1
|
Atabay M, Inci F, Saylan Y. Computational studies for the development of extracellular vesicle-based biosensors. Biosens Bioelectron 2025; 277:117275. [PMID: 39999607 DOI: 10.1016/j.bios.2025.117275] [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: 07/03/2024] [Revised: 12/25/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Cancer affects millions of people, and early detection and efficient treatment are two strong levers to hurdle this disease. Recent studies on exosomes, a subset of extracellular vesicles, have deliberately shown the potential to function as a biomarker or treatment tool, thereby attracting the attention of researchers who work on developing biosensors. Due to the ability of computational methods to predict of the behavior of biomolecules, the combination of experimental and computational methods would enhance the analytical performance of the biosensor, including sensitivity, accuracy, and specificity, even detecting such vesicles from bodily fluids. In this regard, the role of computational methods such as molecular docking, molecular dynamics simulation, and density functional theory is overviewed in the development of biosensors. This review highlights the investigations and studies that have been reported using these methods to design exosome-based biosensors. This review concludes with the role of the quantum mechanics/molecular mechanics method in the investigation of chemical processes of biomolecular systems and the deficiencies in using this approach to develop exosome-based biosensors. In addition, the artificial intelligence theory is explained briefly to show its importance in the study of these biosensors.
Collapse
Affiliation(s)
- Maryam Atabay
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, Turkey; Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Yeşeren Saylan
- Department of Chemistry, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
2
|
Katrib B, Adel A, Abadleh M, Daoud S, Taha M. Computational discovery of novel PI3KC2α inhibitors using structure-based pharmacophore modeling, machine learning and molecular dynamic simulation. J Mol Graph Model 2025; 137:109016. [PMID: 40112531 DOI: 10.1016/j.jmgm.2025.109016] [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: 12/12/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
PI3KC2α is a lipid kinase associated with cancer metastasis and thrombosis. In this study, we present a novel computational workflow integrating structure-based pharmacophore modeling, machine learning (ML), and molecular dynamics (MD) simulations to discover new PI3KC2α inhibitors. Key innovations include the generation of diverse pharmacophores from both crystallographic and docking-derived complexes, coupled with data augmentation via ligand conformational sampling to enhance ML robustness. The optimal model, developed using XGBoost with genetic function algorithm (GFA) and Shapley additive explanations (SHAP), identified four critical pharmacophores and three descriptors governing bioactivity. Virtual screening of the NCI database using these pharmacophores yielded three hits, with H_1 (NCI: 725847) demonstrating MD-derived binding stability and affinity comparable to the potent inhibitor PITCOIN1 (IC50 = 95 nM). This study represents the first application of a conformation-augmented ML framework to PI3KC2α inhibition, offering a blueprint for targeting underexplored kinases with limited structural data.
Collapse
Affiliation(s)
- Bana Katrib
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Ahmed Adel
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Mohammed Abadleh
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Mutasem Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
3
|
Awomuti A, Yu Z, Adesina O, Samuel OW, Mumbi AW, Yin D. Predictive modelling of peroxisome proliferator-activated receptor gamma (PPARγ) IC50 inhibition by emerging pollutants using light gradient boosting machine. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2025; 36:145-167. [PMID: 40126364 DOI: 10.1080/1062936x.2025.2478123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), a critical nuclear receptor, plays a pivotal role in regulating metabolic and inflammatory processes. However, various environmental contaminants can disrupt PPARγ function, leading to adverse health effects. This study introduces a novel approach to predict the inhibitory activity (IC50 values) of 140 chemical compounds across 13 categories, including pesticides, organochlorines, dioxins, detergents, flame retardants, and preservatives, on PPARγ. The predictive model, based on the light-gradient boosting machine (LightGBM) algorithm, was trained on a dataset of 1804 molecules showed r2 values of 0.82 and 0.59, Mean Absolute Error (MAE) of 0.38 and 0.58, and Root Mean Square Error (RMSE) of 0.54 and 0.76 for the training and test sets, respectively. This study provides novel insights into the interactions between emerging contaminants and PPARγ, highlighting the potential hazards and risks these chemicals may pose to public health and the environment. The ability to predict PPARγ inhibition by these hazardous contaminants demonstrates the value of this approach in guiding enhanced environmental toxicology research and risk assessment.
Collapse
Affiliation(s)
- A Awomuti
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, PR China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai, PR China
- UNEP-Tongji Institute of Environment for Sustainable Development, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Z Yu
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, PR China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai, PR China
- UNEP-Tongji Institute of Environment for Sustainable Development, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - O Adesina
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, PR China
- UNEP-Tongji Institute of Environment for Sustainable Development, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - O W Samuel
- School of Computing and Data Science Research Centre, University of Derby, Derby, UK
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - A W Mumbi
- Department of Engineering, Harper Adams University, Edgmond, UK
- Harper Adams Business School, Harper Adams University, Newport, UK
| | - D Yin
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, PR China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai, PR China
| |
Collapse
|
4
|
Du J, Xu G, Zhang W, Cong J, Si X, Wei B. Molecular mechanism underlying effect of D93 and D289 protonation states on inhibitor-BACE1 binding: exploration from multiple independent Gaussian accelerated molecular dynamics and deep learning. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:919-947. [PMID: 39512118 DOI: 10.1080/1062936x.2024.2419911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024]
Abstract
BACE1 has been regarded as an essential drug design target for treating Alzheimer's disease (AD). Multiple independent Gaussian accelerated molecular dynamics simulations (GaMD), deep learning (DL), and molecular mechanics general Born surface area (MM-GBSA) method are integrated to elucidate the molecular mechanism underlying the effect of D93 and D289 protonation on binding of inhibitors OV6 and 4B2 to BACE1. The GaMD trajectory-based DL successfully identifies significant function domains. Dynamic analysis shows that the protonation of D93 and D289 strongly affects the structural flexibility and dynamic behaviour of BACE1. Free energy landscapes indicate that inhibitor-bound BACE1s have more conformational states in the protonated states than the wild-type (WT) BACE1, and show more binding poses of inhibitors. Binding affinities calculated using the MM-GBSA method indicate that the protonation of D93 and D289 highly disturbs the binding ability of inhibitors to BACE1. In addition, the protonation of two residues significantly affects the hydrogen bonding interactions (HBIs) of OV6 and 4B2 with BACE1, altering their binding activity to BACE1. The binding hot spots of BACE1 recognized by residue-based free energy estimations provide rational targeting sites for drug design towards BACE1. This study is anticipated to provide theoretical aids for drug development towards treatment of AD.
Collapse
Affiliation(s)
- J Du
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - G Xu
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - W Zhang
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - J Cong
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - X Si
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - B Wei
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
5
|
Ece E, Ölmez K, Hacıosmanoğlu N, Atabay M, Inci F. Advancing 3D printed microfluidics with computational methods for sweat analysis. Mikrochim Acta 2024; 191:162. [PMID: 38411762 PMCID: PMC10899357 DOI: 10.1007/s00604-024-06231-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 02/28/2024]
Abstract
The intricate tapestry of biomarkers, including proteins, lipids, carbohydrates, vesicles, and nucleic acids within sweat, exhibits a profound correlation with the ones in the bloodstream. The facile extraction of samples from sweat glands has recently positioned sweat sampling at the forefront of non-invasive health monitoring and diagnostics. While extant platforms for sweat analysis exist, the imperative for portability, cost-effectiveness, ease of manufacture, and expeditious turnaround underscores the necessity for parameters that transcend conventional considerations. In this regard, 3D printed microfluidic devices emerge as promising systems, offering a harmonious fusion of attributes such as multifunctional integration, flexibility, biocompatibility, a controlled closed environment, and a minimal requisite analyte volume-features that leverage their prominence in the realm of sweat analysis. However, formidable challenges, including high throughput demands, chemical interactions intrinsic to the printing materials, size constraints, and durability concerns, beset the landscape of 3D printed microfluidic devices. Within this paradigm, we expound upon the foundational aspects of 3D printed microfluidic devices and proffer a distinctive perspective by delving into the computational study of printing materials utilizing density functional theory (DFT) and molecular dynamics (MD) methodologies. This multifaceted approach serves manifold purposes: (i) understanding the complexity of microfluidic systems, (ii) facilitating comprehensive analyses, (iii) saving both cost and time, (iv) improving design optimization, and (v) augmenting resolution. In a nutshell, the allure of 3D printing lies in its capacity for affordable and expeditious production, offering seamless integration of diverse components into microfluidic devices-a testament to their inherent utility in the domain of sweat analysis. The synergistic fusion of computational assessment methodologies with materials science not only optimizes analysis and production processes, but also expedites their widespread accessibility, ensuring continuous biomarker monitoring from sweat for end-users.
Collapse
Affiliation(s)
- Emre Ece
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
- Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey
| | - Kadriye Ölmez
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
- Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey
| | - Nedim Hacıosmanoğlu
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
- Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey
| | - Maryam Atabay
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
- Department of Chemistry, Hacettepe University, 06800, Ankara, Turkey
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey.
- Institute of Materials Science and Nanotechnology, Bilkent University, 06800, Ankara, Turkey.
| |
Collapse
|
6
|
Jaradat NJ, Hatmal M, Alqudah D, Taha MO. Computational workflow for discovering small molecular binders for shallow binding sites by integrating molecular dynamics simulation, pharmacophore modeling, and machine learning: STAT3 as case study. J Comput Aided Mol Des 2023; 37:659-678. [PMID: 37597062 DOI: 10.1007/s10822-023-00528-y] [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: 05/02/2023] [Accepted: 07/26/2023] [Indexed: 08/21/2023]
Abstract
STAT3 belongs to a family of seven transcription factors. It plays an important role in activating the transcription of various genes involved in a variety of cellular processes. High levels of STAT3 are detected in several types of cancer. Hence, STAT3 inhibition is considered a promising therapeutic anti-cancer strategy. However, since STAT3 inhibitors bind to the shallow SH2 domain of the protein, it is expected that hydration water molecules play significant role in ligand-binding complicating the discovery of potent binders. To remedy this issue, we herein propose to extract pharmacophores from molecular dynamics (MD) frames of a potent co-crystallized ligand complexed within STAT3 SH2 domain. Subsequently, we employ genetic function algorithm coupled with machine learning (GFA-ML) to explore the optimal combination of MD-derived pharmacophores that can account for the variations in bioactivity among a list of inhibitors. To enhance the dataset, the training and testing lists were augmented nearly a 100-fold by considering multiple conformers of the ligands. A single significant pharmacophore emerged after 188 ns of MD simulation to represent STAT3-ligand binding. Screening the National Cancer Institute (NCI) database with this model identified one low micromolar inhibitor most likely binds to the SH2 domain of STAT3 and inhibits this pathway.
Collapse
Affiliation(s)
- Nour Jamal Jaradat
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, P.O. Box 330127, Zarqa, 13133, Jordan
| | - Mamon Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, P.O. Box 330127, Zarqa, 13133, Jordan
| | - Dana Alqudah
- Cell Therapy Center, the University of Jordan, Amman, 11942, Jordan
| | - Mutasem Omar Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan.
| |
Collapse
|
7
|
Wang Y, Yang F, Yan D, Zeng Y, Wei B, Chen J, He W. Identification Mechanism of BACE1 on Inhibitors Probed by Using Multiple Separate Molecular Dynamics Simulations and Comparative Calculations of Binding Free Energies. Molecules 2023; 28:4773. [PMID: 37375328 DOI: 10.3390/molecules28124773] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
β-amyloid cleaving enzyme 1 (BACE1) is regarded as an important target of drug design toward the treatment of Alzheimer's disease (AD). In this study, three separate molecular dynamics (MD) simulations and calculations of binding free energies were carried out to comparatively determine the identification mechanism of BACE1 for three inhibitors, 60W, 954 and 60X. The analyses of MD trajectories indicated that the presence of three inhibitors influences the structural stability, flexibility and internal dynamics of BACE1. Binding free energies calculated by using solvated interaction energy (SIE) and molecular mechanics generalized Born surface area (MM-GBSA) methods reveal that the hydrophobic interactions provide decisive forces for inhibitor-BACE1 binding. The calculations of residue-based free energy decomposition suggest that the sidechains of residues L91, D93, S96, V130, Q134, W137, F169 and I179 play key roles in inhibitor-BACE1 binding, which provides a direction for future drug design toward the treatment of AD.
Collapse
Affiliation(s)
- Yiwen Wang
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
- School of Aeronautics, Shandong Jiaotong University, Jinan 250357, China
| | - Fen Yang
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
| | - Dongliang Yan
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Yalin Zeng
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
| | - Benzheng Wei
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China
| | - Jianzhong Chen
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Weikai He
- School of Information Science and Electrical Engineering, Shandong Jiaotong University, Jinan 250357, China
- School of Aeronautics, Shandong Jiaotong University, Jinan 250357, China
| |
Collapse
|
8
|
Al-Imam AM, Daoud S, Hatmal MM, Taha MO. Augmenting bioactivity by docking-generated multiple ligand poses to enhance machine learning and pharmacophore modelling: discovery of new TTK inhibitors as case study. Mol Inform 2023; 42:e2300022. [PMID: 37222400 DOI: 10.1002/minf.202300022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/01/2023] [Accepted: 05/23/2023] [Indexed: 05/25/2023]
Abstract
Dual specificity protein kinase threonine/Tyrosine kinase (TTK) is one of the mitotic kinases. High levels of TTK are detected in several types of cancer. Hence, TTK inhibition is considered a promising therapeutic anti-cancer strategy. In this work, we used multiple docked poses of TTK inhibitors to augment training data for machine learning QSAR modeling. Ligand-Receptor Contacts Fingerprints and docking scoring values were used as descriptor variables. Escalating docking-scoring consensus levels were scanned against orthogonal machine learners, and the best learners (Random Forests and XGBoost) were coupled with genetic algorithm and Shapley additive explanations (SHAP) to determine critical descriptors for predicting anti-TTK bioactivity and for pharmacophore generation. Three successful pharmacophores were deduced and subsequently used for in silico screening against the NCI database. A total of 14 hits were evaluated in vitro for their anti-TTK bioactivities. One hit of novel chemotype showed reasonable dose-response curve with experimental IC50 of 1.0 μM. The presented work indicates the validity of data augmentation using multiple docked poses for building successful machine learning models and pharmacophore hypotheses.
Collapse
Affiliation(s)
- Amenah M Al-Imam
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, 11492, Jordan
| | - Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Sciences Private University, Amman, Jordan
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, PO. Box, 330127, Zarqa 13133, Jordan
| | - Mutasem Omar Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, 11492, Jordan
| |
Collapse
|
9
|
Wang Y, Gao Q, Yao P, Yao Q, Zhang J. Multidimensional virtual screening approaches combined with drug repurposing to identify potential covalent inhibitors of SARS-CoV-2 3CL protease. J Biomol Struct Dyn 2023; 41:15262-15285. [PMID: 36961210 DOI: 10.1080/07391102.2023.2193994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/26/2023] [Indexed: 03/25/2023]
Abstract
The outbreak of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has caused an unprecedented global pandemic, and new cases are still on the rise due to the absence of effective medicines. However, developing new drugs within a short time is extremely difficult. Repurposing the existing drugs provides a fast and effective strategy to identify promising inhibitors. Here we focus on the screening of drugs database for discovering potential covalent inhibitors that target 3-chymotrypsin-like protease (3CLpro), an essential enzyme mediating viral replication and transcription. Firstly, we constructed a receptor-ligand pharmacophore model and verified it through decoy set. The importance of pharmacophore features was evaluated by combining molecular dynamics simulation with interaction analyses. Then, covalent docking was used to perform further screening. According to docking score and Prime/Molecular Mechanics Generalized Born Surface Area (MM-GBSA) score, total ten compounds obtained good scores and successfully established covalent bonds with the catalytic Cys145 residue. They also formed favorable interactions with key residues in active sites and closely integrated with 3CLpro with binding modes similar to known 3CLpro inhibitor. Finally, the top four hits DB08732, DB04653, DB01871 and DB07299 were further subjected to 100 ns molecular dynamics (MD) simulation and MM-GBSA binding free energy calculations. The results suggest that the four candidates show good binding affinities for 3CLpro, which warrants further evaluation for their in-vitro/in-vivo activities. Overall, our research methods provide a valuable reference for discovering promising inhibitors against SARS-CoV-2 and help to fight against the epidemic.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ying Wang
- Department of Physical Chemistry, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Qiushuang Gao
- Department of Physical Chemistry, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Peng Yao
- Department of Physical Chemistry, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Qizheng Yao
- School of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ji Zhang
- Department of Physical Chemistry, China Pharmaceutical University, Nanjing, People's Republic of China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
10
|
Jaradat NJ, Alshaer W, Hatmal M, Taha MO. Discovery of new STAT3 inhibitors as anticancer agents using ligand-receptor contact fingerprints and docking-augmented machine learning. RSC Adv 2023; 13:4623-4640. [PMID: 36760267 PMCID: PMC9896621 DOI: 10.1039/d2ra07007c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
STAT3 belongs to a family of seven vital transcription factors. High levels of STAT3 are detected in several types of cancer. Hence, STAT3 inhibition is considered a promising therapeutic anti-cancer strategy. In this work, we used multiple docked poses of STAT3 inhibitors to augment training data for machine learning QSAR modeling. Ligand-Receptor Contact Fingerprints and scoring values were implemented as descriptor variables. Escalating docking-scoring consensus levels were scanned against orthogonal machine learners, and the best learners (Random Forests and XGBoost) were coupled with genetic algorithm and Shapley additive explanations (SHAP) to identify critical descriptors that determine anti-STAT3 bioactivity to be translated into pharmacophore model(s). Two successful pharmacophores were deduced and subsequently used for in silico screening against the National Cancer Institute (NCI) database. A total of 26 hits were evaluated in vitro for their anti-STAT3 bioactivities. Out of which, three hits of novel chemotypes, showed cytotoxic IC50 values in the nanomolar range (35 nM to 6.7 μM). However, two are potent dihydrofolate reductase (DHFR) inhibitors and therefore should have significant indirect STAT3 inhibitory effects. The third hit (cytotoxic IC50 = 0.44 μM) is purely direct STAT3 inhibitor (devoid of DHFR activity) and caused, at its cytotoxic IC50, more than two-fold reduction in the expression of STAT3 downstream genes (c-Myc and Bcl-xL). The presented work indicates that the concept of data augmentation using multiple docked poses is a promising strategy for generating valid machine learning models capable of discriminating active from inactive compounds.
Collapse
Affiliation(s)
- Nour Jamal Jaradat
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan Amman 11492 Jordan +962 6 5339649 +962 6 5355000 ext. 23305
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan Amman 11942 Jordan
| | - Mamon Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University P.O. Box 330127 Zarqa 13133 Jordan
| | - Mutasem Omar Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan Amman 11492 Jordan +962 6 5339649 +962 6 5355000 ext. 23305
| |
Collapse
|
11
|
Abudayah A, Daoud S, Al-Sha'er MA, Omar Taha M. Pharmacophore Modeling of Targets Infested with Activity Cliffs via Molecular Dynamics Simulation Coupled with QSAR and Comparison with other Pharmacophore Generation Methods: KDR as Case Study. Mol Inform 2022; 41:e2200049. [PMID: 35973966 DOI: 10.1002/minf.202200049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/15/2022] [Indexed: 11/07/2022]
Abstract
Activity cliffs (ACs) are defined as pairs of structurally similar compounds with large difference in their potencies against certain biotarget. We recently proposed that potent AC members induce significant entropically-driven conformational modifications of the target that unveil additional binding interactions, while their weakly-potent counterparts are enthalpically-driven binders with little influence on the protein target. We herein propose to extract pharmacophores for ACs-infested target(s) from molecular dynamics (MD) frames of purely "enthalpic" potent binder(s) complexed within the particular target. Genetic function algorithm/machine learning (GFA/ML) can then be employed to search for the best possible combination of MD pharmacophore(s) capable of explaining bioactivity variations within a list of inhibitors. We compared the performance of this approach with established ligand-based and structure-based methods. Kinase inserts domain receptor (KDR) was used as a case study. KDR plays a crucial role in angiogenic signalling and its inhibitors have been approved in cancer treatment. Interestingly, GFA/ML selected, MD-based, pharmacophores were of comparable performances to ligand-based and structure-based pharmacophores. The resulting pharmacophores and QSAR models were used to capture hits from the national cancer institute list of compounds. The most active hit showed anti-KDR IC50 of 2.76 μM.
Collapse
Affiliation(s)
- Alaa Abudayah
- Faculty of Pharmacy, Zarqa University, Zarqa, 13132, Jordan
| | - Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Sciences Private University, 11931, Amman, Jordan
| | | | - Mutasem Omar Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, 11942, Amman, Jordan
| |
Collapse
|
12
|
Al-Tawil MF, Daoud S, Hatmal MM, Taha MO. Discovery of new Cdc2-like kinase 4 (CLK4) inhibitors via pharmacophore exploration combined with flexible docking-based ligand/receptor contact fingerprints and machine learning. RSC Adv 2022; 12:10686-10700. [PMID: 35424985 PMCID: PMC8982525 DOI: 10.1039/d2ra00136e] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/31/2022] [Indexed: 11/21/2022] Open
Abstract
Cdc2-like kinase 4 (CLK4) inhibitors are of potential therapeutic value in many diseases particularly cancer. In this study, we combined extensive ligand-based pharmacophore exploration, ligand-receptor contact fingerprints generated by flexible docking, physicochemical descriptors and machine learning-quantitative structure-activity relationship (ML-QSAR) analysis to investigate the pharmacophoric/binding requirements for potent CLK4 antagonists. Several ML methods were attempted to tie these properties with anti-CLK4 bioactivities including multiple linear regression (MLR), random forests (RF), extreme gradient boosting (XGBoost), probabilistic neural network (PNN), and support vector regression (SVR). A genetic function algorithm (GFA) was combined with each method for feature selection. Eventually, GFA-SVR was found to produce the best self-consistent and predictive model. The model selected three pharmacophores, three ligand-receptor contacts and two physicochemical descriptors. The GFA-SVR model and associated pharmacophore models were used to screen the National Cancer Institute (NCI) structural database for novel CLK4 antagonists. Three potent hits were identified with the best one showing an anti-CLK4 IC50 value of 57 nM.
Collapse
Affiliation(s)
- Mai Fayiz Al-Tawil
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan Amman 11942 Jordan
| | - Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Sciences Private University Amman Jordan
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University PO Box 330127 Zarqa 13133 Jordan
| | - Mutasem Omar Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan Amman 11942 Jordan
| |
Collapse
|
13
|
Mousa LA, Hatmal MM, Taha M. Exploiting activity cliffs for building pharmacophore models and comparison with other pharmacophore generation methods: sphingosine kinase 1 as case study. J Comput Aided Mol Des 2022; 36:39-62. [PMID: 35059939 DOI: 10.1007/s10822-021-00435-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022]
Abstract
Activity cliffs (ACs) are defined as closely analogous compounds of significant affinity discrepancies against certain biotarget. In this paper we propose to use AC pair(s) for extracting valid binding pharmacophores through exposing corresponding protein complexes to stochastic deformation/relaxation followed by applying genetic algorithm/machine learning (GA-ML) for selecting optimal pharmacophore(s) that best classify a long list of inhibitors. We compared the performances of ligand-based and structure-based pharmacophores with counterparts generated by this newly introduced technique. Sphingosine kinase 1 (SPHK-1) was used as case study. SPHK-1 is a lipid kinase that plays pivotal role in the regulation of a variety of biological processes including, cell growth, apoptosis, and inflammation. The new approach proved to yield pharmacophore and ML models of comparable accuracies to established ligand-based and structure-based pharmacophores. The resulting pharmacophores and ML models were used to capture hits from the national cancer institute list of compounds and predict their bioactivity categories. Two hits of novel chemotypes showed selective and low micromolar inhibitory IC50 values against SPHK-1.
Collapse
Affiliation(s)
- Lubabah A Mousa
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, 11942, Jordan
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, PO Box 330127, Zarqa, 13133, Jordan
| | - Mutasem Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
14
|
Mahmoud IS, Hatmal MM, Abuarqoub D, Esawi E, Zalloum H, Wehaibi S, Nsairat H, Alshaer W. 1,4-Naphthoquinone Is a Potent Inhibitor of IRAK1 Kinases and the Production of Inflammatory Cytokines in THP-1 Differentiated Macrophages. ACS OMEGA 2021; 6:25299-25310. [PMID: 34632188 PMCID: PMC8495692 DOI: 10.1021/acsomega.1c03081] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/03/2021] [Indexed: 05/27/2023]
Abstract
Quinones are a class of cyclic organic compounds that are widely distributed in nature and have been shown to exhibit anti-inflammatory, antioxidant, and anticancerous activities. However, the molecular mechanisms/signaling by which these molecules exert their effect are still not fully understood. In this study, a group of quinone-derived compounds were examined for their potential inhibitory effect against human IRAK1 and IRAK4 kinases in vitro. We have identified five compounds: 1,4-naphthoquinone, emodin, shikonin, plumbagin, and menadione (vitamin K3) as active and selective inhibitors of human IRAK1 enzyme in vitro. The biochemical binding and molecular interactions between the active compounds and IRAK1's catalytic site were demonstrated in silico using structural-based docking and dynamic simulation analysis. Also, 1,4-naphthoquinone was found to effectively inhibit the growth of cancer cell lines overexpressing IRAK1. Furthermore, 1,4-naphthoquinone potently suppressed the production and secretion of key proinflammatory cytokine proteins IL-8, IL-1β, IL-10, TNF-α, and IL-6 in LPS-stimulated PMA-induced human THP-1 macrophages. In conclusion, 1,4-naphthoquinone is an effective inhibitor of IRAK1 kinases and their mediated inflammatory cytokines production in LPS-stimulated PMA-induced human THP-1 macrophages.
Collapse
Affiliation(s)
- Ismail Sami Mahmoud
- Department
of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Ma’mon M. Hatmal
- Department
of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Duaa Abuarqoub
- Cell
Therapy Centre, The University of Jordan, Amman 11942, Jordan
- Department
of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical
Sciences, University of Petra, Amman 11180, Jordan
| | - Ezaldeen Esawi
- Cell
Therapy Centre, The University of Jordan, Amman 11942, Jordan
- Department
of Pathology and Laboratory Medicine, King
Hussein Cancer Center, Amman 11941, Jordan
| | - Hiba Zalloum
- Hamdi
Mango Centre for Scientific Research, The
University of Jordan, Amman 11942, Jordan
| | - Suha Wehaibi
- Cell
Therapy Centre, The University of Jordan, Amman 11942, Jordan
| | - Hamdi Nsairat
- Pharmacological
and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Walhan Alshaer
- Cell
Therapy Centre, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
15
|
Hatmal MM, Abuyaman O, Taha M. Docking-generated multiple ligand poses for bootstrapping bioactivity classifying Machine Learning: Repurposing covalent inhibitors for COVID-19-related TMPRSS2 as case study. Comput Struct Biotechnol J 2021; 19:4790-4824. [PMID: 34426763 PMCID: PMC8373588 DOI: 10.1016/j.csbj.2021.08.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 01/10/2023] Open
Abstract
In the present work we introduce the use of multiple docked poses for bootstrapping machine learning-based QSAR modelling. Ligand-receptor contact fingerprints are implemented as descriptor variables. We implemented this method for the discovery of potential inhibitors of the serine protease enzyme TMPRSS2 involved the infectivity of coronaviruses. Several machine learners were scanned, however, Xgboost, support vector machines (SVM) and random forests (RF) were the best with testing set accuracies reaching 90%. Three potential hits were identified upon using the method to scan known untested FDA approved drugs against TMPRSS2. Subsequent molecular dynamics simulation and covalent docking supported the results of the new computational approach.
Collapse
Affiliation(s)
- Ma'mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, PO Box 330127, Zarqa 13133, Jordan
| | - Omar Abuyaman
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, PO Box 330127, Zarqa 13133, Jordan
| | - Mutasem Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
16
|
Lombino J, Gulotta MR, De Simone G, Mekni N, De Rosa M, Carbone D, Parrino B, Cascioferro SM, Diana P, Padova A, Perricone U. Dynamic-shared Pharmacophore Approach as Tool to Design New Allosteric PRC2 Inhibitors, Targeting EED Binding Pocket. Mol Inform 2020; 40:e2000148. [PMID: 32833314 DOI: 10.1002/minf.202000148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/23/2020] [Indexed: 11/09/2022]
Abstract
The Polycomb Repressive complex 2 (PRC2) maintains a repressive chromatin state and silences many genes, acting as methylase on histone tails. This enzyme was found overexpressed in many types of cancer. In this work, we have set up a Computer-Aided Drug Design approach based on the allosteric modulation of PRC2. In order to minimize the possible bias derived from using a single set of coordinates within the protein-ligand complex, a dynamic workflow was developed. In details, molecular dynamic was used as tool to identify the most significant ligand-protein interactions from several crystallized protein structures. The identified features were used for the creation of dynamic pharmacophore models and docking grid constraints for the design of new PRC2 allosteric modulators. Our protocol was retrospectively validated using a dataset of active and inactive compounds, and the results were compared to the classic approaches, through ROC curves and enrichment factor. Our approach suggested some important interaction features to be adopted for virtual screening performance improvement.
Collapse
Affiliation(s)
- Jessica Lombino
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy.,Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Maria Rita Gulotta
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy.,Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | | | - Nedra Mekni
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy
| | - Maria De Rosa
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy
| | - Daniela Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Stella Maria Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | | | - Ugo Perricone
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy
| |
Collapse
|
17
|
Wang Y, Zhou Y, Shi S, Lu G, Lin X, Xie C, Liu D, Yao D. A rational design for improving the pepsin resistance of cellulase E4 isolated from T. fusca based on the evaluation of the transition complex and molecular structure. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2019.107417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
18
|
Identification of novel inhibitors of p-hydroxyphenylpyruvate dioxygenase using receptor-based virtual screening. J Taiwan Inst Chem Eng 2019. [DOI: 10.1016/j.jtice.2019.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
19
|
Tuffaha GO, Hatmal MM, Taha MO. Discovery of new JNK3 inhibitory chemotypes via QSAR-Guided selection of docking-based pharmacophores and comparison with other structure-based pharmacophore modeling methods. J Mol Graph Model 2019; 91:30-51. [PMID: 31158642 DOI: 10.1016/j.jmgm.2019.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 12/21/2022]
Abstract
The kinase c-Jun N-terminal Kinase 3 (JNK3) plays an important role in neurodegenerative diseases. JNK3 inhibitors have shown promising results in treating Alzheimer's and Parkinson's diseases. This prompted us to model this interesting target via three established structure-based computational workflows; namely, docking-based Comparative Intermolecular Contacts Analysis (db-CICA), pharmacophore modeling via molecular-dynamics based Ligand-Receptor Contact Analysis (md-LRCA), and QSAR-guided selection of crystallographic pharmacophores. Moreover, we compared the performances of resulting pharmacophores against binding models generated via a newly introduced technique, namely, QSAR-guided selection of docking-based pharmacophores. The resulting pharmacophores were validated by receiver operating characteristic (ROC) curve analysis and used as virtual search queries to screen the National Cancer Institute (NCI) database for promising anti-JNK3 hits of novel chemotypes. Eleven nanomolar and low micromolar hits were identified, three of which were captured by QSAR-guided docking-based pharmacophores.
Collapse
Affiliation(s)
- Ghada Omar Tuffaha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Mutasem O Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan.
| |
Collapse
|
20
|
Liu WS, Wang RR, Li WY, Rong M, Liu CL, Ma Y, Wang RL. Investigating the reason for loss-of-function of Src homology 2 domain-containing protein tyrosine phosphatase 2 (SHP2) caused by Y279C mutation through molecular dynamics simulation. J Biomol Struct Dyn 2019; 38:2509-2520. [DOI: 10.1080/07391102.2019.1634641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Wen-Shan Liu
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, China
| | - Rui-Rui Wang
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, China
| | - Wei-Ya Li
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, China
| | - Mei Rong
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, China
| | - Chi-Lu Liu
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, China
| | - Ying Ma
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, China
| | - Run-Ling Wang
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, China
| |
Collapse
|
21
|
Wang RR, Liu WS, Zhou L, Ma Y, Wang RL. Probing the acting mode and advantages of RMC-4550 as an Src-homology 2 domain-containing protein tyrosine phosphatase (SHP2) inhibitor at molecular level through molecular docking and molecular dynamics. J Biomol Struct Dyn 2019; 38:1525-1538. [DOI: 10.1080/07391102.2019.1613266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Rui-Rui Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wen-Shan Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Liang Zhou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Run-Ling Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
22
|
Chaudhuri A, Hudait N, Chakraborty SS. Pharmacophore modeling coupled with molecular dynamic simulation approach to identify new leads for meprin-β metalloprotease. Comput Biol Chem 2019; 80:292-306. [PMID: 31054542 DOI: 10.1016/j.compbiolchem.2019.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 11/26/2022]
Abstract
Human meprin beta metalloprotease, a small subgroup of the astacin family, is a potent drug target for the treatment of several disorders such as fibrosis, neurodegenerative disease in particular Alzheimer and inflammatory bowel diseases. In this study, a ligand-based pharmacophore approach has been used for the selection of potentially active compounds to understand the inhibitory activities of meprin-β by using the sulfonamide scaffold based inhibitors. Using this dataset, a pharmacophore model (Hypo1) was selected on the basis of a highest correlation coefficient (0.959), lowest total cost (105.89) and lowest root mean square deviation (1.31 Å) values. All the pharmacophore hypotheses generated from the candidate inhibitors comprised four features: two hydrogen-bond acceptor, one hydrogen-bond donor and one zinc binder feature. The best validated pharmacophore model (Hypo1) was used for virtual screening of compounds from several databases. The selective hit compounds were filtered by drug likeness property, acceptable ADMET profile, molecular docking and DFT study. Molecular dynamic simulations with the final 10 hit compounds revealed that a large number of non-covalent interactions were formed with the active site and specificity sub-pockets of the meprin beta metalloprotease. This study assists in the development of the new lead molecules as well as gives a better understanding of their interaction with meprin-β.
Collapse
Affiliation(s)
- Ankur Chaudhuri
- Department of Microbiology, West Bengal State University, Barasat, Berunanpukuria, P.O. Malikapur, North 24 Parganas, Kolkata, 700126, West Bengal, India
| | - Nandagopal Hudait
- Department of Microbiology, West Bengal State University, Barasat, Berunanpukuria, P.O. Malikapur, North 24 Parganas, Kolkata, 700126, West Bengal, India; Department of Chemistry, West Bengal State University, Barasat, Berunanpukuria, P.O. Malikapur, North 24 Parganas, Kolkata, 700126, West Bengal, India
| | - Sibani Sen Chakraborty
- Department of Microbiology, West Bengal State University, Barasat, Berunanpukuria, P.O. Malikapur, North 24 Parganas, Kolkata, 700126, West Bengal, India.
| |
Collapse
|
23
|
Al-Sha'er MA, Al-Aqtash RA, Taha MO. Discovery of New Phosphoinositide 3-kinase Delta (PI3Kδ) Inhibitors via Virtual Screening using Crystallography-derived Pharmacophore Modelling and QSAR Analysis. Med Chem 2019; 15:588-601. [PMID: 30799792 DOI: 10.2174/1573406415666190222125333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND PI3Kδ is predominantly expressed in hematopoietic cells and participates in the activation of leukocytes. PI3Kδ inhibition is a promising approach for treating inflammatory diseases and leukocyte malignancies. Accordingly, we decided to model PI3Kδ binding. METHODS Seventeen PI3Kδ crystallographic complexes were used to extract 94 pharmacophore models. QSAR modelling was subsequently used to select the superior pharmacophore(s) that best explain bioactivity variation within a list of 79 diverse inhibitors (i.e., upon combination with other physicochemical descriptors). RESULTS The best QSAR model (r2 = 0.71, r2 LOO = 0.70, r2 press against external testing list of 15 compounds = 0.80) included a single crystallographic pharmacophore of optimal explanatory qualities. The resulting pharmacophore and QSAR model were used to screen the National Cancer Institute (NCI) database for new PI3Kδ inhibitors. Two hits showed low micromolar IC50 values. CONCLUSION Crystallography-based pharmacophores were successfully combined with QSAR analysis for the identification of novel PI3Kδ inhibitors.
Collapse
Affiliation(s)
- Mahmoud A Al-Sha'er
- Faculty of Pharmacy, Zarqa University, P.O. Box 132222, Zarqa, 13132, Jordan
| | - Rua'a A Al-Aqtash
- Faculty of Pharmacy, Zarqa University, P.O. Box 132222, Zarqa, 13132, Jordan
| | - Mutasem O Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
24
|
Hatmal MM, Taha MO. Combining Stochastic Deformation/Relaxation and Intermolecular Contacts Analysis for Extracting Pharmacophores from Ligand-Receptor Complexes. J Chem Inf Model 2018; 58:879-893. [PMID: 29529367 DOI: 10.1021/acs.jcim.7b00708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We previously combined molecular dynamics (classical or simulated annealing) with ligand-receptor contacts analysis as a means to extract valid pharmacophore model(s) from single ligand-receptor complexes. However, molecular dynamics methods are computationally expensive and time-consuming. Here we describe a novel method for extracting valid pharmacophore model(s) from a single crystallographic structure within a reasonable time scale. The new method is based on ligand-receptor contacts analysis following energy relaxation of a predetermined set of randomly deformed complexes generated from the targeted crystallographic structure. Ligand-receptor contacts maintained across many deformed/relaxed structures are assumed to be critical and used to guide pharmacophore development. This methodology was implemented to develop valid pharmacophore models for PI3K-γ, RENIN, and JAK1. The resulting pharmacophore models were validated by receiver operating characteristic (ROC) analysis against inhibitors extracted from the CHEMBL database. Additionally, we implemented pharmacophores extracted from PI3K-γ to search for new inhibitors from the National Cancer Institute list of compounds. The process culminated in new PI3K-γ/mTOR inhibitory leads of low micromolar IC50s.
Collapse
Affiliation(s)
- Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences , The Hashemite University , P.O. Box 330127 , Zarqa 13133 , Jordan
| | - Mutasem O Taha
- Drug Discovery Unit, Department of Pharmaceutical Sciences, Faculty of Pharmacy , University of Jordan , Amman 11942 , Jordan
| |
Collapse
|
25
|
Hatmal MM, Taha MO. Simulated annealing molecular dynamics and ligand-receptor contacts analysis for pharmacophore modeling. Future Med Chem 2017; 9:1141-1159. [PMID: 28722471 DOI: 10.4155/fmc-2017-0061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/20/2017] [Indexed: 12/14/2022] Open
Abstract
AIM Ligand-based pharmacophore modeling requires long list of inhibitors, while pharmacophores based on single ligand-receptor crystallographic structure can be too restricted or promiscuous. METHODOLOGY This prompted us to combine simulated annealing molecular dynamics (SAMD) with ligand-receptor contacts analysis as means to construct pharmacophore model(s) from single ligand-receptor complex. Ligand-receptor contacts that survive numerous heating-cooling SAMD cycles are considered critical and are used to guide pharmacophore development. RESULTS This methodology was implemented to develop pharmacophores for acetylcholinesterase and protein kinase C-θ. The resulting models were validated by receiver-operating characteristic analysis and in vitro bioassay. Assay identified four new protein kinase C-θ inhibitors among captured hits, two of which exhibited nanomolar potencies. CONCLUSION The results illustrate the ability of the new method to extract valid pharmacophores from single ligand-protein complex.
Collapse
Affiliation(s)
- Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan
| | - Mutasem O Taha
- Drug Discovery Unit, Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman 11942, Jordan
| |
Collapse
|