1
|
Achappa S, Aldabaan NA, Desai SV, Muddapur UM, Shaikh IA, Mahnashi MH, Alshehri AA, Mannasaheb BA, Khan AA. Computational Exploration of Potential Pharmacological Inhibitors Targeting the Envelope Protein of the Kyasanur Forest Disease Virus. Pharmaceuticals (Basel) 2024; 17:884. [PMID: 39065734 PMCID: PMC11279457 DOI: 10.3390/ph17070884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/19/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
The limitations of the current vaccination strategy for the Kyasanur Forest Disease virus (KFDV) underscore the critical need for effective antiviral treatments, highlighting the crucial importance of exploring novel therapeutic approaches through in silico drug design. Kyasanur Forest Disease, caused by KFDV, is a tick-borne disease with a mortality of 3-5% and an annual incidence of 400 to 500 cases. In the early stage of infection, the envelope protein plays a crucial role by facilitating host-virus interactions. The objective of this research is to develop effective antivirals targeting the envelope protein to disrupt the virus-host interaction. In line with this, the 3D structure of the envelope protein was modeled and refined through molecular modeling techniques, and subsequently, ligands were designed via de novo design and pharmacophore screening, yielding 12 potential hits followed by ADMET analysis. The top five candidates underwent geometry optimization and molecular docking. Notably, compounds L4 (SA28) and L3 (CNP0247967) are predicted to have significant binding affinities of -8.91 and -7.58 kcal/mol, respectively, toward the envelope protein, based on computational models. Both compounds demonstrated stability during 200 ns molecular dynamics simulations, and the MM-GBSA binding free-energy values were -85.26 ± 4.63 kcal/mol and -66.60 ± 2.92 kcal/mol for the envelope protein L3 and L4 complexes, respectively. Based on the computational prediction, it is suggested that both compounds have potential as drug candidates for controlling host-virus interactions by targeting the envelope protein. Further validation through in-vitro assays would complement the findings of the present in silico investigations.
Collapse
Affiliation(s)
- Sharanappa Achappa
- Department of Biotechnology, KLE Technological University, Hubballi 580031, Karnataka, India; (S.A.); (U.M.M.)
| | | | - Shivalingsarj V. Desai
- Department of Biotechnology, KLE Technological University, Hubballi 580031, Karnataka, India; (S.A.); (U.M.M.)
| | - Uday M. Muddapur
- Department of Biotechnology, KLE Technological University, Hubballi 580031, Karnataka, India; (S.A.); (U.M.M.)
| | - Ibrahim Ahmed Shaikh
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia
| | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia;
| | - Abdullateef A. Alshehri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, P.O. Box 1988, Najran 66462, Saudi Arabia;
| | | | - Aejaz Abdullatif Khan
- Department of General Science, Ibn Sina National College for Medical Studies, Jeddah 21418, Saudi Arabia
| |
Collapse
|
2
|
Wei J, Pan Y, Shen Z, Shen L, Xu L, Yu W, Huang W. A hybrid energy-based and AI-based screening approach for the discovery of novel inhibitors of JAK3. Front Med (Lausanne) 2023; 10:1182227. [PMID: 37886358 PMCID: PMC10598672 DOI: 10.3389/fmed.2023.1182227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
The JAKs protein family is composed of four isoforms, and JAK3 has been regarded as a druggable target for the development of drugs to treat various diseases, including hematologic tumors, cancer, and neuronal death. Therefore, the discovery of JAK3 inhibitors with novel scaffolds possesses the potential to provide additional options for drug development. This article presents a structure-based hybrid high-throughput virtual screening (HTVS) protocol as well as the DeepDock algorithm, which is based on geometric deep learning. These techniques were used to identify inhibitors of JAK3 with a novel sketch from a specific "In-house" database. Using molecular docking with varying precision, MM/GBSA, geometric deep learning scoring, and manual selection, 10 compounds were obtained for subsequent biological evaluation. One of these 10 compounds, compound 8, was found to have inhibitory potency against JAK3 and the MOLM-16 cell line, providing a valuable lead compound for further development of JAK3 inhibitors. To gain a better understanding of the interaction between compound 8 and JAK3, molecular dynamics (MD) simulations were conducted to provide more details on the binding conformation of compound 8 with JAK3 to guide the subsequent structure optimization. In this article, we achieved compound 8 with a novel sketch possessing inhibitory bioactivity against JAK3, and it would provide an acceptable "hit" for further structure optimization and modification to develop JAK3 inhibitors.
Collapse
Affiliation(s)
- Juying Wei
- MDS Center, Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Youlu Pan
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zheyuan Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Liteng Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Wenjuan Yu
- MDS Center, Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenhai Huang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Liya DH, Anand NM, Jainarayanan AK, Elanchezhian M, Seetharaman M, Balakannan D, Pradhan AK. Drug repurposing and sequence analysis in S-glycoprotein variants reveals critical signature patterns and destabilization of receptor-binding domain in omicron variant. J Biomol Struct Dyn 2023; 41:7931-7948. [PMID: 36173706 DOI: 10.1080/07391102.2022.2127902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 09/17/2022] [Indexed: 10/14/2022]
Abstract
The evolution of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus since its emergence in 2019 has yielded several new viral variants with varied infectivity, disease severity, and antigenicity. Although most mutations are expected to be relatively neutral, mutations at the Spike region of the genome have shown to have a major impact on the viral transmission and infection in humans. Therefore, it is crucial to survey the structures of spike protein across the global virus population to contextualize the rate of therapeutic success against these variants. In this study, high-frequency mutational variants from different geographic regions were pooled in order to study the structural evolution of the spike protein through drug docking and MD simulations. We investigated the mutational burden in the spike subregions and have observed that the different variants harbour unique signature patterns in the spike subregions, with certain domains being highly prone to mutations. Further, the MD simulations and docking study revealed that different variants show differential stability when docked for the same set of drug targets. This work sheds light on the mutational burden and the stability landscape of the spike protein across the variants from different geographical regions.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Devang Haresh Liya
- Department of Physical Sciences, Indian Institute of Science Education and Research, Mohali, Manauli, India
| | - Nithishwer Mouroug Anand
- Department of Physical Sciences, Indian Institute of Science Education and Research, Mohali, Manauli, India
| | - Ashwin Kumar Jainarayanan
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
| | - Mirudula Elanchezhian
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Manauli, India
| | - Madhumati Seetharaman
- Department of Physical Sciences, Indian Institute of Science Education and Research, Mohali, Manauli, India
| | - Dhanuush Balakannan
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Manauli, India
| | - Arpit Kumar Pradhan
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munchen, Germany
- Graduate School of Systemic Neuroscience, Ludwig Maximilian University of Munich, Munchen, Germany
| |
Collapse
|
4
|
Bryan DR, Kulp JL, Mahapatra MK, Bryan RL, Viswanathan U, Carlisle MN, Kim S, Schutte WD, Clarke KV, Doan TT, Kulp JL. BMaps: A Web Application for Fragment-Based Drug Design and Compound Binding Evaluation. J Chem Inf Model 2023; 63:4229-4236. [PMID: 37406353 DOI: 10.1021/acs.jcim.3c00209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Fragment-based drug design uses data about where, and how strongly, small chemical fragments bind to proteins, to assemble new drug molecules. Over the past decade, we have been successfully using fragment data, derived from thermodynamically rigorous Monte Carlo fragment-protein binding simulations, in dozens of preclinical drug programs. However, this approach has not been available to the broader research community because of the cost and complexity of doing simulations and using design tools. We have developed a web application, called BMaps, to make fragment-based drug design widely available with greatly simplified user interfaces. BMaps provides access to a large repository (>550) of proteins with 100s of precomputed fragment maps, druggable hot spots, and high-quality water maps. Users can also employ their own structures or those from the Protein Data Bank and AlphaFold DB. Multigigabyte data sets are searched to find fragments in bondable orientations, ranked by a binding-free energy metric. The designers use this to select modifications that improve affinity and other properties. BMaps is unique in combining conventional tools such as docking and energy minimization with fragment-based design, in a very easy to use and automated web application. The service is available at https://www.boltzmannmaps.com.
Collapse
Affiliation(s)
- Daniel R Bryan
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - John L Kulp
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
- Zymergen, Inc., 430 E. 29th Street, Suite 625, New York, New York 10016, United States
| | - Manoj K Mahapatra
- Kanak Manjari Institute of Pharmaceutical Sciences, Rourkela 769015, Odisha, India
| | - Richard L Bryan
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Usha Viswanathan
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Micah N Carlisle
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Surim Kim
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
- Zymergen, Inc., 430 E. 29th Street, Suite 625, New York, New York 10016, United States
| | - William D Schutte
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Kevaughn V Clarke
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Tony T Doan
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - John L Kulp
- Conifer Point Pharmaceuticals, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| |
Collapse
|
5
|
Charles S, Edgar MP, Mahapatra RK. Artificial intelligence based virtual screening study for competitive and allosteric inhibitors of the SARS-CoV-2 main protease. J Biomol Struct Dyn 2023; 41:15286-15304. [PMID: 36943715 DOI: 10.1080/07391102.2023.2188419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/27/2023] [Indexed: 03/23/2023]
Abstract
SARS-CoV-2 is a highly contagious and dangerous coronavirus that first appeared in late 2019 causing COVID-19, a pandemic of acute respiratory illnesses that is still a threat to health and the general public safety. We performed deep docking studies of 800 M unique compounds in both the active and allosteric sites of the SARS-COV-2 Main Protease (Mpro) and the 15 M and 13 M virtual hits obtained were further taken for conventional docking and molecular dynamic (MD) studies. The best XP Glide docking scores obtained were -14.242 and -12.059 kcal/mol by CHEMBL591669 and the highest binding affinities were -10.5 kcal/mol (from 444215) and -11.2 kcal/mol (from NPC95421) for active and allosteric sites, respectively. Some hits can bind both sites making them a great area of concern. Re-docking of 8 random allosteric complexes in the active site shows a significant reduction in docking scores with a t-test P value of 2.532 × 10-11 at 95% confidence. Some specific interactions have higher elevations in docking scores. MD studies on 15 complexes show that single-ligand systems are stable as compared to double-ligand systems, and the allosteric binders identified are shown to modulate the active site binding as evidenced by the changes in the interaction patterns and stability of ligands and active site residues. When an allosteric complex was docked to the second monomer to check for homodimer formation, the validated homodimer could not be re-established, further supporting the potential of the identified allosteric binders. These findings could be important in developing novel therapeutics against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ssemuyiga Charles
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
- Department of Microbiology, Biotechnology and Plant Sciences, School of Biological Sciences, Makerere University, Kampala, Uganda
| | - Mulumba Pius Edgar
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
| | | |
Collapse
|
6
|
Chhetri A, Roy M, Mishra P, Halder AK, Basak S, Gangopadhyay A, Saha A, Bhattacharya P. Genetic algorithm- de novo, molecular dynamics and MMGBSA based modelling of a novel Benz-pyrazole based anticancer ligand to functionally revert mutant P53 into wild type P53. MOLECULAR SIMULATION 2023. [DOI: 10.1080/08927022.2023.2185079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Affiliation(s)
- Ashik Chhetri
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, India
| | - Moloy Roy
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, India
| | - Puja Mishra
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, India
| | - Amit Kumar Halder
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, India
| | - Souvik Basak
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, India
| | - Aditi Gangopadhyay
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Plaban Bhattacharya
- Department of Chemical Technology, University of Calcutta, Kolkata, India
- Orange Business, Vishwaroop IT Park, Navi Mumbai, India
| |
Collapse
|
7
|
Transformation rule-based molecular evolution for automatic gasoline molecule design. Chem Eng Sci 2022. [DOI: 10.1016/j.ces.2022.118119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Fleitmann L, Gertig C, Scheffczyk J, Schilling J, Leonhard K, Bardow A. From Molecules to Heat‐Integrated Processes: Computer‐Aided Design of Solvents and Processes Using Quantum Chemistry. CHEM-ING-TECH 2022. [DOI: 10.1002/cite.202200098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Affiliation(s)
- Lorenz Fleitmann
- ETH Zürich Department of Mechanical and Process Engineering, Energy and Process Systems Engineering Tannenstrasse 3 8092 Zürich Switzerland
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Christoph Gertig
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Jan Scheffczyk
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Johannes Schilling
- ETH Zürich Department of Mechanical and Process Engineering, Energy and Process Systems Engineering Tannenstrasse 3 8092 Zürich Switzerland
| | - Kai Leonhard
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - André Bardow
- ETH Zürich Department of Mechanical and Process Engineering, Energy and Process Systems Engineering Tannenstrasse 3 8092 Zürich Switzerland
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
- Forschungszentrum Jülich GmbH Institute of Energy and Climate Research (IEK-10) Wilhelm-Johnen-Straße 52425 Jülich Germany
| |
Collapse
|
9
|
Polte L, Raßpe‐Lange L, Latz F, Jupke A, Leonhard K. COSMO‐CAMPED – Solvent Design for an Extraction Distillation Considering Molecular, Process, Equipment, and Economic Optimization. CHEM-ING-TECH 2022. [DOI: 10.1002/cite.202200144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Lukas Polte
- RWTH Aachen University Fluid Process Engineering (AVT.FVT) Forckenbeckstraße 51 52074 Aachen Germany
| | - Lukas Raßpe‐Lange
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Filip Latz
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Andreas Jupke
- RWTH Aachen University Fluid Process Engineering (AVT.FVT) Forckenbeckstraße 51 52074 Aachen Germany
| | - Kai Leonhard
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| |
Collapse
|
10
|
Prentis LE, Singleton CD, Bickel JD, Allen WJ, Rizzo RC. A molecular evolution algorithm for ligand design in DOCK. J Comput Chem 2022; 43:1942-1963. [PMID: 36073674 PMCID: PMC9623574 DOI: 10.1002/jcc.26993] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/13/2022] [Accepted: 08/03/2022] [Indexed: 01/11/2023]
Abstract
As a complement to virtual screening, de novo design of small molecules is an alternative approach for identifying potential drug candidates. Here, we present a new 3D genetic algorithm to evolve molecules through breeding, mutation, fitness pressure, and selection. The method, termed DOCK_GA, builds upon and leverages powerful sampling, scoring, and searching routines previously implemented into DOCK6. Three primary experiments were used during development: Single-molecule evolution evaluated three selection methods (elitism, tournament, and roulette), in four clinically relevant systems, in terms of mutation type and crossover success, chemical properties, ensemble diversity, and fitness convergence, among others. Large scale benchmarking assessed performance across 651 different protein-ligand systems. Ensemble-based evolution demonstrated using multiple inhibitors simultaneously to seed growth in a SARS-CoV-2 target. Key takeaways include: (1) The algorithm is robust as demonstrated by the successful evolution of molecules across a large diverse dataset. (2) Users have flexibility with regards to parent input, selection method, fitness function, and molecular descriptors. (3) The program is straightforward to run and only requires a single executable and input file at run-time. (4) The elitism selection method yields more tightly clustered molecules in terms of 2D/3D similarity, with more favorable fitness, followed by tournament and roulette.
Collapse
Affiliation(s)
- Lauren E. Prentis
- Department of Biochemistry & Cell BiologyStony Brook UniversityStony BrookNew YorkUSA
| | | | - John D. Bickel
- Department of ChemistryStony Brook UniversityStony BrookNew YorkUSA
| | - William J. Allen
- Department of Applied Mathematics & StatisticsStony Brook UniversityStony BrookNew YorkUSA
| | - Robert C. Rizzo
- Department of Applied Mathematics & StatisticsStony Brook UniversityStony BrookNew YorkUSA
- Institute of Chemical Biology & Drug DiscoveryStony Brook UniversityStony BrookNew YorkUSA
- Laufer Center for Physical & Quantitative BiologyStony Brook UniversityStony BrookNew YorkUSA
| |
Collapse
|
11
|
Mishra P, Basak S, Mukherjee A, Basu A. Design and Study of In Silico Binding Dynamics of Certain Isoxazole Bearing Leads Against Aβ-42 and BACE-1 Loop in Protein Fibrillation. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666210813120444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aims:
Design isoxazole bearing leads as dual inhibitors against Amyloid β and BACE-1 loop
in protein fibrillation.
Background:
Protein fibrillation is one of the key reasons for several diseases, namely Alzheimer’s, Parkinson’s,
and many others. One of the key strategies of preventing protein fibrillation is destabilizing the
protein fibrils themselves or inhibiting the amyloid fibril-forming pathway in the initial stage.
Introduction:
Attempts have been taken to design newer leads to inhibit protein fibrillation by targeting
the β-amyloidogenesis pathway in the brain. To exploit interfenestration between Amyloid β -42 protein
and BACE-1 (β-site amyloid precursor protein cleaving enzyme) for amyloidogenesis, studies are undertaken
to design dual inhibitors against the same.
Method:
In vitro binding interactions were found using docking, de novo ligand design, and MD simulation
study.
Results:
Three compounds bearing an isoxazole heterocyclic nucleus were designed which could successfully
bind to the hydrophobic raft and salt bridge residues Asp 23-Lys-26 of Amyloid β, destabilizing the
growing fibril. Additionally, one of our candidate compounds exhibited force of interaction with Thr232
at the S3 pocket of BACE-1, interacted with key residue Asp228, Tyr71, and Thr72 of the β-hairpin flap
and hydrogen bonding with Gly11 at loop 10s.
Conclusion:
Protein flexibility dynamics of the Aβ-42 protein revealed that there is a considerable conformational
change of the same with or without ligand binding. The lower RMSF of the bound region and
reprogramming residual contacts within the Aβ-42 protein suggested successful binding of the ligand with
the protein, lowering the access for further β-β dimerization.
Collapse
Affiliation(s)
- Puja Mishra
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, WB, India
| | - Souvik Basak
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, WB, India
| | - Arup Mukherjee
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Kolkata, WB, India
| | - Anindya Basu
- School of Pharmaceutical Sciences, Rajiv
Gandhi Proudyogiki Vishwavidyalaya, Bhopal, India
| |
Collapse
|
12
|
Manzer HS, Villarreal RI, Doran KS. Targeting the BspC-vimentin interaction to develop anti-virulence therapies during Group B streptococcal meningitis. PLoS Pathog 2022; 18:e1010397. [PMID: 35316308 PMCID: PMC8939794 DOI: 10.1371/journal.ppat.1010397] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/25/2022] [Indexed: 12/21/2022] Open
Abstract
Bacterial infections are a major cause of morbidity and mortality worldwide and the rise of antibiotic resistance necessitates development of alternative treatments. Pathogen adhesins that bind to host cells initiate disease pathogenesis and represent potential therapeutic targets. We have shown previously that the BspC adhesin in Group B Streptococcus (GBS), the leading cause of bacterial neonatal meningitis, interacts with host vimentin to promote attachment to brain endothelium and disease development. Here we determined that the BspC variable (V-) domain contains the vimentin binding site and promotes GBS adherence to brain endothelium. Site directed mutagenesis identified a binding pocket necessary for GBS host cell interaction and development of meningitis. Using a virtual structure-based drug screen we identified compounds that targeted the V-domain binding pocket, which blocked GBS adherence and entry into the brain in vivo. These data indicate the utility of targeting the pathogen-host interface to develop anti-virulence therapeutics.
Collapse
Affiliation(s)
- Haider S. Manzer
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| | - Ricardo I. Villarreal
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| | - Kelly S. Doran
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| |
Collapse
|
13
|
Kerstjens A, De Winter H. LEADD: Lamarckian evolutionary algorithm for de novo drug design. J Cheminform 2022; 14:3. [PMID: 35033209 PMCID: PMC8760751 DOI: 10.1186/s13321-022-00582-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/30/2021] [Indexed: 11/10/2022] Open
Abstract
Given an objective function that predicts key properties of a molecule, goal-directed de novo molecular design is a useful tool to identify molecules that maximize or minimize said objective function. Nonetheless, a common drawback of these methods is that they tend to design synthetically unfeasible molecules. In this paper we describe a Lamarckian evolutionary algorithm for de novo drug design (LEADD). LEADD attempts to strike a balance between optimization power, synthetic accessibility of designed molecules and computational efficiency. To increase the likelihood of designing synthetically accessible molecules, LEADD represents molecules as graphs of molecular fragments, and limits the bonds that can be formed between them through knowledge-based pairwise atom type compatibility rules. A reference library of drug-like molecules is used to extract fragments, fragment preferences and compatibility rules. A novel set of genetic operators that enforce these rules in a computationally efficient manner is presented. To sample chemical space more efficiently we also explore a Lamarckian evolutionary mechanism that adapts the reproductive behavior of molecules. LEADD has been compared to both standard virtual screening and a comparable evolutionary algorithm using a standardized benchmark suite and was shown to be able to identify fitter molecules more efficiently. Moreover, the designed molecules are predicted to be easier to synthesize than those designed by other evolutionary algorithms.
Collapse
Affiliation(s)
- Alan Kerstjens
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1A, 2610, Wilrijk, Belgium
| | - Hans De Winter
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1A, 2610, Wilrijk, Belgium.
| |
Collapse
|
14
|
Fragment-to-lead tailored in silico design. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 40:44-57. [PMID: 34916022 DOI: 10.1016/j.ddtec.2021.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/25/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
Fragment-based drug discovery (FBDD) emerged as a disruptive technology and became established during the last two decades. Its rationality and low entry costs make it appealing, and the numerous examples of approved drugs discovered through FBDD validate the approach. However, FBDD still faces numerous challenges. Perhaps the most important one is the transformation of the initial fragment hits into viable leads. Fragment-to-lead (F2L) optimization is resource-intensive and is therefore limited in the possibilities that can be actively pursued. In silico strategies play an important role in F2L, as they can perform a deeper exploration of chemical space, prioritize molecules with high probabilities of being active and generate non-obvious ideas. Here we provide a critical overview of current in silico strategies in F2L optimization and highlight their remarkable impact. While very effective, most solutions are target- or fragment- specific. We propose that fully integrated in silico strategies, capable of automatically and systematically exploring the fast-growing available chemical space can have a significant impact on accelerating the release of fragment originated drugs.
Collapse
|
15
|
COSMO-susCAMPD: Sustainable solvents from combining computer-aided molecular and process design with predictive life cycle assessment. Chem Eng Sci 2021. [DOI: 10.1016/j.ces.2021.116863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
16
|
Guterres H, Park SJ, Cao Y, Im W. CHARMM-GUI Ligand Designer for Template-Based Virtual Ligand Design in a Binding Site. J Chem Inf Model 2021; 61:5336-5342. [PMID: 34757752 DOI: 10.1021/acs.jcim.1c01156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Rational drug design involves a task of finding ligands that would bind to a specific target protein. This work presents CHARMM-GUI Ligand Designer that is an intuitive and interactive web-based tool to design virtual ligands that match the shape and chemical features of a given protein binding site. Ligand Designer provides ligand modification capabilities with 3D visualization that allow researchers to modify and redesign virtual ligands while viewing how the protein-ligand interactions are affected. Virtual ligands can also be parameterized for further molecular dynamics (MD) simulations and free energy calculations. Using 8 targets from 8 different protein classes in the directory of useful decoys, enhanced (DUD-E) data set, we show that Ligand Designer can produce similar ligands to the known active ligands in the crystal structures. Ligand Designer also produces stable protein-ligand complex structures when tested using short MD simulations. We expect that Ligand Designer can be a useful and user-friendly tool to design small molecules in any given potential ligand binding site on a protein of interest.
Collapse
Affiliation(s)
- Hugo Guterres
- Departments of Biological Sciences, Chemistry, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Sang-Jun Park
- Departments of Biological Sciences, Chemistry, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yiwei Cao
- Departments of Biological Sciences, Chemistry, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Wonpil Im
- Departments of Biological Sciences, Chemistry, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
17
|
Gertig C, Fleitmann L, Hemprich C, Hense J, Bardow A, Leonhard K. CAT-COSMO-CAMPD: Integrated in silico design of catalysts and processes based on quantum chemistry. Comput Chem Eng 2021. [DOI: 10.1016/j.compchemeng.2021.107438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
18
|
Prado-Romero D, Medina-Franco JL. Advances in the Exploration of the Epigenetic Relevant Chemical Space. ACS OMEGA 2021; 6:22478-22486. [PMID: 34514220 PMCID: PMC8427648 DOI: 10.1021/acsomega.1c03389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/10/2021] [Indexed: 06/13/2023]
Abstract
Epigenetic drug discovery is a promising avenue to find therapeutic agents for treating several diseases and developing novel chemical probes for research. In order to identify hit and lead compounds, the chemical space has been explored and screened, generating valuable bioactivity information that can be used for multiple purposes such as prediction of the activity of existing chemicals, e.g., small molecules, guiding the design or optimization of compounds, and expanding the epigenetic relevant chemical space. Herein, we review the chemical spaces explored for epigenetic drug discovery and discuss the advances in using structure-activity relationships stored in public chemogenomic databases. We also review current efforts to chart and identify novel regions of the epigenetic relevant chemical space. In particular, we discuss the development and accessibility of two significant types of compound libraries focused on epigenetic targets: commercially available libraries for screening and targeted chemical libraries using de novo design. In this mini-review, we emphasize inhibitors of DNA methyltransferases.
Collapse
|
19
|
Nayarisseri A. Experimental and Computational Approaches to Improve Binding Affinity in Chemical Biology and Drug Discovery. Curr Top Med Chem 2021; 20:1651-1660. [PMID: 32614747 DOI: 10.2174/156802662019200701164759] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug discovery is one of the most complicated processes and establishment of a single drug may require multidisciplinary attempts to design efficient and commercially viable drugs. The main purpose of drug design is to identify a chemical compound or inhibitor that can bind to an active site of a specific cavity on a target protein. The traditional drug design methods involved various experimental based approaches including random screening of chemicals found in nature or can be synthesized directly in chemical laboratories. Except for the long cycle design and time, high cost is also the major issue of concern. Modernized computer-based algorithm including structure-based drug design has accelerated the drug design and discovery process adequately. Surprisingly from the past decade remarkable progress has been made concerned with all area of drug design and discovery. CADD (Computer Aided Drug Designing) based tools shorten the conventional cycle size and also generate chemically more stable and worthy compounds and hence reduce the drug discovery cost. This special edition of editorial comprises the combination of seven research and review articles set emphasis especially on the computational approaches along with the experimental approaches using a chemical synthesizing for the binding affinity in chemical biology and discovery as a salient used in de-novo drug designing. This set of articles exfoliates the role that systems biology and the evaluation of ligand affinity in drug design and discovery for the future.
Collapse
Affiliation(s)
- Anuraj Nayarisseri
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore - 452010, Madhya Pradesh, India
| |
Collapse
|
20
|
Anand NM, Liya DH, Pradhan AK, Tayal N, Bansal A, Donakonda S, Jainarayanan AK. A comprehensive SARS-CoV-2 genomic analysis identifies potential targets for drug repurposing. PLoS One 2021; 16:e0248553. [PMID: 33735271 PMCID: PMC7971693 DOI: 10.1371/journal.pone.0248553] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/01/2021] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which is a novel human coronavirus strain (HCoV) was initially reported in December 2019 in Wuhan City, China. This acute infection caused pneumonia-like symptoms and other respiratory tract illness. Its higher transmission and infection rate has successfully enabled it to have a global spread over a matter of small time. One of the major concerns involving the SARS-COV-2 is the mutation rate, which enhances the virus evolution and genome variability, thereby making the design of therapeutics difficult. In this study, we identified the most common haplotypes from the haplotype network. The conserved genes and population level variants were analysed. Non-Structural Protein 10 (NSP10), Nucleoprotein, Papain-like protease (Plpro or NSP3) and 3-Chymotrypsin like protease (3CLpro or NSP5), which were conserved at the highest threshold, were used as drug targets for molecular dynamics simulations. Darifenacin, Nebivolol, Bictegravir, Alvimopan and Irbesartan are among the potential drugs, which are suggested for further pre-clinical and clinical trials. This particular study provides a comprehensive targeting of the conserved genes. We also identified the mutation frequencies across the viral genome.
Collapse
Affiliation(s)
- Nithishwer Mouroug Anand
- Department of Physical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Devang Haresh Liya
- Department of Physical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Arpit Kumar Pradhan
- Graduate School of Systemic Neuroscience, Ludwig Maximilian University of Munich, Munich, Germany
- Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Nitish Tayal
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Abhinav Bansal
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Sainitin Donakonda
- Institute of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Ashwin Kumar Jainarayanan
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
- Interdisciplinary Bioscience DTP, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Mouchlis VD, Afantitis A, Serra A, Fratello M, Papadiamantis AG, Aidinis V, Lynch I, Greco D, Melagraki G. Advances in de Novo Drug Design: From Conventional to Machine Learning Methods. Int J Mol Sci 2021; 22:1676. [PMID: 33562347 PMCID: PMC7915729 DOI: 10.3390/ijms22041676] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/31/2021] [Accepted: 01/31/2021] [Indexed: 12/11/2022] Open
Abstract
. De novo drug design is a computational approach that generates novel molecular structures from atomic building blocks with no a priori relationships. Conventional methods include structure-based and ligand-based design, which depend on the properties of the active site of a biological target or its known active binders, respectively. Artificial intelligence, including machine learning, is an emerging field that has positively impacted the drug discovery process. Deep reinforcement learning is a subdivision of machine learning that combines artificial neural networks with reinforcement-learning architectures. This method has successfully been employed to develop novel de novo drug design approaches using a variety of artificial networks including recurrent neural networks, convolutional neural networks, generative adversarial networks, and autoencoders. This review article summarizes advances in de novo drug design, from conventional growth algorithms to advanced machine-learning methodologies and highlights hot topics for further development.
Collapse
Affiliation(s)
| | - Antreas Afantitis
- Department of ChemoInformatics, NovaMechanics Ltd., Nicosia 1046, Cyprus;
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (M.F.); (D.G.)
- BioMEdiTech Institute, Tampere University, 33520 Tampere, Finland
| | - Michele Fratello
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (M.F.); (D.G.)
- BioMEdiTech Institute, Tampere University, 33520 Tampere, Finland
| | - Anastasios G. Papadiamantis
- Department of ChemoInformatics, NovaMechanics Ltd., Nicosia 1046, Cyprus;
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Vassilis Aidinis
- Institute for Bioinnovation, Biomedical Sciences Research Center Alexander Fleming, Fleming 34, 16672 Athens, Greece;
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (M.F.); (D.G.)
- BioMEdiTech Institute, Tampere University, 33520 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
- Finnish Center for Alternative Methods (FICAM), Tampere University, 33520 Tampere, Finland
| | - Georgia Melagraki
- Division of Physical Sciences & Applications, Hellenic Military Academy, 16672 Vari, Greece
| |
Collapse
|
22
|
Liu X, IJzerman AP, van Westen GJP. Computational Approaches for De Novo Drug Design: Past, Present, and Future. Methods Mol Biol 2021; 2190:139-165. [PMID: 32804364 DOI: 10.1007/978-1-0716-0826-5_6] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug discovery is time- and resource-consuming. To this end, computational approaches that are applied in de novo drug design play an important role to improve the efficiency and decrease costs to develop novel drugs. Over several decades, a variety of methods have been proposed and applied in practice. Traditionally, drug design problems are always taken as combinational optimization in discrete chemical space. Hence optimization methods were exploited to search for new drug molecules to meet multiple objectives. With the accumulation of data and the development of machine learning methods, computational drug design methods have gradually shifted to a new paradigm. There has been particular interest in the potential application of deep learning methods to drug design. In this chapter, we will give a brief description of these two different de novo methods, compare their application scopes and discuss their possible development in the future.
Collapse
Affiliation(s)
- Xuhan Liu
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Gerard J P van Westen
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.
| |
Collapse
|
23
|
Mahfuz AMUB, Stambuk Opazo F, Aguilar LF, Iqbal MN. Carfilzomib as a potential inhibitor of NADH-dependent enoyl-acyl carrier protein reductases of Klebsiella pneumoniae and Mycobacterium tuberculosis as a drug target enzyme: insights from molecular docking and molecular dynamics. J Biomol Struct Dyn 2020; 40:4021-4037. [PMID: 33251968 DOI: 10.1080/07391102.2020.1852966] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Multiple antibiotic-resistant strains of Klebsiella pneumoniae can cause life-threatening infections. Bacterial enoyl-acyl carrier protein (ACP) reductases (ENRs) are considered critical targets for developing antibiotics. Our current study aims to identify inhibitors of K. pneumoniae ENRs (FabI and FabV). Due to the unavailability of experimental structures, protein models of FabI and FabV were predicted and validated in this study. Virtual screening of the 1930 FDA-approved drug database was conducted against the active site of the FabI protein with the help of the LEA3D server, and carfilzomib was chosen among the screened drugs for further docking studies. Carfilzomib, a proteasome inhibitor used in the treatment of multiple myeloma, was among the best-suited compounds obtained from the virtual screening and was found to be bactericidal in the in vitro experiment. Carfilzomib was docked against the active sites of the FabI and FabV proteins, and the ENR of Mycobacterium tuberculosis, InhA. Carfilzomib showed a high binding affinity with all three proteins. Molecular dynamics (MD) simulations were conducted following the docking studies. MD simulations revealed that carfilzomib binds strongly to the active sites of the above mentioned ENRs. Our study found that carfilzomib is a potential inhibitor of the ENRs of K. pneumoniae and M. tuberculosis. This is a possible mechanism of its bactericidal property against M. tuberculosis observed in vitro in addition to its predicted actions on zinc-dependent metalloprotease-1 and peptide deformylase, two other drug target enzymes of M. tuberculosis. Our study suggests that this drug could be used as a lead compound to develop antibiotics that can selectively act against ENRs of bacteria, without interfering with the activities of human proteasome. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- A M U B Mahfuz
- Department of Biotechnology & Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka, Bangladesh
| | - Felipe Stambuk Opazo
- Laboratorio de Genética e Inmunología Molecular, Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Luis F Aguilar
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Muhammad Nasir Iqbal
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad, ICT, Pakistan
| |
Collapse
|
24
|
Verma SK, Ratre P, Jain AK, Liang C, Gupta GD, Thareja S. De novo designing, assessment of target affinity and binding interactions against aromatase: Discovery of novel leads as anti-breast cancer agents. Struct Chem 2020. [DOI: 10.1007/s11224-020-01673-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
25
|
Gertig C, Fleitmann L, Schilling J, Leonhard K, Bardow A. Rx‐COSMO‐CAMPD: Enhancing Reactions by Integrated Computer‐Aided Design of Solvents and Processes based on Quantum Chemistry. CHEM-ING-TECH 2020. [DOI: 10.1002/cite.202000112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Christoph Gertig
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Lorenz Fleitmann
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Johannes Schilling
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - Kai Leonhard
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
| | - André Bardow
- RWTH Aachen University Institute of Technical Thermodynamics Schinkelstraße 8 52062 Aachen Germany
- Forschungszentrum Jülich GmbH Institute of Energy and Climate Research – Energy Systems Engineering (IEK-10) Wilhelm-Johnen-Straße 52425 Jülich Germany
- ETH Zurich Department of Mechanical and Process Engineering, Energy & Process Systems Engineering Tannenstrasse 3 8092 Zürich Switzerland
| |
Collapse
|
26
|
Copertino DC, Duarte RRR, Powell TR, de Mulder Rougvie M, Nixon DF. Montelukast drug activity and potential against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). J Med Virol 2020; 93:187-189. [PMID: 32658304 PMCID: PMC7405283 DOI: 10.1002/jmv.26299] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Dennis C Copertino
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, New York
| | - Rodrigo R R Duarte
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, New York
| | - Timothy R Powell
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, New York
| | | | - Douglas F Nixon
- Division of Infectious Diseases, Weill Cornell Medicine, Cornell University, New York, New York
| |
Collapse
|
27
|
Chatterjee S, Maity A, Chowdhury S, Islam MA, Muttinini RK, Sen D. In silico analysis and identification of promising hits against 2019 novel coronavirus 3C-like main protease enzyme. J Biomol Struct Dyn 2020; 39:5290-5303. [PMID: 32608329 DOI: 10.1080/07391102.2020.1787228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The recent outbreak of the 2019 novel coronavirus disease (COVID-19) has been proved as a global threat. No particular drug or vaccine has not yet been discovered which may act specifically against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and causes COVID-19. For this highly infectious virus, 3CL-like main protease (3CLpro) plays a key role in the virus life cycle and can be considered as a pivotal drug target. Structure-based virtual screening of DrugBank database resulted in 20 hits against 3CLpro. Atomistic 100 ns molecular dynamics of five top hits and binding energy calculation analyses were performed for main protease-hit complexes. Among the top five hits, Nafarelin and Icatibant affirmed the binding energy (g_MMPBSA) of -712.94 kJ/mol and -851.74 kJ/mol, respectively. Based on binding energy and stability of protein-ligand complex; the present work reports these two drug-like hits against SARS-CoV-2 main protease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shilpa Chatterjee
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Arindam Maity
- School of Pharmaceutical Technology, Adamas University, Kolkata, India
| | - Suchana Chowdhury
- BCDA College of Pharmaceutical Technology, Hridaypur, Kolkata, India
| | - Md Ataul Islam
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,School of Health Sciences, University of Kwazulu-Natal, Durban, South Africa
| | | | - Debanjan Sen
- BCDA College of Pharmaceutical Technology, Hridaypur, Kolkata, India
| |
Collapse
|
28
|
Banjare L, Verma SK, Jain AK, Thareja S. Lead Molecules as Novel Aromatase Inhibitors: In Silico De Novo Designing and Binding Affinity Studies. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190703152659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Aromatase inhibitors emerged as a pivotal moiety to selectively block estrogen production, prevention and treatment of tumour growth in breast cancer. De novo drug design is an alternative approach to blind virtual screening for successful designing of the novel molecule against various therapeutic targets.Objective:In the present study, we have explored the de novo approach to design novel aromatase inhibitors.Method:The e-LEA3D, a computational-aided drug design web server was used to design novel drug-like candidates against the target aromatase. For drug-likeness ADME parameters (molecular weight, H-bond acceptors, H-bond donors, LogP and number of rotatable bonds) of designed molecules were calculated in TSAR software package, geometry optimization and energy minimization was accomplished using Chem Office. Further, molecular docking study was performed in Molegro Virtual Docker (MVD).Results:Among 17 generated molecules using the de novo pathway, 13 molecules passed the Lipinski filter pertaining to their bioavailability characteristics. De novo designed molecules with drug-likeness were further docked into the mapped active site of aromatase to scale up their affinity and binding fitness with the target. Among de novo fabricated drug like candidates (1-13), two molecules (5, 6) exhibited higher affinity with aromatase in terms of MolDock score (-150.650, -172.680 Kcal/mol, respectively) while molecule 8 showed lowest target affinity (-85.588 Kcal/mol).Conclusion:The binding patterns of lead molecules (5, 6) could be used as a pharmacophore for medicinal chemists to explore these molecules for their aromatase inhibitory potential.
Collapse
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| | - Sant Kumar Verma
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| | - Suresh Thareja
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495009 (C.G.), India
| |
Collapse
|
29
|
Singh N, Chaput L, Villoutreix BO. Virtual screening web servers: designing chemical probes and drug candidates in the cyberspace. Brief Bioinform 2020; 22:1790-1818. [PMID: 32187356 PMCID: PMC7986591 DOI: 10.1093/bib/bbaa034] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The interplay between life sciences and advancing technology drives a continuous cycle of chemical data growth; these data are most often stored in open or partially open databases. In parallel, many different types of algorithms are being developed to manipulate these chemical objects and associated bioactivity data. Virtual screening methods are among the most popular computational approaches in pharmaceutical research. Today, user-friendly web-based tools are available to help scientists perform virtual screening experiments. This article provides an overview of internet resources enabling and supporting chemical biology and early drug discovery with a main emphasis on web servers dedicated to virtual ligand screening and small-molecule docking. This survey first introduces some key concepts and then presents recent and easily accessible virtual screening and related target-fishing tools as well as briefly discusses case studies enabled by some of these web services. Notwithstanding further improvements, already available web-based tools not only contribute to the design of bioactive molecules and assist drug repositioning but also help to generate new ideas and explore different hypotheses in a timely fashion while contributing to teaching in the field of drug development.
Collapse
Affiliation(s)
- Natesh Singh
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Ludovic Chaput
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Bruno O Villoutreix
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 Drugs and Molecules for Living Systems, F-59000 Lille, France
| |
Collapse
|
30
|
PETRA: Drug Engineering via Rigidity Analysis. Molecules 2020; 25:molecules25061304. [PMID: 32178472 PMCID: PMC7144111 DOI: 10.3390/molecules25061304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 11/23/2022] Open
Abstract
Rational drug design aims to develop pharmaceutical agents that impart maximal therapeutic benefits via their interaction with their intended biological targets. In the past several decades, advances in computational tools that inform wet-lab techniques have aided the development of a wide variety of new medicines with high efficacies. Nonetheless, drug development remains a time and cost intensive process. In this work, we have developed a computational pipeline for assessing how individual atoms contribute to a ligand’s effect on the structural stability of a biological target. Our approach takes as input a protein-ligand resolved PDB structure file and systematically generates all possible ligand variants. We assess how the atomic-level edits to the ligand alter the drug’s effect via a graph theoretic rigidity analysis approach. We demonstrate, via four case studies of common drugs, the utility of our pipeline and corroborate our analyses with known biophysical properties of the medicines, as reported in the literature.
Collapse
|
31
|
Perez C, Soler D, Soliva R, Guallar V. FragPELE: Dynamic Ligand Growing within a Binding Site. A Novel Tool for Hit-To-Lead Drug Design. J Chem Inf Model 2020; 60:1728-1736. [PMID: 32027130 DOI: 10.1021/acs.jcim.9b00938] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The early stages of drug discovery rely on hit-to-lead programs, where initial hits undergo partial optimization to improve binding affinities for their biological target. This is an expensive and time-consuming process, requiring multiple iterations of trial and error designs, an ideal scenario for applying computer simulation. However, most state-of-the-art modeling techniques fail to provide a fast and reliable answer to the Induced-Fit protein-ligand problem. To aid in this matter, we present FragPELE, a new tool for in silico hit-to-lead drug design, capable of growing a fragment from a bound core while exploring the protein-ligand conformational space. We tested the ability of FragPELE to predict crystallographic data, even in cases where cryptic sub-pockets open because of the presence of particular R-groups. Additionally, we evaluated the potential of the software on growing and scoring five congeneric series from the 2015 FEP+ dataset, comparing them to FEP+, SP and Induced-Fit Glide, and MMGBSA simulations. Results show that FragPELE could be useful not only for finding new cavities and novel binding modes in cases where standard docking tools cannot but also to rank ligand activities in a reasonable amount of time and with acceptable precision.
Collapse
Affiliation(s)
- Carles Perez
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona 08034, Spain
| | - Daniel Soler
- Nostrum Biodiscovery, Carrer Jordi Girona 29, Nexus II D128, 08034 Barcelona, Spain
| | - Robert Soliva
- Nostrum Biodiscovery, Carrer Jordi Girona 29, Nexus II D128, 08034 Barcelona, Spain
| | - Victor Guallar
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona 08034, Spain.,ICREA: Institució Catalana de Recerca i Estudis Avançats, Passeig Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
32
|
Huang Y, Wei L, Han X, Chen H, Ren Y, Xu Y, Song R, Rao L, Su C, Peng C, Feng L, Wan J. Discovery of novel allosteric site and covalent inhibitors of FBPase with potent hypoglycemic effects. Eur J Med Chem 2019; 184:111749. [DOI: 10.1016/j.ejmech.2019.111749] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/20/2019] [Accepted: 09/28/2019] [Indexed: 12/21/2022]
|
33
|
Gertig C, Kröger L, Fleitmann L, Scheffczyk J, Bardow A, Leonhard K. Rx-COSMO-CAMD: Computer-Aided Molecular Design of Reaction Solvents Based on Predictive Kinetics from Quantum Chemistry. Ind Eng Chem Res 2019. [DOI: 10.1021/acs.iecr.9b03232] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Christoph Gertig
- Institute of Technical Thermodynamics, RWTH Aachen University, Schinkelstraße 8, 52062 Aachen, Germany
| | - Leif Kröger
- Institute of Technical Thermodynamics, RWTH Aachen University, Schinkelstraße 8, 52062 Aachen, Germany
| | - Lorenz Fleitmann
- Institute of Technical Thermodynamics, RWTH Aachen University, Schinkelstraße 8, 52062 Aachen, Germany
| | - Jan Scheffczyk
- Institute of Technical Thermodynamics, RWTH Aachen University, Schinkelstraße 8, 52062 Aachen, Germany
| | - André Bardow
- Institute of Technical Thermodynamics, RWTH Aachen University, Schinkelstraße 8, 52062 Aachen, Germany
- Institute of Energy and Climate Research—Energy Systems Engineering (IEK-10), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52425 Jülich, Germany
| | - Kai Leonhard
- Institute of Technical Thermodynamics, RWTH Aachen University, Schinkelstraße 8, 52062 Aachen, Germany
| |
Collapse
|
34
|
Hsu HH, Huang CH, Lin ST. New Data Structure for Computational Molecular Design with Atomic or Fragment Resolution. J Chem Inf Model 2019; 59:3703-3713. [PMID: 31393721 DOI: 10.1021/acs.jcim.9b00478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A new molecular data structure and molecular structure operation algorithms are proposed for general purpose molecular design. The data structure allows for a variety of molecular operations for creating new molecules. Two types of molecular operations were developed, unimolecular and bimolecular operations. In unimolecular operations, a child molecule can be created from a parent via addition of a functional group, deletion of a fragment, mutation of an atom, etc. In bimolecular operations, children molecules are generated from two parent molecules through combination or crossover (hybridization). These molecular operations are essential for the creation and modification of molecules for the purpose of molecular design. The data structure is capable of representing linear, branched, multifunctional, and multivalent compounds. Algorithms are developed for deriving the molecular data structure of a molecule from its atomic coordinates and vice versa. We show that this new molecular data structure and the developed algorithms, referred to as Molecular Assembling and Representation Suite, allow one to generate a comprehensive library of new molecules via performing every possible molecular structure modification.
Collapse
Affiliation(s)
- Hsuan-Hao Hsu
- Department of Chemical Engineering , National Taiwan University , Taipei 10617 , Taiwan
| | - Chen-Hsuan Huang
- Department of Chemical Engineering , National Taiwan University , Taipei 10617 , Taiwan
| | - Shiang-Tai Lin
- Department of Chemical Engineering , National Taiwan University , Taipei 10617 , Taiwan
| |
Collapse
|
35
|
Islam MA, Pillay TS. Identification of promising anti-DNA gyrase antibacterial compounds using de novo design, molecular docking and molecular dynamics studies. J Biomol Struct Dyn 2019; 38:1798-1809. [PMID: 31084271 DOI: 10.1080/07391102.2019.1617785] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The rapidly increasing rate of antibiotic resistance is of great concern. Approximately two million deaths result annually from bacterial infections worldwide. Therefore, there is a paramount requirement to develop innovative and novel antibacterial agents with new mechanisms of action and activity against resistant bacterial strains. For this purpose, a set of benzothiazole and N-phenylpyrrolamides derivatives reported as DNA Gyrase B (GyrB) inhibitors were collected from the literature and docked inside the receptor cavity of DNA Gyrase B (PDB ID: 5L3J). The best 10 docked complexes were used to identify novel antibacterial chemical agents through a de novo design approach. Out of initial 300 chemical analogues, the best six analogues were identified using screening with a set of criteria followed by pharmacokinetic analysis. The binding interactions of the best six analogues revealed that all molecules formed a number of critical interactions with catalytic amino residues of DNA Gyrase B with high binding energy. The predicted inhibitory constant biological activity based on binding energy supported the potential of the molecules as DNA Gyrase B ligands. The RMSD, RMSF, and radius of gyration parameters obtained from the 100 ns molecular dynamics simulation study clearly demonstrated that all six analogues were efficient enough to form stable complexes with DNA Gyrase B. High negative binding energy of all ligands obtained from MM-GBSA approach undoubtedly explained the strong affinity toward the DNA Gyrase B. Therefore, the proposed de novo designed molecules can be considered as promising antibacterial chemical agents subject to experimental validation, in vitro.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Md Ataul Islam
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria and National Health Laboratory Service Tshwane Academic Division, Pretoria, South Africa.,School of Health Sciences, University of Kwazulu-Natal, Durban, South Africa.,Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Tahir S Pillay
- Department of Chemical Pathology, Faculty of Health Sciences, University of Pretoria and National Health Laboratory Service Tshwane Academic Division, Pretoria, South Africa.,Division of Chemical Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
36
|
Chu Y, He X. MoleGear: A Java-Based Platform for Evolutionary De Novo Molecular Design. Molecules 2019; 24:E1444. [PMID: 30979097 PMCID: PMC6479339 DOI: 10.3390/molecules24071444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 11/17/2022] Open
Abstract
A Java-based platform, MoleGear, is developed for de novo molecular design based on the chemistry development kit (CDK) and other Java packages. MoleGear uses evolutionary algorithm (EA) to explore chemical space, and a suite of fragment-based operators of growing, crossover, and mutation for assembling novel molecules that can be scored by prediction of binding free energy or a weighted-sum multi-objective fitness function. The EA can be conducted in parallel over multiple nodes to support large-scale molecular optimizations. Some complementary utilities such as fragment library design, chemical space analysis, and graphical user interface are also integrated into MoleGear. The candidate molecules as inhibitors for the human immunodeficiency virus 1 (HIV-1) protease were designed by MoleGear, which validates the potential capability for de novo molecular design.
Collapse
Affiliation(s)
- Yunhan Chu
- Department of Chemical Engineering, Norwegian University of Science and Technology, N-7491 Trondheim, Norway.
| | - Xuezhong He
- Department of Chemical Engineering, Norwegian University of Science and Technology, N-7491 Trondheim, Norway.
| |
Collapse
|
37
|
Chen JJ, Schmucker LN, Visco DP. Identifying de-NEDDylation inhibitors: Virtual high-throughput screens targeting SENP8. Chem Biol Drug Des 2019; 93:590-604. [PMID: 30560590 DOI: 10.1111/cbdd.13457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/21/2018] [Accepted: 11/24/2018] [Indexed: 12/16/2022]
Abstract
Protein modification can have far-reaching effects. NEDDylation, a protein modification process with the protein NEDD8, stabilizes and modifies how the targeted protein interacts with other proteins. Its role in system regulation makes it a prime therapeutic target, and virtual high-throughput screening has already identified new NEDD8 inhibitors. SENP8 matures the NEDD8 proenzyme into the active form and regulates NEDDylation by removing NEDD8 from over-NEDDylated proteins. In this work, SENP8 inhibitor candidates were identified in two rounds of virtual high-throughput screening. Of the ten candidates identified in the first round of screening, four were active in validation experiments to yield an experimental hit rate of 40%. Of the five candidates identified in the second round of screening, one was active in validation experiments to yield an experimental hit rate of 20%. Results indicate virtual high-throughput screening improved hit rates over traditional high-throughput screening. The SENP8 inhibitor candidates can be used to interrogate the NEDDylation regulation mechanism.
Collapse
Affiliation(s)
| | - Lyndsey N Schmucker
- Department of Chemical and Biomolecular Engineering, University of Akron, Akron, OH
| | - Donald P Visco
- Department of Chemical and Biomolecular Engineering, University of Akron, Akron, OH
| |
Collapse
|
38
|
Xiao S, Wei L, Hong Z, Rao L, Ren Y, Wan J, Feng L. Design, synthesis and algicides activities of thiourea derivatives as the novel scaffold aldolase inhibitors. Bioorg Med Chem 2019; 27:805-812. [PMID: 30711311 DOI: 10.1016/j.bmc.2019.01.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 12/25/2022]
Abstract
By using a new Fragment-Based Virtual Screen strategy, two series of novel FBA-II inhibitors (thiourea derivatives) were de novo discovered based on the active site of fructose-1, 6-bisphosphate aldolase from Cyanobacterial (CyFBA). In comparison, most of the N-(2-benzoylhydrazine-1-carbonothioyl) benzamide derivatives (L14∼L22) exhibit higher CyFBA-II inhibitory activities compared to N-(phenylcarbamothioyl) benzamide derivatives (L1∼L13). Especially, compound L14 not only shows higher CyFBA-II activity (Ki = 0.65 μM), but also exhibits most potent in vivo activity against Synechocystis sp. PCC 6803 (EC50 = 0.09 ppm), higher (7-fold) than that of our previous inhibitor (EC50 = 0.6 ppm). The binding modes of compound L14 and CyFBA-II were further elucidated by jointly using DOX computational protocol, MM-PBSA and site-directed mutagenesis assays. The positive results suggest that strategy adopted in this study was promising to rapidly discovery the potent inhibitors with novel scaffolds. The satisfactory algicide activities suggest that the thiourea derivatives is very likely to be a promising lead for the development of novel specific algicides to solve Cyanobacterial harmful algal blooms (CHABs).
Collapse
Affiliation(s)
- Shan Xiao
- International Cooperation Base of Pesticide and Green Synthesis (Hubei), Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, Department of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Lin Wei
- International Cooperation Base of Pesticide and Green Synthesis (Hubei), Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, Department of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Zongqin Hong
- International Cooperation Base of Pesticide and Green Synthesis (Hubei), Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, Department of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Li Rao
- International Cooperation Base of Pesticide and Green Synthesis (Hubei), Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, Department of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Yanliang Ren
- International Cooperation Base of Pesticide and Green Synthesis (Hubei), Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, Department of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Jian Wan
- International Cooperation Base of Pesticide and Green Synthesis (Hubei), Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, Department of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Lingling Feng
- International Cooperation Base of Pesticide and Green Synthesis (Hubei), Key Laboratory of Pesticide & Chemical Biology (CCNU), Ministry of Education, Department of Chemistry, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
39
|
Chen JJ, Schmucker LN, Visco DP. Virtual high-throughput screens identifying hPK-M2 inhibitors: Exploration of model extrapolation. Comput Biol Chem 2019; 78:317-329. [PMID: 30623877 DOI: 10.1016/j.compbiolchem.2018.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 10/27/2022]
Abstract
Glycolysis with PK-M2 occurs typically in anaerobic conditions and atypically in aerobic conditions, which is known as the Warburg effect. The Warburg effect is found in many oncogenic situations and is believed to provide energy and biomass for oncogenesis to persist. The work presented targets human PK-M2 (hPK-M2) in a virtual high-throughput screen to identify new inhibitors and leads for further study. In the initial screen, one of the 12 candidates selected for experimental validation showed biological activity (hit-rate = 8.13%). In the second screen with retrained models, six of 11 candidates selected for experimental validation showed biological activity (hit-rate: 54.5%). Additionally, four different scaffolds were identified for further analysis when examining the tested candidates and compounds in the training data. Finally, extrapolation was necessary to identify a sufficient number of candidates to test in the second screen. Examination of the results suggested stepwise extrapolation to maximize efficiency.
Collapse
Affiliation(s)
- Jonathan J Chen
- Department of Biology, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA.
| | - Lyndsey N Schmucker
- Department of Chemical and Biomolecular Engineering, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA.
| | - Donald P Visco
- Department of Chemical and Biomolecular Engineering, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA.
| |
Collapse
|
40
|
Chen JJ, Schmucker LN, Visco DP. Identifying new clotting factor XIa inhibitors in virtual high-throughput screens using PCA-GA-SVM models and signature. Biotechnol Prog 2018; 34:1553-1565. [PMID: 30009405 DOI: 10.1002/btpr.2693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/08/2018] [Accepted: 06/28/2018] [Indexed: 12/17/2022]
Abstract
Blood Clotting Factor XI is an important actor in the clotting mechanism: it activates downstream zymogen involved in the clotting process. It can be targeted for activation or inhibition depending on treatment goals to enhance or inhibit clotting. In terms of antithrombosis treatment, Factor XI has emerged as a promising target to focus on. In this work, an iterative virtual high-throughput screening pipeline was proposed that can supplement current efforts to find inhibitors. The first iteration identified 11 compounds to test with 3 active for a hit-rate of 27.3%. The second iteration of the pipeline identified another 11 compounds to test with 7 active for a hit-rate of 63.6%. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1553-1565, 2018.
Collapse
Affiliation(s)
- Jonathan J Chen
- Dept. of Biology, The University of Akron, 302 Buchtel Common, Akron, OH, 44325
| | - Lyndsey N Schmucker
- Dept. of Chemical and Biomolecular Engineering, The University of Akron, 302 Buchtel Common, Akron, OH, 44325
| | - Donald P Visco
- Dept. of Chemical and Biomolecular Engineering, The University of Akron, 302 Buchtel Common, Akron, OH, 44325
| |
Collapse
|
41
|
Hsu HH, Huang CH, Lin ST. Fully Automated Molecular Design with Atomic Resolution for Desired Thermophysical Properties. Ind Eng Chem Res 2018. [DOI: 10.1021/acs.iecr.8b01004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hsuan-Hao Hsu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Chen-Hsuan Huang
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Shiang-Tai Lin
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
42
|
Chen JJ, Schmucker LN, Visco DP. Pharmaceutical Machine Learning: Virtual High-Throughput Screens Identifying Promising and Economical Small Molecule Inhibitors of Complement Factor C1s. Biomolecules 2018; 8:E24. [PMID: 29735903 PMCID: PMC6023033 DOI: 10.3390/biom8020024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 12/17/2022] Open
Abstract
When excessively activated, C1 is insufficiently regulated, which results in tissue damage. Such tissue damage causes the complement system to become further activated to remove the resulting tissue damage, and a vicious cycle of activation/tissue damage occurs. Current Food and Drug Administration approved treatments include supplemental recombinant C1 inhibitor, but these are extremely costly and a more economical solution is desired. In our work, we have utilized an existing data set of 136 compounds that have been previously tested for activity against C1. Using these compounds and the activity data, we have created models using principal component analysis, genetic algorithm, and support vector machine approaches to characterize activity. The models were then utilized to virtually screen the 72 million compound PubChem repository. This first round of virtual high-throughput screening identified many economical and promising inhibitor candidates, a subset of which was tested to validate their biological activity. These results were used to retrain the models and rescreen PubChem in a second round vHTS. Hit rates for the first round vHTS were 57%, while hit rates for the second round vHTS were 50%. Additional structure⁻property analysis was performed on the active and inactive compounds to identify interesting scaffolds for further investigation.
Collapse
Affiliation(s)
- Jonathan J Chen
- Department of Biology, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA.
| | - Lyndsey N Schmucker
- Department of Chemical and Biomolecular Engineering, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA.
| | - Donald P Visco
- Department of Chemical and Biomolecular Engineering, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA.
| |
Collapse
|
43
|
Mohanty PS, Bansal AK, Naaz F, Gupta UD, Dwivedi VD, Yadava U. Ribonucleotide reductase as a drug target against drug resistance Mycobacterium leprae: A molecular docking study. INFECTION GENETICS AND EVOLUTION 2018; 60:58-65. [PMID: 29454978 DOI: 10.1016/j.meegid.2018.02.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/28/2017] [Accepted: 02/09/2018] [Indexed: 10/18/2022]
Abstract
Leprosy is a chronic infection of skin and nerve caused by Mycobacterium leprae. The treatment is based on standard multi drug therapy consisting of dapsone, rifampicin and clofazamine. The use of rifampicin alone or with dapsone led to the emergence of rifampicin-resistant Mycobacterium leprae strains. The emergence of drug-resistant leprosy put a hurdle in the leprosy eradication programme. The present study aimed to predict the molecular model of ribonucleotide reductase (RNR), the enzyme responsible for biosynthesis of nucleotides, to screen new drugs for treatment of drug-resistant leprosy. The study was conducted by retrieving RNR of M. leprae from GenBank. A molecular 3D model of M. leprae was predicted using homology modelling and validated. A total of 325 characters were included in the analysis. The predicted 3D model of RNR showed that the ϕ and φ angles of 251 (96.9%) residues were positioned in the most favoured regions. It was also conferred that 18 α-helices, 6 β turns, 2 γ turns and 48 helix-helix interactions contributed to the predicted 3D structure. Virtual screening of Food and Drug Administration approved drug molecules recovered 1829 drugs of which three molecules, viz., lincomycin, novobiocin and telithromycin, were taken for the docking study. It was observed that the selected drug molecules had a strong affinity towards the modelled protein RNR. This was evident from the binding energy of the drug molecules towards the modelled protein RNR (-6.10, -6.25 and -7.10). Three FDA-approved drugs, viz., lincomycin, novobiocin and telithromycin, could be taken for further clinical studies to find their efficacy against drug resistant leprosy.
Collapse
Affiliation(s)
- Partha Sarathi Mohanty
- Department of Epidemiology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, M. Miyazaki Marg, Tajganj, Agra, India.
| | - Avi Kumar Bansal
- Department of Epidemiology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, M. Miyazaki Marg, Tajganj, Agra, India
| | - Farah Naaz
- Department of Epidemiology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, M. Miyazaki Marg, Tajganj, Agra, India
| | - Umesh Datta Gupta
- National JALMA Institute for Leprosy and Other Mycobacterial Diseases, M. Miyazaki Marg, Tajganj, Agra, India
| | - Vivek Dhar Dwivedi
- Department of Epidemiology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, M. Miyazaki Marg, Tajganj, Agra, India
| | - Umesh Yadava
- Department of Physics, Deen Dayal Upadhyay Gorakhpur University, Civil Lines, Gorakhpur, India
| |
Collapse
|
44
|
Hassanzadeh P, Atyabi F, Dinarvand R. Linkers: The key elements for the creation of efficient nanotherapeutics. J Control Release 2018; 270:260-267. [DOI: 10.1016/j.jconrel.2017.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/09/2017] [Accepted: 12/11/2017] [Indexed: 01/16/2023]
|
45
|
Suryanarayanan V, Panwar U, Chandra I, Singh SK. De Novo Design of Ligands Using Computational Methods. Methods Mol Biol 2018; 1762:71-86. [PMID: 29594768 DOI: 10.1007/978-1-4939-7756-7_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
De novo design technique is complementary to high-throughput virtual screening and is believed to contribute in pharmaceutical development of novel drugs with desired properties at a very low cost and time-efficient manner. In this chapter, we outline the basic de novo design concepts based on computational methods with an example.
Collapse
Affiliation(s)
- Venkatesan Suryanarayanan
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Umesh Panwar
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Ishwar Chandra
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
46
|
Ningthoujam SS, Talukdar AD, Sarker SD, Nahar L, Choudhury MD. Prediction of Medicinal Properties Using Mathematical Models and Computation, and Selection of Plant Materials. COMPUTATIONAL PHYTOCHEMISTRY 2018. [PMCID: PMC7149595 DOI: 10.1016/b978-0-12-812364-5.00002-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In any phytochemical drug discovery programme, one of the major issues is the appropriate selection of target plant species that may provide lead for new drug discovery and development. Conducting research without any working hypotheses may produce serendipitous discoveries, but the chances of success are much slimmer than any information-based targeted approach. Therefore, the plant selection process is extremely important for ensuring success. In recent years, there have been significant amounts of work involving applications of various mathematical modelling and computational techniques to predict medicinal properties of plants, and thus to provide information-based selection of plant materials for further studies aiming at potential drug discovery and development. This chapter presents an overview of methods and processes involved in plant selection by utilizing various mathematical modelling and computational techniques.
Collapse
Affiliation(s)
| | | | | | - Lutfun Nahar
- Liverpool John Moores University, Liverpool, United Kingdom
| | | |
Collapse
|
47
|
Identifying novel factor XIIa inhibitors with PCA-GA-SVM developed vHTS models. Eur J Med Chem 2017; 140:31-41. [DOI: 10.1016/j.ejmech.2017.08.056] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/21/2017] [Accepted: 08/23/2017] [Indexed: 01/18/2023]
|
48
|
Allen WJ, Fochtman BC, Balius TE, Rizzo RC. Customizable de novo design strategies for DOCK: Application to HIVgp41 and other therapeutic targets. J Comput Chem 2017; 38:2641-2663. [PMID: 28940386 DOI: 10.1002/jcc.25052] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/03/2017] [Indexed: 12/12/2022]
Abstract
De novo design can be used to explore vast areas of chemical space in computational lead discovery. As a complement to virtual screening, from-scratch construction of molecules is not limited to compounds in pre-existing vendor catalogs. Here, we present an iterative fragment growth method, integrated into the program DOCK, in which new molecules are built using rules for allowable connections based on known molecules. The method leverages DOCK's advanced scoring and pruning approaches and users can define very specific criteria in terms of properties or features to customize growth toward a particular region of chemical space. The code was validated using three increasingly difficult classes of calculations: (1) Rebuilding known X-ray ligands taken from 663 complexes using only their component parts (focused libraries), (2) construction of new ligands in 57 drug target sites using a library derived from ∼13M drug-like compounds (generic libraries), and (3) application to a challenging protein-protein interface on the viral drug target HIVgp41. The computational testing confirms that the de novo DOCK routines are robust and working as envisioned, and the compelling results highlight the potential utility for designing new molecules against a wide variety of important protein targets. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- William J Allen
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, 11794
| | - Brian C Fochtman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, 11794
| | - Trent E Balius
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, 94158
| | - Robert C Rizzo
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, 11794.,Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, 11794.,Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York, 11794
| |
Collapse
|
49
|
Mello JDFRE, Gomes RA, Vital-Fujii DG, Ferreira GM, Trossini GHG. Fragment-based drug discovery as alternative strategy to the drug development for neglected diseases. Chem Biol Drug Des 2017; 90:1067-1078. [PMID: 28547936 DOI: 10.1111/cbdd.13030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/12/2017] [Accepted: 05/08/2017] [Indexed: 12/24/2022]
Abstract
Neglected diseases (NDs) affect large populations and almost whole continents, representing 12% of the global health burden. In contrast, the treatment available today is limited and sometimes ineffective. Under this scenery, the Fragment-Based Drug Discovery emerged as one of the most promising alternatives to the traditional methods of drug development. This method allows achieving new lead compounds with smaller size of fragment libraries. Even with the wide Fragment-Based Drug Discovery success resulting in new effective therapeutic agents against different diseases, until this moment few studies have been applied this approach for NDs area. In this article, we discuss the basic Fragment-Based Drug Discovery process, brief successful ideas of general applications and show a landscape of its use in NDs, encouraging the implementation of this strategy as an interesting way to optimize the development of new drugs to NDs.
Collapse
Affiliation(s)
- Juliana da Fonseca Rezende E Mello
- Litec, Laboratório de Integração Entre Técnicas Computacionais e Experimentais no Planejamento de Fármacos, Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Renan Augusto Gomes
- Litec, Laboratório de Integração Entre Técnicas Computacionais e Experimentais no Planejamento de Fármacos, Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Drielli Gomes Vital-Fujii
- Litec, Laboratório de Integração Entre Técnicas Computacionais e Experimentais no Planejamento de Fármacos, Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Glaucio Monteiro Ferreira
- Litec, Laboratório de Integração Entre Técnicas Computacionais e Experimentais no Planejamento de Fármacos, Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil.,Programa de Pós-graduação em Toxicologia e Análises Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Gustavo Henrique Goulart Trossini
- Litec, Laboratório de Integração Entre Técnicas Computacionais e Experimentais no Planejamento de Fármacos, Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil.,Programa de Pós-graduação em Toxicologia e Análises Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
50
|
Liu T, Naderi M, Alvin C, Mukhopadhyay S, Brylinski M. Break Down in Order To Build Up: Decomposing Small Molecules for Fragment-Based Drug Design with eMolFrag. J Chem Inf Model 2017; 57:627-631. [PMID: 28346786 PMCID: PMC5433162 DOI: 10.1021/acs.jcim.6b00596] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
![]()
Constructing high-quality
libraries of molecular building blocks
is essential for successful fragment-based drug discovery. In this
communication, we describe eMolFrag, a new open-source
software to decompose organic compounds into nonredundant fragments
retaining molecular connectivity information. Given a collection of
molecules, eMolFrag generates a set of unique fragments
comprising larger moieties, bricks, and smaller linkers connecting
bricks. These building blocks can subsequently be used to construct
virtual screening libraries for targeted drug discovery. The robustness
and computational performance of eMolFrag is assessed
against the Directory of Useful Decoys, Enhanced database conducted
in serial and parallel modes with up to 16 computing cores. Further,
the application of eMolFrag in de novo drug design
is illustrated using the adenosine receptor. eMolFrag
is implemented in Python, and it is available as stand-alone software
and a web server at www.brylinski.org/emolfrag and https://github.com/liutairan/eMolFrag.
Collapse
Affiliation(s)
| | | | - Chris Alvin
- Department of Computer Science and Information Systems, Bradley University , Peoria, Illinois 61625, United States
| | | | | |
Collapse
|