1
|
Wei G, Zhao F, Zhang Z, Regenstein JM, Sang Y, Zhou P. Identification and characterization of umami-ACE inhibitory peptides from traditional fermented soybean curds. Food Chem 2025; 465:142160. [PMID: 39579405 DOI: 10.1016/j.foodchem.2024.142160] [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: 06/21/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Fermented soybean curds (FSC) are popular because of its umami taste. Its bioactivities are of interest. Peptides in FSC were identified using nano-HPLC-MS/MS, and 11 candidate peptides showing potential umami and ACE inhibitory activities were screened using various databases. Pharmacophore model analysis showed their high probability of ACE inhibition with fit values >2, which showed the peptides bound to umami receptors and ACE mainly through hydrogen bond, and electrostatic and hydrophobic interactions. Additionally, their docking and interaction energy were independent of the peptide length. Three high umami-ACE inhibitory peptides (VE, FEF, and WEEF) were synthesized. Their umami thresholds were WEEF (0.32 mM) < FEF (0.55 mM) < VE (1.10 mM), while their IC50 were WEEF (85 ± 2 μM) < FEF (170 ± 10 μM) < VE (205 ± 5 μM). NO and ET-1 production were dose-dependent with WEEF showing the best ACE inhibitory activity. The results allowed identification of effective umami agents and ACE inhibitory peptides from fermented soybean products. It could also be useful method for screening potential umami-ACE inhibitory peptides.
Collapse
Affiliation(s)
- Guanmian Wei
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei Province 071001, PR China; School of Food Science, Jiangnan University, Wuxi, Jiangsu Province 214122, PR China
| | - Feiran Zhao
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei Province 071001, PR China
| | - Ziyi Zhang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei Province 071001, PR China
| | - Joe M Regenstein
- Department of Food Science, Cornell University, Ithaca, NY 14853-7201, USA
| | - Yaxin Sang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei Province 071001, PR China.
| | - Peng Zhou
- School of Food Science, Jiangnan University, Wuxi, Jiangsu Province 214122, PR China.
| |
Collapse
|
2
|
Dong C, Huang YP, Lin X, Zhang H, Gao YQ. DSDPFlex: Flexible-Receptor Docking with GPU Acceleration. J Chem Inf Model 2024; 64:8537-8548. [PMID: 39514506 DOI: 10.1021/acs.jcim.4c01715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Molecular docking is an essential tool in structure-based drug discovery, widely utilized to model ligand-protein interactions and enrich potential hits. Among the different docking strategies, semiflexible docking (rigid-receptor and flexible-ligand model) is the most popular, benefiting from its balance of docking accuracy and speed. However, this approach ignores the conformational changes of proteins and hence demands suitable protein conformations as input. When the binding interaction adheres to an induced-fit model, flexible methods such as molecular dynamics simulation can be utilized, but they are computationally demanding. To balance between speed and accuracy, the flexible docking approach is an effective choice, as exemplified by AutoDock Vina and AutoDockFR, which treat selected protein side chains as flexible parts. However, the efficiency of flexible docking methods is yet to be improved for virtual screening usage. In this article, we introduce DSDPFlex, an improved flexible-receptor docking method accelerated by GPU parallelization. Beyond acceleration, optimizations with respect to sampling, scoring, and search space are implemented in DSDPFlex to further improve its capability in flexible tasks. In cross-docking evaluation, DSDPFlex demonstrates superior accuracy compared to AutoDock Vina and is 100 times faster than Vina in flexible-receptor tasks. We also show the advantage of flexible-receptor methods on suboptimal pockets and validate the advantage of DSDPFlex in screening on apo and AlphaFold2-predicted structures. With improvements in both efficiency and accuracy, DSDPFlex is expected to hold potential in future docking-based studies.
Collapse
Affiliation(s)
- Chengwei Dong
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yu-Peng Huang
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xiaohan Lin
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Hong Zhang
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102200, China
| | - Yi Qin Gao
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102200, China
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, China
| |
Collapse
|
3
|
Zheng G, Wu D, Wei X, Xu D, Mao T, Yan D, Han W, Shang X, Chen Z, Qiu J, Tang K, Cao Z, Qiu T. PbsNRs: predict the potential binders and scaffolds for nuclear receptors. Brief Bioinform 2024; 26:bbae710. [PMID: 39798999 PMCID: PMC11724720 DOI: 10.1093/bib/bbae710] [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: 07/24/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025] Open
Abstract
Nuclear receptors (NRs) are a class of essential proteins that regulate the expression of specific genes and are associated with multiple diseases. In silico methods for prescreening potential NR binders with predictive binding ability are highly desired for NR-related drug development but are rarely reported. Here, we present the PbsNRs (Predicting binders and scaffolds for Nuclear Receptors), a user-friendly web server designed to predict the potential NR binders and scaffolds through proteochemometric modeling. The utility of PbsNRs was systemically evaluated using both chemical compounds and natural products. Results indicated that PbsNRs achieved a good prediction performance for chemical compounds on internal (ROC-AUC = 0.906, where ROC is Receiver-Operating Characteristic curve and AUC is the Area Under the Curve) and external (ROC-AUC = 0.783) datasets, outperforming both compound-ligand interaction tools and NR-specific predictors. PbsNRs also successfully identified bioactive chemical scaffolds for NRs by screening massive natural products. Moreover, the predicted bioactive and inactive natural products for NR2B1 were experimentally validated using biosensors. PbsNRs not only aids in screening potential therapeutic NR binders but also helps discover the essential molecular scaffold and guide the drug discovery for multiple NR-related diseases. The PbsNRs web server is available at http://pbsnrs.badd-cao.net.
Collapse
Affiliation(s)
- Genhui Zheng
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
- Oden Institute for Computational Engineering and Sciences (ICES), University of Texas at Austin, No. 201 East 24th Street, Austin 78712, TX, United States
| | - Dingfeng Wu
- National Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou 310052, China
| | - Xiuxia Wei
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Dongpo Xu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Tiantian Mao
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Deyu Yan
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Wenhao Han
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Xiaoxiao Shang
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
- Department of Mathematics and Statistics, McGill University, 805 Sherbrooke Street West, Montreal H3A 0B9, Quebec, Canada
| | - Zikun Chen
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Jingxuan Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Kailin Tang
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Zhiwei Cao
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
| | - Tianyi Qiu
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
4
|
Chiang CH, Wang Y, Hussain A, Brooks CL, Narayan ARH. Ancestral Sequence Reconstruction to Enable Biocatalytic Synthesis of Azaphilones. J Am Chem Soc 2024; 146:30194-30203. [PMID: 39441831 DOI: 10.1021/jacs.4c08761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Biocatalysis can be powerful in organic synthesis but is often limited by enzymes' substrate scope and selectivity. Developing a biocatalytic step involves identifying an initial enzyme for the target reaction followed by optimization through rational design, directed evolution, or both. These steps are time consuming, resource-intensive, and require expertise beyond typical organic chemistry. Thus, an effective strategy for streamlining the process from enzyme identification to implementation is essential to expanding biocatalysis. Here, we present a strategy combining bioinformatics-guided enzyme mining and ancestral sequence reconstruction (ASR) to resurrect enzymes for biocatalytic synthesis. Specifically, we achieve an enantioselective synthesis of azaphilone natural products using two ancestral enzymes: a flavin-dependent monooxygenase (FDMO) for stereodivergent oxidative dearomatization and a substrate-selective acyltransferase (AT) for the acylation of the enzymatically installed hydroxyl group. This cascade, stereocomplementary to established chemoenzymatic routes, expands access to enantiomeric linear tricyclic azaphilones. By leveraging the co-occurrence and coevolution of FDMO and AT in azaphilone biosynthetic pathways, we identified an AT candidate, CazE, and addressed its low solubility and stability through ASR, obtaining a more soluble, stable, promiscuous, and reactive ancestral AT (AncAT). Sequence analysis revealed AncAT as a chimeric composition of its descendants with enhanced reactivity likely due to ancestral promiscuity. Flexible receptor docking and molecular dynamics simulations showed that the most reactive AncAT promotes a reactive geometry between substrates. We anticipate that our bioinformatics-guided, ASR-based approach can be broadly applied in target-oriented synthesis, reducing the time required to develop biocatalytic steps and efficiently access superior biocatalysts.
Collapse
Affiliation(s)
- Chang-Hwa Chiang
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ye Wang
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Azam Hussain
- Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Charles L Brooks
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Enhanced Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Alison R H Narayan
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
5
|
Hwang W, Austin SL, Blondel A, Boittier ED, Boresch S, Buck M, Buckner J, Caflisch A, Chang HT, Cheng X, Choi YK, Chu JW, Crowley MF, Cui Q, Damjanovic A, Deng Y, Devereux M, Ding X, Feig MF, Gao J, Glowacki DR, Gonzales JE, Hamaneh MB, Harder ED, Hayes RL, Huang J, Huang Y, Hudson PS, Im W, Islam SM, Jiang W, Jones MR, Käser S, Kearns FL, Kern NR, Klauda JB, Lazaridis T, Lee J, Lemkul JA, Liu X, Luo Y, MacKerell AD, Major DT, Meuwly M, Nam K, Nilsson L, Ovchinnikov V, Paci E, Park S, Pastor RW, Pittman AR, Post CB, Prasad S, Pu J, Qi Y, Rathinavelan T, Roe DR, Roux B, Rowley CN, Shen J, Simmonett AC, Sodt AJ, Töpfer K, Upadhyay M, van der Vaart A, Vazquez-Salazar LI, Venable RM, Warrensford LC, Woodcock HL, Wu Y, Brooks CL, Brooks BR, Karplus M. CHARMM at 45: Enhancements in Accessibility, Functionality, and Speed. J Phys Chem B 2024; 128:9976-10042. [PMID: 39303207 PMCID: PMC11492285 DOI: 10.1021/acs.jpcb.4c04100] [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] [Received: 06/20/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
Since its inception nearly a half century ago, CHARMM has been playing a central role in computational biochemistry and biophysics. Commensurate with the developments in experimental research and advances in computer hardware, the range of methods and applicability of CHARMM have also grown. This review summarizes major developments that occurred after 2009 when the last review of CHARMM was published. They include the following: new faster simulation engines, accessible user interfaces for convenient workflows, and a vast array of simulation and analysis methods that encompass quantum mechanical, atomistic, and coarse-grained levels, as well as extensive coverage of force fields. In addition to providing the current snapshot of the CHARMM development, this review may serve as a starting point for exploring relevant theories and computational methods for tackling contemporary and emerging problems in biomolecular systems. CHARMM is freely available for academic and nonprofit research at https://academiccharmm.org/program.
Collapse
Affiliation(s)
- Wonmuk Hwang
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
- Department
of Materials Science and Engineering, Texas
A&M University, College Station, Texas 77843, United States
- Department
of Physics and Astronomy, Texas A&M
University, College Station, Texas 77843, United States
- Center for
AI and Natural Sciences, Korea Institute
for Advanced Study, Seoul 02455, Republic
of Korea
| | - Steven L. Austin
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Arnaud Blondel
- Institut
Pasteur, Université Paris Cité, CNRS UMR3825, Structural
Bioinformatics Unit, 28 rue du Dr. Roux F-75015 Paris, France
| | - Eric D. Boittier
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Stefan Boresch
- Faculty of
Chemistry, Department of Computational Biological Chemistry, University of Vienna, Wahringerstrasse 17, 1090 Vienna, Austria
| | - Matthias Buck
- Department
of Physiology and Biophysics, Case Western
Reserve University, School of Medicine, Cleveland, Ohio 44106, United States
| | - Joshua Buckner
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Amedeo Caflisch
- Department
of Biochemistry, University of Zürich, CH-8057 Zürich, Switzerland
| | - Hao-Ting Chang
- Institute
of Bioinformatics and Systems Biology, National
Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan, ROC
| | - Xi Cheng
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yeol Kyo Choi
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Jhih-Wei Chu
- Institute
of Bioinformatics and Systems Biology, Department of Biological Science
and Technology, Institute of Molecular Medicine and Bioengineering,
and Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung
University, Hsinchu 30010, Taiwan,
ROC
| | - Michael F. Crowley
- Renewable
Resources and Enabling Sciences Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| | - Qiang Cui
- Department
of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
- Department
of Physics, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
- Department
of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, Massachusetts 02215, United States
| | - Ana Damjanovic
- Department
of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department
of Physics and Astronomy, Johns Hopkins
University, Baltimore, Maryland 21218, United States
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yuqing Deng
- Shanghai
R&D Center, DP Technology, Ltd., Shanghai 201210, China
| | - Mike Devereux
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Xinqiang Ding
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Michael F. Feig
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| | - Jiali Gao
- School
of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen, Guangdong 518055, China
- Department
of Chemistry and Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - David R. Glowacki
- CiTIUS
Centro Singular de Investigación en Tecnoloxías Intelixentes
da USC, 15705 Santiago de Compostela, Spain
| | - James E. Gonzales
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Mehdi Bagerhi Hamaneh
- Department
of Physiology and Biophysics, Case Western
Reserve University, School of Medicine, Cleveland, Ohio 44106, United States
| | | | - Ryan L. Hayes
- Department
of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, Irvine, California 92697, United States
| | - Jing Huang
- Key Laboratory
of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Yandong Huang
- College
of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Phillip S. Hudson
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
- Medicine
Design, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Wonpil Im
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Shahidul M. Islam
- Department
of Chemistry, Delaware State University, Dover, Delaware 19901, United States
| | - Wei Jiang
- Computational
Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Michael R. Jones
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Silvan Käser
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Fiona L. Kearns
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Nathan R. Kern
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Jeffery B. Klauda
- Department
of Chemical and Biomolecular Engineering, Institute for Physical Science
and Technology, Biophysics Program, University
of Maryland, College Park, Maryland 20742, United States
| | - Themis Lazaridis
- Department
of Chemistry, City College of New York, New York, New York 10031, United States
| | - Jinhyuk Lee
- Disease
Target Structure Research Center, Korea
Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
- Department
of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34141, Republic of Korea
| | - Justin A. Lemkul
- Department
of Biochemistry, Virginia Polytechnic Institute
and State University, Blacksburg, Virginia 24061, United States
| | - Xiaorong Liu
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yun Luo
- Department
of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California 91766, United States
| | - Alexander D. MacKerell
- Department
of Pharmaceutical Sciences, University of
Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Dan T. Major
- Department
of Chemistry and Institute for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Markus Meuwly
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
- Department
of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Kwangho Nam
- Department
of Chemistry and Biochemistry, University
of Texas at Arlington, Arlington, Texas 76019, United States
| | - Lennart Nilsson
- Karolinska
Institutet, Department of Biosciences and
Nutrition, SE-14183 Huddinge, Sweden
| | - Victor Ovchinnikov
- Harvard
University, Department of Chemistry
and Chemical Biology, Cambridge, Massachusetts 02138, United States
| | - Emanuele Paci
- Dipartimento
di Fisica e Astronomia, Universitá
di Bologna, Bologna 40127, Italy
| | - Soohyung Park
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Richard W. Pastor
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Amanda R. Pittman
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Carol Beth Post
- Borch Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Samarjeet Prasad
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jingzhi Pu
- Department
of Chemistry and Chemical Biology, Indiana
University Indianapolis, Indianapolis, Indiana 46202, United States
| | - Yifei Qi
- School
of Pharmacy, Fudan University, Shanghai 201203, China
| | | | - Daniel R. Roe
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Benoit Roux
- Department
of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | | | - Jana Shen
- Department
of Pharmaceutical Sciences, University of
Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Andrew C. Simmonett
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Alexander J. Sodt
- Eunice
Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kai Töpfer
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Meenu Upadhyay
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Arjan van der Vaart
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | | | - Richard M. Venable
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Luke C. Warrensford
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - H. Lee Woodcock
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Yujin Wu
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Charles L. Brooks
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bernard R. Brooks
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Martin Karplus
- Harvard
University, Department of Chemistry
and Chemical Biology, Cambridge, Massachusetts 02138, United States
- Laboratoire
de Chimie Biophysique, ISIS, Université
de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
6
|
Lopez DH, Yalkowsky SH. The Relationship Between Molecular Symmetry and Physicochemical Properties Involving Boiling and Melting of Organic Compounds. Pharm Res 2023; 40:2801-2815. [PMID: 37561323 DOI: 10.1007/s11095-023-03576-z] [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: 04/20/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVE AND METHODS The reliable estimation of phase transition physicochemical properties such as boiling and melting points can be valuable when designing compounds with desired physicochemical properties. This study explores the role of external rotational symmetry in determining boiling and melting points of select organic compounds. Using experimental data from the literature, the entropies of boiling and fusion were obtained for 541 compounds. The statistical significance of external rotational symmetry number on entropies of phase change was determined by using multiple linear regression. In addition, a series of aliphatic hydrocarbons, polysubstituted benzenes, and di-substituted napthalenes are used as examples to demonstrate the role of external symmetry on transition temperature. RESULTS The results reveal that symmetry is not well correlated with boiling point but is statistically significant in melting point. CONCLUSION The lack of correlation between the boiling point and the symmetry number reflects the fact that molecules have a high degree of rotational freedom in both the liquid and the vapor. On the other hand, the strong relationship between symmetry and melting point reflects the fact that molecules are rotationally restricted in the crystal but not in the liquid. Since the symmetry number is equal to the number of ways that the molecule can be properly oriented for incorporation into the crystal lattice, it is a significant determinant of the melting point.
Collapse
Affiliation(s)
- David Humberto Lopez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA.
| | - Samuel Hyman Yalkowsky
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
7
|
Yasir M, Park J, Lee Y, Han ET, Park WS, Han JH, Kwon YS, Lee HJ, Chun W. Discovery of GABA Aminotransferase Inhibitors via Molecular Docking, Molecular Dynamic Simulation, and Biological Evaluation. Int J Mol Sci 2023; 24:16990. [PMID: 38069313 PMCID: PMC10707509 DOI: 10.3390/ijms242316990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
γ-Aminobutyric acid aminotransferase (GABA-AT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that degrades γ-aminobutyric (GABA) in the brain. GABA is an important inhibitory neurotransmitter that plays important neurological roles in the brain. Therefore, GABA-AT is an important drug target that regulates GABA levels. Novel and potent drug development to inhibit GABA-AT is still a very challenging task. In this study, we aimed to devise novel and potent inhibitors against GABA-AT using computer-aided drug design (CADD) tools. Since the crystal structure of human GABA-AT was not yet available, we utilized a homologous structure derived from our previously published paper. To identify highly potent compounds relative to vigabatrin, an FDA-approved drug against human GABA-AT, we developed a pharmacophore analysis protocol for 530,000 Korea Chemical Bank (KCB) compounds and selected the top 50 compounds for further screening. Preliminary biological analysis was carried out for these 50 compounds and 16 compounds were further assessed. Subsequently, molecular docking, molecular dynamics (MD) simulations, and binding free energy calculations were carried out. In the results, four predicted compounds, A07, B07, D08, and H08, were found to be highly potent and were further evaluated by a biological activity assay to confirm the results of the GABA-AT activity inhibition assay.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| | - Yuno Lee
- Drug Information Platform Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea;
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (E.-T.H.); (J.-H.H.)
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea;
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (E.-T.H.); (J.-H.H.)
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Hee-Jae Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.); (H.-J.L.)
| |
Collapse
|
8
|
Wang Z, Ma Q, Zheng P, Xie S, Yao K, Zhang J, Shao B, Jiang H. Generation of broad-spectrum recombinant antibody and construction of colorimetric immunoassay for tropane alkaloids: Recognition mechanism and application. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132247. [PMID: 37597393 DOI: 10.1016/j.jhazmat.2023.132247] [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: 05/14/2023] [Revised: 07/27/2023] [Accepted: 08/06/2023] [Indexed: 08/21/2023]
Abstract
Tropane alkaloids (TAs) have emerged as plant toxins, related to poisoning events. The development of stable antibodies is crucial to ensure the effectiveness of immunological methods in quickly and accurately monitoring these alkaloids. In this study, based on hybridoma, the variable region gene of monoclonal antibody (mAb) was amplified, and the recombinant antibody (rAb) gene sequence (VH-Linker-VL) was successfully constructed and expressed in HEK293F. The obtained rAb has kept the same performance as mAb, and the IC50 of 29 TAs ranged from 0.12 to 2642.78 ng/mL. In the recognition mechanism, the docking and dynamics model identified hydrophobic interaction as the most critical force. Substituent will impact recognition by influencing the spatial structure and hydrophobic properties. Then, a colorimetric immunoassay based on rAb was established, five types of water and thirty-nine nectars of honey were tested. The results demonstrated the absence of TAs in environmental water, whereas atropine was detected in more than 13.47% of honey samples at concentrations exceeding 1 μg/kg. The results show a good correlation with UHPLC-MS/MS, suggesting that the immunoassay has excellent screening ability. The data on TAs in honey and water could serve as a foundation for developing relevant policies.
Collapse
Affiliation(s)
- Zile Wang
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Qiang Ma
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Pimiao Zheng
- College of Veterinary Medicine, Shandong Agricultural University, Taian 271018, People's Republic of China
| | - Sanlei Xie
- College of Veterinary Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Kai Yao
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, People's Republic of China
| | - Jing Zhang
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, People's Republic of China
| | - Bing Shao
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, People's Republic of China
| | - Haiyang Jiang
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China.
| |
Collapse
|
9
|
Yasir M, Park J, Han ET, Park WS, Han JH, Kwon YS, Lee HJ, Chun W. Vismodegib Identified as a Novel COX-2 Inhibitor via Deep-Learning-Based Drug Repositioning and Molecular Docking Analysis. ACS OMEGA 2023; 8:34160-34170. [PMID: 37744812 PMCID: PMC10515398 DOI: 10.1021/acsomega.3c05425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023]
Abstract
Artificial intelligence algorithms have been increasingly applied in drug development due to their efficiency and effectiveness. Deep-learning-based drug repurposing can contribute to the identification of novel therapeutic applications for drugs with other indications. The current study used a trained deep-learning model to screen an FDA-approved drug library for novel COX-2 inhibitors. Reference COX-2 data sets, composed of active and decoy compounds, were obtained from the DUD-E database. To extract molecular features, compounds were subjected to RDKit, a cheminformatic toolkit. GraphConvMol, a graph convolutional network model from DeepChem, was applied to obtain a predictive model from the DUD-E data sets. Then, the COX-2 inhibitory potential of the FDA-approved drugs was predicted using the trained deep-learning model. Vismodegib, an anticancer agent that inhibits the hedgehog signaling pathway by binding to smoothened, was predicted to inhibit COX-2. Noticeably, some compounds that exhibit high potential from the prediction were known to be COX-2 inhibitors, indicating the prediction model's liability. To confirm the COX-2 inhibition activity of vismodegib, molecular docking was carried out with the reference compounds of the COX-2 inhibitor, celecoxib, and ibuprofen. Furthermore, the experimental examination of COX-2 inhibition was also carried out using a cell culture study. Results showed that vismodegib exhibited a highly comparable COX-2 inhibitory activity compared to celecoxib and ibuprofen. In conclusion, the deep-learning model can efficiently improve the virtual screening of drugs, and vismodegib can be used as a novel COX-2 inhibitor.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department
of Pharmacology, Kangwon National University
School of Medicine, Chuncheon24341, Republic
of Korea
| | - Jinyoung Park
- Department
of Pharmacology, Kangwon National University
School of Medicine, Chuncheon24341, Republic
of Korea
| | - Eun-Taek Han
- Department
of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Won Sun Park
- Department
of Physiology, Kangwon National University
School of Medicine, Chuncheon24341, Republic
of Korea
| | - Jin-Hee Han
- Department
of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Yong-Soo Kwon
- College
of Pharmacy, Kangwon National University
School of Medicine, Chuncheon24341, Republic
of Korea
| | - Hee-Jae Lee
- Department
of Pharmacology, Kangwon National University
School of Medicine, Chuncheon24341, Republic
of Korea
| | - Wanjoo Chun
- Department
of Pharmacology, Kangwon National University
School of Medicine, Chuncheon24341, Republic
of Korea
| |
Collapse
|
10
|
Wang Y, Ye R, Fan L, Zhao X, Li L, Zheng H, Qiu Y, He X, Lu Y. A TNF-α blocking peptide that reduces NF-κB and MAPK activity for attenuating inflammation. Bioorg Med Chem 2023; 92:117420. [PMID: 37573821 DOI: 10.1016/j.bmc.2023.117420] [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: 04/28/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023]
Abstract
Overexpression of tumor necrosis factor-α (TNF-α) is implicated in many inflammatory diseases, including septic shock, hepatitis, asthma, insulin resistance and autoimmune diseases, such as rheumatoid arthritis and Crohn's disease. The TNF-α signaling pathway is a valuable target, and anti-TNF-α drugs are successfully used to treat autoimmune and inflammatory diseases. Here, we study anti-inflammatory activity of an anti-TNF-α peptide (SN1-13, DEFHLELHLYQSW). In the cellular level assessment, SN1-13 inhibited TNF-α-induced cytotoxicity and blocks TNF-α-triggered signaling activities (IC50 = 15.40 μM). Moreover, the potential binding model between SN1-13 and TNF-α/TNFRs conducted through molecular docking revealed that SN1-13 could stunt TNF-α mediated signaling thought blocking TNF-α and its receptor TNFR1 and TNFR2. These results suggest that SN1-13 would be a potential lead peptide to treat TNF-α-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yue Wang
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun 130013, China
| | - Ruiwei Ye
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Liming Fan
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai 201299, China
| | - Xin Zhao
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072,China
| | - Linxue Li
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Hao Zheng
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yan Qiu
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai 201299, China.
| | - Xiuxia He
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun 130013, China.
| | - Yiming Lu
- School of Medicine, Shanghai University, Shanghai 200444, China; Department of Critical Care Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072,China.
| |
Collapse
|
11
|
Buckner J, Liu X, Chakravorty A, Wu Y, Cervantes LF, Lai TT, Brooks CL. pyCHARMM: Embedding CHARMM Functionality in a Python Framework. J Chem Theory Comput 2023; 19:3752-3762. [PMID: 37267404 PMCID: PMC10504603 DOI: 10.1021/acs.jctc.3c00364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CHARMM is rich in methodology and functionality as one of the first programs addressing problems of molecular dynamics and modeling of biological macromolecules and their partners, e.g., small molecule ligands. When combined with the highly developed CHARMM parameters for proteins, nucleic acids, small molecules, lipids, sugars, and other biologically relevant building blocks, and the versatile CHARMM scripting language, CHARMM has been a trendsetting platform for modeling studies of biological macromolecules. To further enhance the utility of accessing and using CHARMM functionality in increasingly complex workflows associated with modeling biological systems, we introduce pyCHARMM, Python bindings, functions, and modules to complement and extend the extensive set of modeling tools and methods already available in CHARMM. These include access to CHARMM function-generated variables associated with the system (psf), coordinates, velocities and forces, atom selection variables, and force field related parameters. The ability to augment CHARMM forces and energies with energy terms or methods derived from machine learning or other sources, written in Python, CUDA, or OpenCL and expressed as Python callable routines is introduced together with analogous functions callable during dynamics calculations. Integration of Python-based graphical engines for visualization of simulation models and results is also accessible. Loosely coupled parallelism is available for workflows such as free energy calculations, using MBAR/TI approaches or high-throughput multisite λ-dynamics (MSλD) free energy methods, string path optimization calculations, replica exchange, and molecular docking with a new Python-based CDOCKER module. CHARMM accelerated platform kernels through the CHARMM/OpenMM API, CHARMM/DOMDEC, and CHARMM/BLaDE API are also readily integrated into this Python framework. We anticipate that pyCHARMM will be a robust platform for the development of comprehensive and complex workflows utilizing Python and its extensive functionality as well as an optimal platform for users to learn molecular modeling methods and practices within a Python-friendly environment such as Jupyter Notebooks.
Collapse
Affiliation(s)
- Joshua Buckner
- Department of Chemistry, University of Michigan, Ann Arbor, MI
| | - Xiaorong Liu
- Department of Chemistry, University of Michigan, Ann Arbor, MI
| | | | - Yujin Wu
- Department of Chemistry, University of Michigan, Ann Arbor, MI
| | - Luis F. Cervantes
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI
| | - Thanh T. Lai
- Biophysics Program, University of Michigan, Ann Arbor, MI
| | - Charles L. Brooks
- Department of Chemistry, University of Michigan, Ann Arbor, MI
- Biophysics Program, University of Michigan, Ann Arbor, MI
| |
Collapse
|
12
|
Yang R, Liu L, Jiang D, Liu L, Yang H, Xu H, Qin M, Wang P, Gu J, Xing Y. Identification of Potential TMPRSS2 Inhibitors for COVID-19 Treatment in Chinese Medicine by Computational Approaches and Surface Plasmon Resonance Technology. J Chem Inf Model 2023; 63:3005-3017. [PMID: 37155923 DOI: 10.1021/acs.jcim.2c01643] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
BACKGROUND Coronavirus disease-19 (COVID-19) pneumonia continues to spread in the entire globe with limited medication available. In this study, the active compounds in Chinese medicine (CM) recipes targeting the transmembrane serine protease 2 (TMPRSS2) protein for the treatment of COVID-19 were explored. METHODS The conformational structure of TMPRSS2 protein (TMPS2) was built through homology modeling. A training set covering TMPS2 inhibitors and decoy molecules was docked to TMPS2, and their docking poses were re-scored with scoring schemes. A receiver operating characteristic (ROC) curve was applied to select the best scoring function. Virtual screening of the candidate compounds (CCDs) in the six highly effective CM recipes against TMPS2 was conducted based on the validated docking protocol. The potential CCDs after docking were subject to molecular dynamics (MD) simulations and surface plasmon resonance (SPR) experiment. RESULTS A training set of 65 molecules were docked with modeled TMPS2 and LigScore2 with the highest area under the curve, AUC, value (0.886) after ROC analysis selected to best differentiate inhibitors from decoys. A total of 421 CCDs in the six recipes were successfully docked into TMPS2, and the top 16 CCDs with LigScore2 higher than the cutoff (4.995) were screened out. MD simulations revealed a stable binding between these CCDs and TMPS2 due to the negative binding free energy. Lastly, SPR experiments validated the direct combination of narirutin, saikosaponin B1, and rutin with TMPS2. CONCLUSIONS Specific active compounds including narirutin, saikosaponin B1, and rutin in CM recipes potentially target and inhibit TMPS2, probably exerting a therapeutic effect on COVID-19.
Collapse
Affiliation(s)
- Rong Yang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Linhua Liu
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Dansheng Jiang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Lei Liu
- Department of Infectious Diseases, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Huili Yang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Hongling Xu
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Meirong Qin
- National Medical Products Administration, Shenzhen Institute for Drug Control, Shenzhen 518057, China
| | - Ping Wang
- National Medical Products Administration, Shenzhen Institute for Drug Control, Shenzhen 518057, China
| | - Jiangyong Gu
- Research Centre for Integrative Medicine, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yufeng Xing
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
13
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [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
|
14
|
Liao Y, Cao P, Luo L. Identification of Novel Arachidonic Acid 15-Lipoxygenase Inhibitors Based on the Bayesian Classifier Model and Computer-Aided High-Throughput Virtual Screening. Pharmaceuticals (Basel) 2022; 15:1440. [PMID: 36422570 PMCID: PMC9695033 DOI: 10.3390/ph15111440] [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: 10/08/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 08/29/2023] Open
Abstract
Ferroptosis is an iron-dependent lipid peroxidative form of cell death that is distinct from apoptosis and necrosis. ALOX15, also known as arachidonic acid 15-lipoxygenase, promotes ferroptosis by converting intracellular unsaturated lipids into oxidized lipid intermediates and is an important ferroptosis target. In this study, a naive Bayesian machine learning classifier with a structure-based, high-throughput screening approach and a molecular docking program were combined to screen for three compounds with excellent target-binding potential. In the absorption, distribution, metabolism, excretion, and toxicity characterization, three candidate molecules were predicted to exhibit drug-like properties. The subsequent molecular dynamics simulations confirmed their stable binding to the targets. The findings indicated that the compounds exhibited excellent potential ALOX15 inhibitor capacity, thereby providing novel candidates for the treatment of inflammatory ischemia-related diseases caused by ferroptosis.
Collapse
Affiliation(s)
- Yinglin Liao
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China
| | - Peng Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
| |
Collapse
|
15
|
Combining virtual screening and in vitro evaluation for the discovery of potential CYP11B2 inhibitors. Future Med Chem 2022; 14:1239-1250. [PMID: 35912798 DOI: 10.4155/fmc-2022-0119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: To search for highly bioactive hits for CYP11B2 inhibitors by virtual screening and in vitro evaluation. Materials & methods: Virtual screening of potential CYP11B2 inhibitors was performed by molecular docking and molecular dynamics simulation. Compound activity was determined by in vitro evaluation using MTT and ELISA assays. Results & conclusion: Based on the results of molecular docking and molecular dynamics simulation, nine lead hits were selected for in vitro biochemical testing. All hits in in vitro experiments had lower inhibitory effects on cell proliferation and certain inhibitory effects on aldosterone secretion. These hits may be excellent candidates for CYP11B2 inhibitors.
Collapse
|