1
|
Wu MH, Xie Z, Zhi D. A Folding-Docking-Affinity framework for protein-ligand binding affinity prediction. Commun Chem 2025; 8:108. [PMID: 40195508 PMCID: PMC11977223 DOI: 10.1038/s42004-025-01506-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 03/25/2025] [Indexed: 04/09/2025] Open
Abstract
Accurate protein-ligand binding affinity prediction is crucial in drug discovery. Existing methods are predominately docking-free, without explicitly considering atom-level interaction between proteins and ligands in scenarios where crystallized protein-ligand binding conformations are unavailable. Now, with breakthroughs in deep learning AI-based protein folding and binding conformation prediction, can we improve binding affinity prediction? This study introduces a framework, Folding-Docking-Affinity (FDA), which folds proteins, determines protein-ligand binding conformations, and predicts binding affinities from three-dimensional protein-ligand binding structures. Our experimental results indicate that FDA performs comparably to state-of-the-art docking-free methods. We anticipate that our proposed framework serves as a starting point for integrating binding structures for more accurate binding affinity prediction.
Collapse
Affiliation(s)
- Ming-Hsiu Wu
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Ziqian Xie
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Degui Zhi
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
2
|
Stelitano G, Bettoni C, Marczyk J, Chiarelli LR. Artificial Intuition and accelerating the process of antimicrobial drug discovery. Comput Biol Med 2025; 188:109833. [PMID: 39954396 DOI: 10.1016/j.compbiomed.2025.109833] [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: 10/24/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
New drug development is a very challenging, expensive, and usually time-consuming process. This issue is very important with regard to antimicrobials, which are affected by the global issue of the development and spread of resistance. This framework underscores the urgency of accelerating drug development processes while reducing their costs. In this context, new bioinformatics tools can provide important support for drug development by limiting and shortening in vitro evaluation of the best outcomes, thereby minimizing costs. Recently, new Artificial Intelligence (AI)-based tools have been developed for de novo design of new molecules, or for the identification of features of inhibitors among a large set of molecules that can guide rational design. With this work, we present an Artificial Intuition (AI4)-based pharmacological analysis of a series of antimicrobial compounds that are known to be active against Mycobacterium tuberculosis. The compounds have been subjected to Molecular Dynamic Simulation (MDS), and the respective outputs processed with a Quantitative Complexity Management (QCM) tool in order to determine the corresponding complexity profiles. The comparison of different analogues in each series revealed a relationship between the complexity of the various chemical moieties and their importance for the biological activity of each compound, suggesting that QCM may be a useful tool in guiding the optimization process. This first attempt to apply the tool in the field of drug development has yielded interesting results, indicating that QCM, which powers AI4, can be implemented for rational drug design in the near future.
Collapse
Affiliation(s)
- Giovanni Stelitano
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Christian Bettoni
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | | | - Laurent R Chiarelli
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, 27100, Pavia, Italy.
| |
Collapse
|
3
|
Mahdizadeh S, Eriksson LA. iScore: A ML-Based Scoring Function for De Novo Drug Discovery. J Chem Inf Model 2025; 65:2759-2772. [PMID: 40036330 PMCID: PMC11938276 DOI: 10.1021/acs.jcim.4c02192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
In the quest for accelerating de novo drug discovery, the development of efficient and accurate scoring functions represents a fundamental challenge. This study introduces iScore, a novel machine learning (ML)-based scoring function designed to predict the binding affinity of protein-ligand complexes with remarkable speed and precision. Uniquely, iScore circumvents the conventional reliance on explicit knowledge of protein-ligand interactions and a full picture of atomic contacts, instead leveraging a set of ligand and binding pocket descriptors to directly evaluate binding affinity. This approach enables skipping the inefficient and slow conformational sampling stage, thereby enabling the rapid screening of ultrahuge molecular libraries, a crucial advancement given the practically infinite dimensions of chemical space. iScore was rigorously trained and validated using the PDBbind 2020 refined set, CASF 2016, CSAR NRC-HiQ Set1/2, DUD-E, and target fishing data sets, employing three distinct ML methodologies: Deep neural network (iScore-DNN), random forest (iScore-RF), and eXtreme gradient boosting (iScore-XGB). A hybrid model, iScore-Hybrid, was subsequently developed to incorporate the strengths of these individual base learners. The hybrid model demonstrated a Pearson correlation coefficient (R) of 0.78 and a root-mean-square error (RMSE) of 1.23 in cross-validation, outperforming the individual base learners and establishing new benchmarks for scoring power (R = 0.814, RMSE = 1.34), ranking power (ρ = 0.705), and screening power (success rate at top 10% = 73.7%). Moreover, iScore-Hybrid demonstrated great performance in the target fishing benchmarking study.
Collapse
Affiliation(s)
- Sayyed
Jalil Mahdizadeh
- Department of Chemistry and
Molecular Biology, University of Gothenburg, Göteborg 405 30, Sweden
| | - Leif A. Eriksson
- Department of Chemistry and
Molecular Biology, University of Gothenburg, Göteborg 405 30, Sweden
| |
Collapse
|
4
|
Wang ZC, Zeng Y, Sun JY, Chen XQ, Wu HC, Li YY, Mu YG, Zheng LZ, Gao ZB, Li WF. An efficient deep learning-based strategy to screen inhibitors for GluN1/GluN3A receptor. Acta Pharmacol Sin 2025:10.1038/s41401-025-01513-x. [PMID: 40069493 DOI: 10.1038/s41401-025-01513-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/12/2025] [Indexed: 03/15/2025]
Abstract
The GluN1/GluN3A receptor, a unique excitatory glycine receptor recently identified in the central nervous system, challenges traditional perspectives of N-methyl-D-aspartate (NMDA) receptor diversity and glycinergic signaling. Its role in emotional regulation positions it as a potential therapeutic target for neuropsychiatric disorders. However, pharmacological research on GluN1/GluN3A receptors remains at an early stage. Traditional high-throughput screening methods for ion channel drug discovery often lack efficiency, particularly when applied to large compound libraries. To address this concern, we designed a deep learning-based strategy that balances efficiency and accuracy for identifying GluN1/GluN3A inhibitors. First, a sequence-based scoring function was developed to rapidly screen a library containing 18 million compounds, reducing the pool to approximately 105 candidates. Next, two complex-based scoring functions, IGModel and RTMScore, were employed to precisely score and rank the remaining candidates. Finally, an active molecule with an IC50 of 2.87 ± 0.80 μM for the GluN1/GluN3A receptor was confirmed through whole-cell voltage-clamp electrophysiology. This study also presents a paradigm for integrating deep learning into rapid and precise high-throughput screening.
Collapse
Affiliation(s)
- Ze-Chen Wang
- School of Physics, Shandong University, Jinan, 250100, China
| | - Yue Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 200032, China
| | - Jin-Yuan Sun
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue-Qin Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hao-Chen Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yang-Yang Li
- School of Physics, Shandong University, Jinan, 250100, China
| | - Yu-Guang Mu
- School of Biological Science, Nanyang Technological University, Singapore, 637551, Singapore
| | - Liang-Zhen Zheng
- Shenzhen Zelixir Biotech Co. Ltd, Hengtaiyu Park, Shenzhen, 518107, China
| | - Zhao-Bing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Wei-Feng Li
- School of Physics, Shandong University, Jinan, 250100, China.
| |
Collapse
|
5
|
Xie A, Zhao G, Liang H, Gao T, Gao X, Hou N, Wei F, Li J, Zhao H, Xu X. LeScore: a scoring function incorporating hydrogen bonding penalty for protein-ligand docking. J Mol Model 2025; 31:106. [PMID: 40029439 DOI: 10.1007/s00894-025-06328-5] [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: 10/28/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
CONTEXT Molecular docking is vital for structure-based virtual screening and heavily depends on accurate and robust scoring functions. Scoring functions often inadequately account for the breakage of solvent hydrogen bonds, hindering the accuracy of predicting binding energy. Here, we introduce LeScore, a novel scoring function that specifically incorporates the hydrogen bonding penalty (HBP) in an aqueous environment, aiming to penalize unfavorable polar interactions when hydrogen bonds with water are broken but the energy loss is not fully compensated by newly formed protein-ligand interactions. LeScore was optimized for descriptor combinations and subsequently validated using a testing data set, achieving a Pearson correlation coefficient (rp) of 0.53 in the training set and 0.52 in the testing set. To evaluate its screening capability, a subset of the Directory of Useful Decoys: Enhanced (DUD-E) was used. And LeScore achieved an AUC of 0.71 for specific targets, outperforming models without HBP and enhancing the ranking and classification of active compounds. Overall, LeScore provides a robust tool for improving virtual screening, especially in cases where hydrogen bonding is crucial for ligand binding. METHOD LeScore is formulated as a linear combination of descriptors, including van der Waals interactions, hydrogen bond energy, ligand strain energy, and newly integrated HBP. The function was optimized using multiple linear regression (MLR) on the PDBbind 2019 dataset. Evaluation metrics, such as Pearson and Spearman correlation coefficients were utilized to assess the performance of 12 descriptor combinations. Additionally, the study employed datasets from the DUD-E to evaluate LeScore's ability to distinguish active ligands from decoys across multiple target proteins.
Collapse
Affiliation(s)
- Aowei Xie
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China
| | - Guangjian Zhao
- School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Marine Biomedical Research Institute of Qingdao, Chinese Ministry of Education, Ocean University of China, Qingda, 266003, Shandong, People's Republic of China
| | - Huicong Liang
- School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Marine Biomedical Research Institute of Qingdao, Chinese Ministry of Education, Ocean University of China, Qingda, 266003, Shandong, People's Republic of China
| | - Ting Gao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China
| | - Xinru Gao
- School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Marine Biomedical Research Institute of Qingdao, Chinese Ministry of Education, Ocean University of China, Qingda, 266003, Shandong, People's Republic of China
| | - Ning Hou
- School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Marine Biomedical Research Institute of Qingdao, Chinese Ministry of Education, Ocean University of China, Qingda, 266003, Shandong, People's Republic of China
| | - Fengjiao Wei
- School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Marine Biomedical Research Institute of Qingdao, Chinese Ministry of Education, Ocean University of China, Qingda, 266003, Shandong, People's Republic of China
| | - Jiajie Li
- School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Marine Biomedical Research Institute of Qingdao, Chinese Ministry of Education, Ocean University of China, Qingda, 266003, Shandong, People's Republic of China
| | - Hongtao Zhao
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, 43183, Gothenburg, Sweden.
| | - Ximing Xu
- School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Marine Biomedical Research Institute of Qingdao, Chinese Ministry of Education, Ocean University of China, Qingda, 266003, Shandong, People's Republic of China.
| |
Collapse
|
6
|
Nada H, Meanwell NA, Gabr MT. Virtual screening: hope, hype, and the fine line in between. Expert Opin Drug Discov 2025; 20:145-162. [PMID: 39862145 PMCID: PMC11844436 DOI: 10.1080/17460441.2025.2458666] [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: 12/11/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Technological advancements in virtual screening (VS) have rapidly accelerated its application in drug discovery, as reflected by the exponential growth in VS-related publications. However, a significant gap remains between the volume of computational predictions and their experimental validation. This discrepancy has led to a rise in the number of unverified 'claimed' hits which impedes the drug discovery efforts. AREAS COVERED This perspective examines the current VS landscape, highlighting essential practices and identifying critical challenges, limitations, and common pitfalls. Using case studies and practices, this perspective aims to highlight strategies that can effectively mitigate or overcome these challenges. Furthermore, the perspective explores common approaches for addressing pharmacodynamic and pharmacokinetic issues in optimizing VS hits. EXPERT OPINION VS has become a tried-and-true technique of drug discovery due to the rapid advances in computational methods and machine learning (ML) over the past two decades. Although each VS workflow varies depending on the chosen approach and methodology, integrated strategies that combine biological and in silico data have consistently yielded higher success rates. Moreover, the widespread adoption of ML has enhanced the integration of VS into the drug discovery pipeline. However, the absence of standardized evaluation criteria hinders the objective assessment of VS studies' success and the identification of optimal adoption methods.
Collapse
Affiliation(s)
- Hossam Nada
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
| | - Nicholas A. Meanwell
- Baruch S. Blumberg Institute, Doylestown, PA, USA; School of Pharmacy, University of Michigan, Ann Arbor, MI, USA
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Moustafa T. Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
7
|
Lalnunfela C, Lalthanpuii PB, Lalremsanga HT, Zothansiama, Lalmuansangi C, Zosangzuali M, Kumar NS, Lalhriatpuii T, Lalchhandama K. Anticancer activity of Ilex khasiana, a rare and endemic species of holly in Northeast India, against murine lymphoma. Heliyon 2025; 11:e41839. [PMID: 39885875 PMCID: PMC11780953 DOI: 10.1016/j.heliyon.2025.e41839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 02/01/2025] Open
Abstract
Ilex khasiana Purkay. is a lesser-known species of holly (family Aquifoliaceae) that is endemic to Northeast India. Designated as critically endangered, the plant is used in the treatments of bacterial infections, cancer, intestinal helminthiasis, tuberculosis, and viral infections. A methanol extract of the leaves was prepared from which 16 different compounds were identified using gas chromatography-mass spectroscopy. An alkylated phenol, 2,6-di-tert-butylphenol, was the predominant compound. Acute toxicity test indicated that the plant extract was non-toxic even at the highest dosage tested, i.e., 2000 mg/kg body weight. The plant extract caused considerable prolongation of survival in mice transplanted with Dalton's lymphoma ascites, extending life by 33 %, with median survival time of 35.5 and average survival time of 22.83 days, and with a treatment to control ratio of 131.37 %. Reduction of body mass, lipid peroxidation, alanine transaminase, aspartate aminotransferase, and creatinine were seen in DLA-transplanted mice after treatment with the plant extract. On the other hand, glutathione level, glutathione S-transferase and superoxide dismutase activity increased. Alkaline comet assay showed that the plant extract effectively induced DNA damage, producing a tail length of 11.89 μm and Olive moment of 2.36 at 250 mg/kg bwt, the most effective dosage. Molecular docking revealed high ligand binding ability of 2,6-di-tert-butylphenol to chemokine receptor CXCR4, DNA topoisomerase 2-alpha, DNA topoisomerase 2-beta, histone deacetylases (HDAC1, HDAC2, HDAC3), Janus kinase 1 and programmed cell death protein 1. The safety and anticancer activity in the present study substantiate the therapeutic importance of I. khasiana as acclaimed in the Mizo traditional medicine. Additionally, the study advocates further pharmacological investigations as well as the conservation and propagation of the endangered plant for future research.
Collapse
Affiliation(s)
- Charles Lalnunfela
- Department of Zoology, Mizoram University, Tanhril, 796004, Mizoram, India
| | - Pawi Bawitlung Lalthanpuii
- DBT-BUILDER National Laboratory, Department of Life Sciences, Pachhunga University College, Aizawl, 796001, Mizoram, India
| | | | - Zothansiama
- Department of Zoology, Mizoram University, Tanhril, 796004, Mizoram, India
| | | | - Mary Zosangzuali
- Department of Zoology, Mizoram University, Tanhril, 796004, Mizoram, India
| | | | - Tochhawng Lalhriatpuii
- Department of Pharmacy, Regional Institute of Paramedical and Nursing Sciences, Zemabawk, 796017, Mizoram, India
| | - Kholhring Lalchhandama
- DBT-BUILDER National Laboratory, Department of Life Sciences, Pachhunga University College, Aizawl, 796001, Mizoram, India
| |
Collapse
|
8
|
Onikanni SA, Fadaka AO, Dao TNP, Munyembaraga V, Nyau V, Sibuyi NRS, Ajayi MG, Nhung NTA, Ejiofor E, Ajiboye BO, Le MH, Chang HH. In silico identification of the anticataract target of βB2-crystallin from Phaseolus vulgaris: a new insight into cataract treatment. Front Chem 2025; 12:1421534. [PMID: 39896137 PMCID: PMC11782562 DOI: 10.3389/fchem.2024.1421534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 12/12/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction Severe protein clumping in the lens can block light and lead to vision issues in cataract patients. Recent studies have linked β-crystallins, which are key proteins in the lens, to the development of cataracts. Specifically, the S175G/H181Q mutation in the βB2-crystallin gene plays a major role in cataract formation. Methods To understand how this mutation can be activated, we utilized computational methods to predict activators from Phaseolus vulgaris. The Schrödinger platform was employed to screen bioactive compounds and simulate molecular interactions in order to analyze binding and structural changes. Results Our results indicated that these phytochemicals are stable near S175G/H181Q. Discussion These findings suggest novel approaches that could potentially be developed into effective anticataract medications through further refinement and additional testing, ultimately resulting in the creation of more potent agents for cataract treatment.
Collapse
Affiliation(s)
- Sunday Amos Onikanni
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Chemical Sciences, Biochemistry Unit, Afe-Babalola University, Ado-Ekiti, Nigeria
| | | | - Tran Nhat Phong Dao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Valens Munyembaraga
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung, Taiwan
- University Teaching Hospital of Butare, Huye, Rwanda
| | - Vincent Nyau
- Department of Food Science and Nutrition, School of Agricultural Sciences, University of Zambia, Lusaka, Zambia
| | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, University of the Western Cape, Bellville, South Africa
| | - Morenike Grace Ajayi
- Department of Chemical Sciences, Bamidele Olumilua University of Education, Science and Technology, Ikere, Nigeria
| | - Nguyen Thi Ai Nhung
- Department of Chemistry, University of Sciences, Hue University, Hue, Vietnam
| | - Emmanuel Ejiofor
- Department of Chemical Sciences, Faculty of Science, Clifford University, Owerrinta, Nigeria
| | - Basiru Olaitan Ajiboye
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye Ekiti, Oye Ekiti, Nigeria
- Institute of Drug Research and Development, SE Bogoro Center, Afe Babalola University, PMB5454, Ado-Ekiti, Nigeria
| | - Minh Hoang Le
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Centre, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
9
|
Xin M, Wang Z, Wang Z, Qu Y, Yang Y, Li YQ, Zhao M, Zheng L, Mu Y, Li W. Evaluations of the Perturbation Resistance of the Deep-Learning-Based Ligand Conformation Optimization Algorithm. J Chem Inf Model 2025; 65:41-49. [PMID: 39724561 DOI: 10.1021/acs.jcim.4c01096] [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: 12/28/2024]
Abstract
In recent years, the deep learning (DL) technique has rapidly developed and shown great success in scoring the protein-ligand binding affinities. The protein-ligand conformation optimization based on DL-derived scoring functions holds broad application prospects, for instance, drug design and enzyme engineering. In this study, we evaluated the robustness of a DL-based ligand conformation optimization protocol (DeepRMSD+Vina) for optimizing structures with input perturbations by examining the predicted ligand binding poses and scoring. Our results clearly indicated that compared to traditional optimization algorithms (such as Prime MM-GBSA and Vina optimization), DeepRMSD+Vina exhibits higher performance when treating diverse protein-ligand cases. The DeepRMSD+Vina is robust and can always generate the correct binding structures even if perturbations (up to 3 Å) are introduced to the input structure. The success rate is 62% for perturbation with a RMSD within 2-3 Å. However, the success rate dramatically drops to 11% for large perturbations, with RMSD extending to 3-4 Å. Furthermore, compared to the widely used optimization protocol of AutoDock Vina, the DL-generated conformation shows a balanced performance for all of the systems under examination. Overall, the DL-based DeepRMSD+Vina is unarguably more reliable than the traditional methods, which is attributed to the physically inspired design of the neural networks in DeepRMSD+Vina where the distance-transformed features describing the atomic interactions between the protein and the ligand have been explicitly considered and modeled. The outstanding robustness of the DL-based ligand conformational optimization algorithm further validates its superiority in the field of conformational optimization.
Collapse
Affiliation(s)
- Minghui Xin
- School of Physics, Shandong University, Jinan 250100, China
| | - Zechen Wang
- School of Physics, Shandong University, Jinan 250100, China
| | - Zhihao Wang
- School of Physics, Shandong University, Jinan 250100, China
| | - Yuanyuan Qu
- School of Physics, Shandong University, Jinan 250100, China
| | - Yanmei Yang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| | - Yong-Qiang Li
- School of Physics, Shandong University, Jinan 250100, China
| | - Mingwen Zhao
- School of Physics, Shandong University, Jinan 250100, China
| | - Liangzhen Zheng
- Shenzhen Zelixir Biotech Co. Ltd, Hengtaiyu Park, Shenzhen 518107, China
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore, Singapore
| | - Weifeng Li
- School of Physics, Shandong University, Jinan 250100, China
| |
Collapse
|
10
|
Cao C, Qiu X, Yang Z, Jin Y. New insights into the evolution and function of the UMAMIT (USUALLY MULTIPLE ACIDS MOVE IN AND OUT TRANSPORTER) gene family. JOURNAL OF PLANT RESEARCH 2025; 138:3-17. [PMID: 39531163 DOI: 10.1007/s10265-024-01596-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
UMAMIT proteins have been known as key players in amino acid transport. In Arabidopsis, functions of several UMAMITs have been characterized, but their precise mechanism, evolutionary history and functional divergence remain elusive. In this study, we conducted phylogenetic analysis of the UMAMIT gene family across key species in the evolutionary history of plants, ranging from algae to angiosperms. Our findings indicate that UMAMIT proteins underwent a substantial expansion from algae to angiosperms, accompanied by the stabilization of the EamA (the main domain of UMAMIT) structure. Phylogenetic studies suggest that UMAMITs may have originated from green algae and be divided into four subfamilies. These proteins first diversified in bryophytes and subsequently experienced gene duplication events in seed plants. Subfamily I was potentially associated with amino acid transport in seeds. Regarding subcellular localization, UMAMITs were predominantly localized in the plasma membrane and chloroplasts. However, members from clade 8 in subfamily III exhibited specific localization in the tonoplast. These members may have multiple functions, such as plant disease resistance and root development. Furthermore, our protein structure prediction revealed that the four-helix bundle motif is crucial in controlling the UMAMIT switch for exporting amino acid. We hypothesize that the specific amino acids in the amino acid binding region determine the type of amino acids being transported. Additionally, subfamily II contains genes that are specifically expressed in reproductive organs and roots in angiosperms, suggesting neofunctionalization. Our study highlights the evolutionary complexity of UMAMITs and underscores their crucial role in the adaptation and diversification of seed plants.
Collapse
Affiliation(s)
- Chenhao Cao
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Xinbao Qiu
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Zhongnan Yang
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Yue Jin
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China.
| |
Collapse
|
11
|
Ramasamy S, Jeyaram K, Narayanan A, Arunachalam S, Ethiraj S, Sankar M, Pandian B. Repurposing fluvoxamine as an inhibitor for NUDT5 in breast cancer cell: an in silico and in vitro study. In Silico Pharmacol 2024; 13:5. [PMID: 39726906 PMCID: PMC11668718 DOI: 10.1007/s40203-024-00293-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Drug repurposing is necessary to accelerate drug discovery and meet the drug needs. This study investigated the possibility of using fluvoxamine to inhibit the cellular metabolizing enzyme NUDT5 in breast cancer. Computational and experimental techniques were used to evaluate the structural flexibility, binding stability, and chemical reactivity of the drugs. These findings indicated that fluvoxamine effectively suppressed the activity of NUDT5, as evidenced by a binding score of - 8.514 kcal/mol. Furthermore, the binding positions of fluvoxamine and NUDT5 were optimized. Fluvoxamine attachment to the active sites of Trp28, Trp46, Glu47, Arg51, Arg84, and Leu98 in NUDT5 has been shown to alter the metabolism of ADPr. These alterations play a role in ATP production in the breast cancer cells. In addition, an MTT assay conducted on the MCF-7 cell line using fluvoxamine revealed an IC50 value of 53.86 ± 0.05 µM. Fluvoxamine-induced apoptosis was confirmed as evidenced by AO/EtBr and DAPI staining. Graphical abstract Effect of fluvoxamine on breast cancer cells.
Collapse
Affiliation(s)
- Sumathi Ramasamy
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126 Tamil Nadu India
| | - Kanimozhi Jeyaram
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126 Tamil Nadu India
| | - Aathimoolam Narayanan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, 626126 Tamil Nadu India
| | | | - Selvarajan Ethiraj
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603203 Tamil Nadu India
| | - Muthumanickam Sankar
- Department of Bioinformatics, Alagappa University, Karaikudi, 630003 Tamil Nadu India
| | - Boomi Pandian
- Department of Bioinformatics, Alagappa University, Karaikudi, 630003 Tamil Nadu India
| |
Collapse
|
12
|
Garg P, Singhal G, Kulkarni P, Horne D, Salgia R, Singhal SS. Artificial Intelligence-Driven Computational Approaches in the Development of Anticancer Drugs. Cancers (Basel) 2024; 16:3884. [PMID: 39594838 PMCID: PMC11593155 DOI: 10.3390/cancers16223884] [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: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The integration of AI has revolutionized cancer drug development, transforming the landscape of drug discovery through sophisticated computational techniques. AI-powered models and algorithms have enhanced computer-aided drug design (CADD), offering unprecedented precision in identifying potential anticancer compounds. Traditionally, cancer drug design has been a complex, resource-intensive process, but AI introduces new opportunities to accelerate discovery, reduce costs, and optimize efficiency. This manuscript delves into the transformative applications of AI-driven methodologies in predicting and developing anticancer drugs, critically evaluating their potential to reshape the future of cancer therapeutics while addressing their challenges and limitations.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Gargi Singhal
- Department of Medical Sciences, S.N. Medical College, Agra 282002, Uttar Pradesh, India
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
13
|
Hong Y, Ha J, Sim J, Lim CJ, Oh KS, Chandrasekaran R, Kim B, Choi J, Ko J, Shin WH, Lee J. Accurate prediction of protein-ligand interactions by combining physical energy functions and graph-neural networks. J Cheminform 2024; 16:121. [PMID: 39497201 PMCID: PMC11536843 DOI: 10.1186/s13321-024-00912-2] [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: 01/22/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024] Open
Abstract
We introduce an advanced model for predicting protein-ligand interactions. Our approach combines the strengths of graph neural networks with physics-based scoring methods. Existing structure-based machine-learning models for protein-ligand binding prediction often fall short in practical virtual screening scenarios, hindered by the intricacies of binding poses, the chemical diversity of drug-like molecules, and the scarcity of crystallographic data for protein-ligand complexes. To overcome the limitations of existing machine learning-based prediction models, we propose a novel approach that fuses three independent neural network models. One classification model is designed to perform binary prediction of a given protein-ligand complex pose. The other two regression models are trained to predict the binding affinity and root-mean-square deviation of a ligand conformation from an input complex structure. We trained the model to account for both deviations in experimental and predicted binding affinities and pose prediction uncertainties. By effectively integrating the outputs of the triplet neural networks with a physics-based scoring function, our model showed a significantly improved performance in hit identification. The benchmark results with three independent decoy sets demonstrate that our model outperformed existing models in forward screening. Our model achieved top 1% enrichment factors of 32.7 and 23.1 with the CASF2016 and DUD-E benchmark sets, respectively. The benchmark results using the LIT-PCBA set further confirmed its higher average enrichment factors, emphasizing the model's efficiency and generalizability. The model's efficiency was further validated by identifying 23 active compounds from 63 candidates in experimental screening for autotaxin inhibitors, demonstrating its practical applicability in hit discovery.Scientific contributionOur work introduces a novel training strategy for a protein-ligand binding affinity prediction model by integrating the outputs of three independent sub-models and utilizing expertly crafted decoy sets. The model showcases exceptional performance across multiple benchmarks. The high enrichment factors in the LIT-PCBA benchmark demonstrate its potential to accelerate hit discovery.
Collapse
Affiliation(s)
- Yiyu Hong
- Arontier Co., 241, Gangnam-daero, Seocho-gu, Seoul, 06735, Republic of Korea
| | - Junsu Ha
- Arontier Co., 241, Gangnam-daero, Seocho-gu, Seoul, 06735, Republic of Korea
| | - Jaemin Sim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chae Jo Lim
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Kwang-Seok Oh
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | | | - Bomin Kim
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jieun Choi
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Junsu Ko
- Arontier Co., 241, Gangnam-daero, Seocho-gu, Seoul, 06735, Republic of Korea.
| | - Woong-Hee Shin
- Arontier Co., 241, Gangnam-daero, Seocho-gu, Seoul, 06735, Republic of Korea.
- Department of Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| | - Juyong Lee
- Arontier Co., 241, Gangnam-daero, Seocho-gu, Seoul, 06735, Republic of Korea.
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea.
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
14
|
Hu Q, Wang Z, Meng J, Li W, Guo J, Mu Y, Wang S, Zheng L, Wei Y. OpenDock: a pytorch-based open-source framework for protein-ligand docking and modelling. Bioinformatics 2024; 40:btae628. [PMID: 39432683 PMCID: PMC11552628 DOI: 10.1093/bioinformatics/btae628] [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: 07/04/2024] [Revised: 09/19/2024] [Accepted: 10/19/2024] [Indexed: 10/23/2024] Open
Abstract
MOTIVATION Molecular docking is an invaluable computational tool with broad applications in computer-aided drug design and enzyme engineering. However, current molecular docking tools are typically implemented in languages such as C++ for calculation speed, which lack flexibility and user-friendliness for further development. Moreover, validating the effectiveness of external scoring functions for molecular docking and screening within these frameworks is challenging, and implementing more efficient sampling strategies is not straightforward. RESULTS To address these limitations, we have developed an open-source molecular docking framework, OpenDock, based on Python and PyTorch. This framework supports the integration of multiple scoring functions; some can be utilized during molecular docking and pose optimization, while others can be used for post-processing scoring. In terms of sampling, the current version of this framework supports simulated annealing and Monte Carlo optimization. Additionally, it can be extended to include methods such as genetic algorithms and particle swarm optimization for sampling docking poses and protein side chain orientations. Distance constraints are also implemented to enable covalent docking, restricted docking or distance map constraints guided pose sampling. Overall, this framework serves as a valuable tool in drug design and enzyme engineering, offering significant flexibility for most protein-ligand modelling tasks. AVAILABILITY AND IMPLEMENTATION OpenDock is publicly available at: https://github.com/guyuehuo/opendock.
Collapse
Affiliation(s)
- Qiuyue Hu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zechen Wang
- School of Physics, Shangdong University, Jinan, 250100, China
| | - Jintao Meng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| | - Weifeng Li
- School of Physics, Shangdong University, Jinan, 250100, China
| | - Jingjing Guo
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR, 999078, China
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Sheng Wang
- Shanghai Zelixir Biotech Co. Ltd, Shanghai, 201203, China
| | | | - Yanjie Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| |
Collapse
|
15
|
Ma Z, Ajibade A, Zou X. Docking strategies for predicting protein-ligand interactions and their application to structure-based drug design. COMMUNICATIONS IN INFORMATION AND SYSTEMS 2024; 24:199-230. [PMID: 39584017 PMCID: PMC11583305 DOI: 10.4310/cis.241021221101] [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] [Indexed: 11/26/2024]
Abstract
Molecular docking stands as a pivotal element in the realm of computer-aided drug design (CADD), consistently contributing to advancements in pharmaceutical research. In essence, it employs computer algorithms to identify the "best" match between two molecules, akin to solving intricate three-dimensional jigsaw puzzles. At a more stringent level, the molecular docking challenge entails predicting the accurate bound association state based on the atomic coordinates of two molecules. This process assumes particular significance in unraveling the mechanistic intricacies of physicochemical interactions at the atomic scale. Notably, the application of docking, especially in the context of protein-small molecule interactions, holds wide-ranging implications for structure-based drug design, given the prevalent use of small compounds as drug candidates. This study provides an overview of docking methodologies, delves into recent key developments, elucidates the physicochemical underpinnings of molecular recognition in protein-ligand interactions, and concludes by addressing the applications of docking in virtual screening, alongside current challenges within existing docking methods.
Collapse
Affiliation(s)
- Zhiwei Ma
- Dalton Cardiovascular Research Center, University of Missouri-Columbia USA
| | - Abeeb Ajibade
- Dalton Cardiovascular Research Center, University of Missouri-Columbia
- Department of Physics and Astronomy, University of Missouri-Columbia USA
| | - Xiaoqin Zou
- Dalton Cardiovascular Research Center, University of Missouri-Columbia
- Department of Physics and Astronomy, University of Missouri-Columbia
- Department of Biochemistry, University of Missouri-Columbia
- Institute for Data Science and Informatics, University of Missouri-Columbia USA
| |
Collapse
|
16
|
Dey J, Mahapatra SR, Raj TK, Misra N, Suar M. Identification of potential flavonoid compounds as antibacterial therapeutics against Klebsiella pneumoniae infection using structure-based virtual screening and molecular dynamics simulation. Mol Divers 2024; 28:3111-3128. [PMID: 37801217 DOI: 10.1007/s11030-023-10738-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023]
Abstract
Klebsiella pneumoniae, which is among the top three pathogens on WHO's priority list, is one of the gram-negative bacteria that doctors and researchers around the world have fought for decades. Capsular polysaccharide (CPS) protein is extensively recognized as an important K. pneumoniae virulence factor. Thus, CPS has become the most characterized target for the discovery of novel drug candidates. The ineffectiveness of currently existing antibiotics urges the search for potent antimicrobial compounds. Flavonoids are a group of plant metabolites that have antibacterial potential and can enhance the present medications to elicit improved results against diverse diseases without adverse reactions. Henceforth, the present study aims to illustrate the inhibitory potential of flavonoids with varying pharmacological properties, targeting the CPS protein of K. pneumoniae by in silico approaches. The flavonoid compounds (n = 169) were retrieved from the PubChem database and screened using the structure-based virtual screening approach. Compounds with the highest binding score were estimated through their pharmacokinetic effects by ADMET descriptors. Finally, four potential inhibitors with PubChem CID: (4301534, 5213, 5481948, and 637080) were selected after molecular docking and drug-likeness analysis. All four lead compounds were employed for the MDS analysis of a 100 ns time period. Various studies were undertaken to assess the stability of the protein-ligand complexes. The binding free energy was computed using MM-PBSA, and the outcomes indicated that the molecules are having stable interactions with the binding site of the target protein. The results revealed that all four compounds can be employed as potential therapeutics against K. pneumoniae.
Collapse
Affiliation(s)
- Jyotirmayee Dey
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India
| | - Soumya Ranjan Mahapatra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India
| | - T Kiran Raj
- Department of Biotechnology & Bioinformatics, School of Life Sciences, JSS Academy of Higher Education & Research, Mysore, India
| | - Namrata Misra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
| | - Mrutyunjay Suar
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
| |
Collapse
|
17
|
Luo Q, Wang S, Li HY, Zheng L, Mu Y, Guo J. Benchmarking reverse docking through AlphaFold2 human proteome. Protein Sci 2024; 33:e5167. [PMID: 39276010 PMCID: PMC11400627 DOI: 10.1002/pro.5167] [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: 05/24/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024]
Abstract
Predicting the binding of ligands to the human proteome via reverse-docking methods enables the understanding of ligand's interactions with potential protein targets in the human body, thereby facilitating drug repositioning and the evaluation of potential off-target effects or toxic side effects of drugs. In this study, we constructed 11 reverse docking pipelines by integrating site prediction tools (PointSite and SiteMap), docking programs (Glide and AutoDock Vina), and scoring functions (Glide, Autodock Vina, RTMScore, DeepRMSD, and OnionNet-SFCT), and then thoroughly benchmarked their predictive capabilities. The results show that the Glide_SFCT (PS) pipeline exhibited the best target prediction performance based on the atomic structure models in AlphaFold2 human proteome. It achieved a success rate of 27.8% when considering the top 100 ranked prediction. This pipeline effectively narrows the range of potential targets within the human proteome, laying a foundation for drug target prediction, off-target assessment, and toxicity prediction, ultimately boosting drug development. By facilitating these critical aspects of drug discovery and development, our work has the potential to ultimately accelerate the identification of new therapeutic agents and improve drug safety.
Collapse
Affiliation(s)
- Qing Luo
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Sheng Wang
- Shanghai Zelixir Biotech Company Ltd., China
| | - Hoi Yeung Li
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Liangzhen Zheng
- Shenzhen Zelixir Biotech Company Ltd., China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Jingjing Guo
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| |
Collapse
|
18
|
Alsedfy MY, Ebnalwaled AA, Moustafa M, Said AH. Investigating the binding affinity, molecular dynamics, and ADMET properties of curcumin-IONPs as a mucoadhesive bioavailable oral treatment for iron deficiency anemia. Sci Rep 2024; 14:22027. [PMID: 39322646 PMCID: PMC11424638 DOI: 10.1038/s41598-024-72577-8] [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: 06/10/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
Iron deficiency anemia (IDA) is a common health issue, and researchers are interested in overcoming it. Nanotechnology green synthesis is one of the recent approaches to making efficient drugs. In this study, we modeled curcumin-coated iron oxide nanoparticles (cur-IONPs) to study their predicted toxicity and drug-likeness properties, then to investigate mucoadhesive behavior by docking cur-IONPs with two main mucin proteins in gastrointestinal tract (GIT) mucosa (muc 5AC and muc 2). Furthermore, the stability of cur-IONPs/protein complexes was assessed by molecular dynamics. Our in-silico studies results showed that cur-IONPs were predicted to be potential candidates to treat IDA due to its mucoadhesive properties, which could enhance the bioavailability, time residency, and iron absorbance through GIT, in addition to its high safety profile with high drug-likeness properties and oral bioavailability. Finally, molecular dynamic simulation studies revealed stable complexes supporting strength docking studies. Our results focus on the high importance of in-silico drug design studies; however, they need to be supported with in vitro and in vivo studies to reveal the efficacy, toxicity, and bioavailability of cur-IONPs.
Collapse
Affiliation(s)
- M Yasser Alsedfy
- Electronics and Nano Devices Lab, Faculty of Science, South Valley University, Qena, 83523, Egypt.
- Department of Radiology, Faculty of Applied Health Sciences, Sphinx University, New Assiut, Egypt.
| | - A A Ebnalwaled
- Electronics and Nano Devices Lab, Faculty of Science, South Valley University, Qena, 83523, Egypt
| | - Mona Moustafa
- Physics Department, Faculty of Science, Minia University, Minya, Egypt
| | - Alaa Hassan Said
- Electronics and Nano Devices Lab, Faculty of Science, South Valley University, Qena, 83523, Egypt
| |
Collapse
|
19
|
Lam HYI, Guan JS, Ong XE, Pincket R, Mu Y. Protein language models are performant in structure-free virtual screening. Brief Bioinform 2024; 25:bbae480. [PMID: 39327890 PMCID: PMC11427677 DOI: 10.1093/bib/bbae480] [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: 05/24/2024] [Revised: 08/17/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
Hitherto virtual screening (VS) has been typically performed using a structure-based drug design paradigm. Such methods typically require the use of molecular docking on high-resolution three-dimensional structures of a target protein-a computationally-intensive and time-consuming exercise. This work demonstrates that by employing protein language models and molecular graphs as inputs to a novel graph-to-transformer cross-attention mechanism, a screening power comparable to state-of-the-art structure-based models can be achieved. The implications thereof include highly expedited VS due to the greatly reduced compute required to run this model, and the ability to perform early stages of computer-aided drug design in the complete absence of 3D protein structures.
Collapse
Affiliation(s)
- Hilbert Yuen In Lam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr, Singapore 637551, Singapore, Republic of Singapore
- MagMol Pte. Ltd., 68 Circular Road, #02-01, Singapore 049422, Singapore, Republic of Singapore
| | - Jia Sheng Guan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr, Singapore 637551, Singapore, Republic of Singapore
| | - Xing Er Ong
- MagMol Pte. Ltd., 68 Circular Road, #02-01, Singapore 049422, Singapore, Republic of Singapore
| | - Robbe Pincket
- Heliovision, Asstraat 5, 3000 Leuven, Leuven, Kingdom of Belgium
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr, Singapore 637551, Singapore, Republic of Singapore
- MagMol Pte. Ltd., 68 Circular Road, #02-01, Singapore 049422, Singapore, Republic of Singapore
| |
Collapse
|
20
|
Wang Z, Zhou F, Wang Z, Hu Q, Li YQ, Wang S, Wei Y, Zheng L, Li W, Peng X. Fully Flexible Molecular Alignment Enables Accurate Ligand Structure Modeling. J Chem Inf Model 2024; 64:6205-6215. [PMID: 39074901 DOI: 10.1021/acs.jcim.4c00669] [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: 07/31/2024]
Abstract
Accurate protein-ligand binding poses are the prerequisites of structure-based binding affinity prediction and provide the structural basis for in-depth lead optimization in small molecule drug design. However, it is challenging to provide reasonable predictions of binding poses for different molecules due to the complexity and diversity of the chemical space of small molecules. Similarity-based molecular alignment techniques can effectively narrow the search range, as structurally similar molecules are likely to have similar binding modes, with higher similarity usually correlated to higher success rates. However, molecular similarity is not consistently high because molecules often require changes to achieve specific purposes, leading to reduced alignment precision. To address this issue, we propose a new alignment method─Z-align. This method uses topological structural information as a criterion for evaluating similarity, reducing the reliance on molecular fingerprint similarity. Our method has achieved success rates significantly higher than those of other methods at moderate levels of similarity. Additionally, our approach can comprehensively and flexibly optimize bond lengths and angles of molecules, maintaining a high accuracy even when dealing with larger molecules. Consequently, our proposed solution helps in achieving more accurate binding poses in protein-ligand docking problems, facilitating the development of small molecule drugs. Z-align is freely available as a web server at https://cloud.zelixir.com/zalign/home.
Collapse
Affiliation(s)
- Zhihao Wang
- School of Physics, Shandong University, Jinan, 250100, China
| | - Fan Zhou
- Shanghai Zelixir Biotech, Shanghai, 200030, China
| | - Zechen Wang
- School of Physics, Shandong University, Jinan, 250100, China
| | - Qiuyue Hu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yong-Qiang Li
- School of Physics, Shandong University, Jinan, 250100, China
| | - Sheng Wang
- Shanghai Zelixir Biotech, Shanghai, 200030, China
| | - Yanjie Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liangzhen Zheng
- Shanghai Zelixir Biotech, Shanghai, 200030, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Weifeng Li
- School of Physics, Shandong University, Jinan, 250100, China
| | - Xiangda Peng
- Shanghai Zelixir Biotech, Shanghai, 200030, China
| |
Collapse
|
21
|
Fan Z, Jia W. High-confidence structural annotation of substances via multi-layer molecular network reveals the system-wide constituent alternations in milk interfered with diphenylolpropane. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134334. [PMID: 38642498 DOI: 10.1016/j.jhazmat.2024.134334] [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: 02/24/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/22/2024]
Abstract
The spectral database-based mass spectrometry (MS) matching strategy is versatile for structural annotating in ingredient fluctuation profiling mediated by external interferences. However, the systematic variability of MS pool attributable to aliasing peaks and inadequacy of present spectral database resulted in a substantial metabolic feature depletion. An amended procedure termed multiple-charges overlap peaks extraction algorithm (MCOP) was proposed involving identifying collision-trigged dissociation precursor ions through iteratively matching mass features of fragmentations to expand the spectral reference library. We showcased the versatility and utility of established strategy in an investigation centered on the stimulation of milk mediated by diphenylolpropane (BPA). MCOP enabled efficient unknown annotations at metabolite-lipid-protein level, which elevated the accuracy of substance annotation to 85.3% after manual validation. Arginase and α-amylase (|r| > 0.75, p < 0.05) were first identified as the crucial issues via graph neural network-based virtual screening in the abnormal metabolism of urea triggered by BPA, resulting in the accumulation of arginine (original: 1.7 μg kg-1 1.7 times) and maltodextrin (original: 6.9 μg kg-1 2.9 times) and thus, exciting the potential dietary risks. Conclusively, MCOP demonstrated generalisation and scalability and substantially advanced the discovery of unknown metabolites for complex matrix samples, thus deciphering dark matter in multi-omics.
Collapse
Affiliation(s)
- Zibian Fan
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China; Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, China.
| |
Collapse
|
22
|
Qu X, Dong L, Luo D, Si Y, Wang B. Water Network-Augmented Two-State Model for Protein-Ligand Binding Affinity Prediction. J Chem Inf Model 2024; 64:2263-2274. [PMID: 37433009 DOI: 10.1021/acs.jcim.3c00567] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Water network rearrangement from the ligand-unbound state to the ligand-bound state is known to have significant effects on the protein-ligand binding interactions, but most of the current machine learning-based scoring functions overlook these effects. In this study, we endeavor to construct a comprehensive and realistic deep learning model by incorporating water network information into both ligand-unbound and -bound states. In particular, extended connectivity interaction features were integrated into graph representation, and graph transformer operator was employed to extract features of the ligand-unbound and -bound states. Through these efforts, we developed a water network-augmented two-state model called ECIFGraph::HM-Holo-Apo. Our new model exhibits satisfactory performance in terms of scoring, ranking, docking, screening, and reverse screening power tests on the CASF-2016 benchmark. In addition, it can achieve superior performance in large-scale docking-based virtual screening tests on the DEKOIS2.0 data set. Our study highlights that the use of a water network-augmented two-state model can be an effective strategy to bolster the robustness and applicability of machine learning-based scoring functions, particularly for targets with hydrophilic or solvent-exposed binding pockets.
Collapse
Affiliation(s)
- Xiaoyang Qu
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Lina Dong
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Ding Luo
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Yubing Si
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Binju Wang
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen 361005, P. R. China
| |
Collapse
|
23
|
Rayka M, Mirzaei M, Mohammad Latifi A. An ensemble-based approach to estimate confidence of predicted protein-ligand binding affinity values. Mol Inform 2024; 43:e202300292. [PMID: 38358080 DOI: 10.1002/minf.202300292] [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: 10/25/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 02/16/2024]
Abstract
When designing a machine learning-based scoring function, we access a limited number of protein-ligand complexes with experimentally determined binding affinity values, representing only a fraction of all possible protein-ligand complexes. Consequently, it is crucial to report a measure of confidence and quantify the uncertainty in the model's predictions during test time. Here, we adopt the conformal prediction technique to evaluate the confidence of a prediction for each member of the core set of the CASF 2016 benchmark. The conformal prediction technique requires a diverse ensemble of predictors for uncertainty estimation. To this end, we introduce ENS-Score as an ensemble predictor, which includes 30 models with different protein-ligand representation approaches and achieves Pearson's correlation of 0.842 on the core set of the CASF 2016 benchmark. Also, we comprehensively investigate the residual error of each data point to assess the normality behavior of the distribution of the residual errors and their correlation to the structural features of the ligands, such as hydrophobic interactions and halogen bonding. In the end, we provide a local host web application to facilitate the usage of ENS-Score. All codes to repeat results are provided at https://github.com/miladrayka/ENS_Score.
Collapse
Affiliation(s)
- Milad Rayka
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Morteza Mirzaei
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Latifi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Wang Z, Wang S, Li Y, Guo J, Wei Y, Mu Y, Zheng L, Li W. A new paradigm for applying deep learning to protein-ligand interaction prediction. Brief Bioinform 2024; 25:bbae145. [PMID: 38581420 PMCID: PMC10998640 DOI: 10.1093/bib/bbae145] [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: 11/06/2023] [Revised: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 04/08/2024] Open
Abstract
Protein-ligand interaction prediction presents a significant challenge in drug design. Numerous machine learning and deep learning (DL) models have been developed to accurately identify docking poses of ligands and active compounds against specific targets. However, current models often suffer from inadequate accuracy or lack practical physical significance in their scoring systems. In this research paper, we introduce IGModel, a novel approach that utilizes the geometric information of protein-ligand complexes as input for predicting the root mean square deviation of docking poses and the binding strength (pKd, the negative value of the logarithm of binding affinity) within the same prediction framework. This ensures that the output scores carry intuitive meaning. We extensively evaluate the performance of IGModel on various docking power test sets, including the CASF-2016 benchmark, PDBbind-CrossDocked-Core and DISCO set, consistently achieving state-of-the-art accuracies. Furthermore, we assess IGModel's generalizability and robustness by evaluating it on unbiased test sets and sets containing target structures generated by AlphaFold2. The exceptional performance of IGModel on these sets demonstrates its efficacy. Additionally, we visualize the latent space of protein-ligand interactions encoded by IGModel and conduct interpretability analysis, providing valuable insights. This study presents a novel framework for DL-based prediction of protein-ligand interactions, contributing to the advancement of this field. The IGModel is available at GitHub repository https://github.com/zchwang/IGModel.
Collapse
Affiliation(s)
- Zechen Wang
- School of Physics, Shandong University, South Shanda Road, 250100 Shandong, China
| | - Sheng Wang
- Shanghai Zelixir Biotech, Xiangke Road, 200030, Shanghai, China
| | - Yangyang Li
- School of Physics, Shandong University, South Shanda Road, 250100 Shandong, China
| | - Jingjing Guo
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Rua de Luís Gonzaga Gomes, Macao, China
| | - Yanjie Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Road 1068, Shenzhen, 518055 Guang Dong, China
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Liangzhen Zheng
- Shanghai Zelixir Biotech, Xiangke Road, 200030, Shanghai, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Road 1068, Shenzhen, 518055 Guang Dong, China
| | - Weifeng Li
- School of Physics, Shandong University, South Shanda Road, 250100 Shandong, China
| |
Collapse
|
25
|
Tan LH, Kwoh CK, Mu Y. RmsdXNA: RMSD prediction of nucleic acid-ligand docking poses using machine-learning method. Brief Bioinform 2024; 25:bbae166. [PMID: 38695120 PMCID: PMC11063749 DOI: 10.1093/bib/bbae166] [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: 12/21/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 05/04/2024] Open
Abstract
Small molecule drugs can be used to target nucleic acids (NA) to regulate biological processes. Computational modeling methods, such as molecular docking or scoring functions, are commonly employed to facilitate drug design. However, the accuracy of the scoring function in predicting the closest-to-native docking pose is often suboptimal. To overcome this problem, a machine learning model, RmsdXNA, was developed to predict the root-mean-square-deviation (RMSD) of ligand docking poses in NA complexes. The versatility of RmsdXNA has been demonstrated by its successful application to various complexes involving different types of NA receptors and ligands, including metal complexes and short peptides. The predicted RMSD by RmsdXNA was strongly correlated with the actual RMSD of the docked poses. RmsdXNA also outperformed the rDock scoring function in ranking and identifying closest-to-native docking poses across different structural groups and on the testing dataset. Using experimental validated results conducted on polyadenylated nuclear element for nuclear expression triplex, RmsdXNA demonstrated better screening power for the RNA-small molecule complex compared to rDock. Molecular dynamics simulations were subsequently employed to validate the binding of top-scoring ligand candidates selected by RmsdXNA and rDock on MALAT1. The results showed that RmsdXNA has a higher success rate in identifying promising ligands that can bind well to the receptor. The development of an accurate docking score for a NA-ligand complex can aid in drug discovery and development advancements. The code to use RmsdXNA is available at the GitHub repository https://github.com/laiheng001/RmsdXNA.
Collapse
Affiliation(s)
- Lai Heng Tan
- Interdisciplinary Graduate School, Nanyang Technological University, 61 Nanyang Drive, 637335 Singapore, Singapore
| | - Chee Keong Kwoh
- School of Computer Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore, Singapore
| |
Collapse
|
26
|
Heidari A, Dehghanian E, Razmara Z, Shahraki S, Samareh Delarami H, Heidari Majd M. Effect of Cu(II) compound containing dipicolinic acid on DNA damage: a study of antiproliferative activity and DNA interaction properties by spectroscopic, molecular docking and molecular dynamics approaches. J Biomol Struct Dyn 2024:1-16. [PMID: 38498382 DOI: 10.1080/07391102.2024.2329308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
A polymeric compound formulized as [Cu(µ-dipic)2{Na2(µ-H2O)4]n.2nH2O (I), where dipic is 2,6-pyridine dicarboxylic acid (dipicolinic acid, H2dipic), was synthesized by sonochemical irradiation. The initial in-vitro cytotoxic activity of this complex compared with renowned anticancer drugs like cisplatin, versus HCT116 colon cell lines, shows promising results. This study investigated the interaction mode between compound (I) and calf-thymus DNA utilizing a range of analytical techniques including spectrophotometry, fluorimetry, partition coefficient analysis, viscometry, gel electrophoresis and molecular docking technique. The results obtained from experimental methods reveal complex (I) could bind to CT-DNA via hydrogen bonding and van der Waals forces and the theoretical methods support it. Also, complex (I) indicates nuclease activity in the attendance of H2O2 and can act as an artificial nuclease to cleave DNA with high efficiency.
Collapse
Affiliation(s)
- Ameneh Heidari
- Department of Chemistry, Faculty of Science, University of Zabol, Zabol, Iran
| | - Effat Dehghanian
- Department of Chemistry, University of Sistan and Baluchestan, Zahedan, Iran
| | - Zohreh Razmara
- Department of Chemistry, Faculty of Science, University of Zabol, Zabol, Iran
| | - Somaye Shahraki
- Department of Chemistry, Faculty of Science, University of Zabol, Zabol, Iran
| | | | | |
Collapse
|
27
|
Opoku F, Govender P, Shonhai A, Simelane MB. Iso-mukaadial acetate and ursolic acid acetate bind to Plasmodium Falciparum heat shock protein 70: towards targeting parasite protein folding pathway. BMC Chem 2024; 18:55. [PMID: 38500145 PMCID: PMC10949600 DOI: 10.1186/s13065-024-01159-6] [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: 01/15/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024] Open
Abstract
Plasmodium falciparum is the most lethal malaria parasite. P. falciparum Hsp70 (PfHsp70) is an essential molecular chaperone (facilitates protein folding) and is deemed a prospective antimalarial drug target. The present study investigates the binding capabilities of select plant derivatives, iso-mukaadial acetate (IMA) and ursolic acid acetate (UAA), against P. falciparum using an in silico docking approach. The interaction between the ligands and PfHsp70 was evaluated using plasmon resonance (SPR) analysis. Molecular docking, binding free energy analysis and molecular dynamics simulations were conducted towards understanding the mechanisms by which the compounds bind to PfHsp70. The molecular docking results revealed ligand flexibilities, conformations and positions of key amino acid residues and protein-ligand interactions as crucial factors accounting for selective inhibition of Hsp70. The simulation results also suggest protein-ligand van der Waals forces as the driving force guiding the interaction of these compounds with PfHsp70. Of the two compounds, UAA and IMA bound to PfHsp70 within the micromolar range based on surface plasmon resonance (SPR) based binding assay. Our findings pave way for future rational design of new selective compounds targeting PfHsp70.
Collapse
Affiliation(s)
- Francis Opoku
- Department of Chemical Sciences (formerly Department of Applied Chemistry), University of Johannesburg, Doornfontein Campus, P.O. Box 17011, Johannesburg, 2028, South Africa
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Penny Govender
- Department of Chemical Sciences (formerly Department of Applied Chemistry), University of Johannesburg, Doornfontein Campus, P.O. Box 17011, Johannesburg, 2028, South Africa
| | - Addmore Shonhai
- Department of Biochemistry & Microbiology, University of Venda, Thohoyandou, South Africa
| | | |
Collapse
|
28
|
Chen D, Liu J, Wei GW. TopoFormer: Multiscale Topology-enabled Structure-to-Sequence Transformer for Protein-Ligand Interaction Predictions. RESEARCH SQUARE 2024:rs.3.rs-3640878. [PMID: 38405777 PMCID: PMC10889053 DOI: 10.21203/rs.3.rs-3640878/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Pre-trained deep Transformers have had tremendous success in a wide variety of disciplines. However, in computational biology, essentially all Transformers are built upon the biological sequences, which ignores vital stereochemical information and may result in crucial errors in downstream predictions. On the other hand, three-dimensional (3D) molecular structures are incompatible with the sequential architecture of Transformer and natural language processing (NLP) models in general. This work addresses this foundational challenge by a topological Transformer (TopoFormer). TopoFormer is built by integrating NLP and a multiscale topology techniques, the persistent topological hyperdigraph Laplacian (PTHL), which systematically converts intricate 3D protein-ligand complexes at various spatial scales into a NLP-admissible sequence of topological invariants and homotopic shapes. Element-specific PTHLs are further developed to embed crucial physical, chemical, and biological interactions into topological sequences. TopoFormer surges ahead of conventional algorithms and recent deep learning variants and gives rise to exemplary scoring accuracy and superior performance in ranking, docking, and screening tasks in a number of benchmark datasets. The proposed topological sequences can be extracted from all kinds of structural data in data science to facilitate various NLP models, heralding a new era in AI-driven discovery.
Collapse
Affiliation(s)
- Dong Chen
- Department of Mathematics, Michigan State University, MI, 48824, USA
| | - Jian Liu
- Department of Mathematics, Michigan State University, MI, 48824, USA
- Mathematical Science Research Center, Chongqing University of Technology, Chongqing 400054, China
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, MI, 48824, USA
- Department of Electrical and Computer Engineering, Michigan State University, MI 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, MI 48824, USA
| |
Collapse
|
29
|
Cai H, Shen C, Jian T, Zhang X, Chen T, Han X, Yang Z, Dang W, Hsieh CY, Kang Y, Pan P, Ji X, Song J, Hou T, Deng Y. CarsiDock: a deep learning paradigm for accurate protein-ligand docking and screening based on large-scale pre-training. Chem Sci 2024; 15:1449-1471. [PMID: 38274053 PMCID: PMC10806797 DOI: 10.1039/d3sc05552c] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The expertise accumulated in deep neural network-based structure prediction has been widely transferred to the field of protein-ligand binding pose prediction, thus leading to the emergence of a variety of deep learning-guided docking models for predicting protein-ligand binding poses without relying on heavy sampling. However, their prediction accuracy and applicability are still far from satisfactory, partially due to the lack of protein-ligand binding complex data. To this end, we create a large-scale complex dataset containing ∼9 M protein-ligand docking complexes for pre-training, and propose CarsiDock, the first deep learning-guided docking approach that leverages pre-training of millions of predicted protein-ligand complexes. CarsiDock contains two main stages, i.e., a deep learning model for the prediction of protein-ligand atomic distance matrices, and a translation, rotation and torsion-guided geometry optimization procedure to reconstruct the matrices into a credible binding pose. The pre-training and multiple innovative architectural designs facilitate the dramatically improved docking accuracy of our approach over the baselines in terms of multiple docking scenarios, thereby contributing to its outstanding early recognition performance in several retrospective virtual screening campaigns. Further explorations demonstrate that CarsiDock can not only guarantee the topological reliability of the binding poses but also successfully reproduce the crucial interactions in crystalized structures, highlighting its superior applicability.
Collapse
Affiliation(s)
- Heng Cai
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Chao Shen
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Tianye Jian
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Xujun Zhang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Tong Chen
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Xiaoqi Han
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Zhuo Yang
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Wei Dang
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Chang-Yu Hsieh
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yu Kang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Peichen Pan
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Xiangyang Ji
- Department of Automation, Tsinghua University Beijing 100084 China
| | - Jianfei Song
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Tingjun Hou
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yafeng Deng
- Hangzhou Carbonsilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| |
Collapse
|
30
|
Rodrigues TCML, Dias AL, dos Santos AMF, Messias Monteiro AF, Oliveira MCN, Oliveira Pires HF, de Sousa NF, Salvadori MGDSS, Scotti MT, Scotti L. Multi-target Phenylpropanoids Against Epilepsy. Curr Neuropharmacol 2024; 22:2168-2190. [PMID: 38847378 PMCID: PMC11337686 DOI: 10.2174/1570159x22666240524160126] [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: 07/31/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 06/13/2024] Open
Abstract
Epilepsy is a neurological disease with no defined cause, characterized by recurrent epileptic seizures. These occur due to the dysregulation of excitatory and inhibitory neurotransmitters in the central nervous system (CNS). Psychopharmaceuticals have undesirable side effects; many patients require more than one pharmacotherapy to control crises. With this in mind, this work emphasizes the discovery of new substances from natural products that can combat epileptic seizures. Using in silico techniques, this review aims to evaluate the antiepileptic and multi-target activity of phenylpropanoid derivatives. Initially, ligand-based virtual screening models (LBVS) were performed with 468 phenylpropanoid compounds to predict biological activities. The LBVS were developed for the targets alpha- amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), voltage-gated calcium channel Ttype (CaV), gamma-aminobutyric acid A (GABAA), gamma-aminobutyric acid transporter type 1 (GAT-1), voltage-gated potassium channel of the Q family (KCNQ), voltage-gated sodium channel (NaV), and N-methyl D-aspartate (NMDA). The compounds that had good results in the LBVS were analyzed for the absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters, and later, the best molecules were evaluated in the molecular docking consensus. The TR430 compound showed the best results in pharmacokinetic parameters; its oral absorption was 99.03%, it did not violate any Lipinski rule, it showed good bioavailability, and no cytotoxicity was observed either from the molecule or from the metabolites in the evaluated parameters. TR430 was able to bind with GABAA (activation) and AMPA (inhibition) targets and demonstrated good binding energy and significant interactions with both targets. The studied compound showed to be a promising molecule with a possible multi-target activity in both fundamental pharmacological targets for the treatment of epilepsy.
Collapse
Affiliation(s)
| | - Arthur Lins Dias
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Aline Matilde Ferreira dos Santos
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Alex France Messias Monteiro
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
| | - Mayara Cecile Nascimento Oliveira
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Hugo Fernandes Oliveira Pires
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, João Pessoa, Paraíba, Brazil
| | - Natália Ferreira de Sousa
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
| | | | - Marcus Tullius Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
| | - Luciana Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, João Pessoa, Paraíba, Brazil
- Teaching and Research Management, University Hospital Lauro Wanderley, Federal University of Paraíba, 58050-585, João Pessoa, PB, Brazil
| |
Collapse
|
31
|
Ariffin NHM, Hasham R, Hamzah MAAM, Park CS. Skin hydration modulatory activities of Ficus deltoidea extract. Fitoterapia 2024; 172:105755. [PMID: 38000761 DOI: 10.1016/j.fitote.2023.105755] [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: 06/27/2023] [Revised: 11/11/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023]
Abstract
Ficus deltoidea was known for its potent antioxidant, anti-melanogenic and photoprotective skin barrier activities. These properties are contributed by its biomarkers which are vitexin and isovitexin. This study aims to optimize the yield of methanolic extraction of Ficus deltoidea leaves (EFD) and evaluate their effects on skin barrier function and hydration. For optimization, Box-Behnken design was utilized to investigate the effects of methanol concentration, sonication time, and solvent-to-sample ratio on the yields of vitexin and isovitexin in EFD. The optimal yields obtained were 32.29 mg/g for vitexin and 35.87 mg/g for isovitexin. The optimum extraction conditions were 77.66% methanol concentration, 20.03 min sonication time, and 19.88 mL/g solvent-to-sample ratio. The quantitative real-time polymerase chain reaction was utilized to measure variant marker genes of transglutaminase-1, caspase 14, ceramide synthase 3, involucrin, and filaggrin of EFD-induced keratinocyte differentiation by in vitro study. Exposure to EFD has elevated the mRNA levels of all tested marker genes by 0.7-9.2 folds. Then, in vivo efficacy study was conducted on 20 female subjects for 14 days to evaluate skin biophysical assessment of hydration. EFD topical formulation treatment successfully increased skin hydration on day 7 (43.74%) and day 14 (47.23%). In silico study by molecular docking was performed to identify intermolecular binding interactions of vitexin and isovitexin with the interested proteins of tested marker genes. The result of molecular docking to the interested proteins revealed a similar trend with real-time PCR data. In conclusion, EFD potentially enhanced the skin barrier function and hydration of human skin cells.
Collapse
Affiliation(s)
- Nor Hazwani Mohd Ariffin
- Department of Bioprocess and Polymer Engineering, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Rosnani Hasham
- Department of Bioprocess and Polymer Engineering, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia.
| | - Mohd Amir Asyraf Mohd Hamzah
- Department of Bioprocess and Polymer Engineering, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Chang Seo Park
- Department of Chemical and Biochemical Engineering, Dongguk University, 3-26, Pil-dong, Chung-gu, Seoul 100-715, Republic of Korea.
| |
Collapse
|
32
|
He B, Guo J, Tong HHY, To WM. Artificial Intelligence in Drug Discovery: A Bibliometric Analysis and Literature Review. Mini Rev Med Chem 2024; 24:1353-1367. [PMID: 38243944 DOI: 10.2174/0113895575271267231123160503] [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: 07/09/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 01/22/2024]
Abstract
Drug discovery is a complex and iterative process, making it ideal for using artificial intelligence (AI). This paper uses a bibliometric approach to reveal AI's trend and underlying structure in drug discovery (AIDD). A total of 4310 journal articles and reviews indexed in Scopus were analyzed, revealing that AIDD has been rapidly growing over the past two decades, with a significant increase after 2017. The United States, China, and the United Kingdom were the leading countries in research output, with academic institutions, particularly the Chinese Academy of Sciences and the University of Cambridge, being the most productive. In addition, industrial companies, including both pharmaceutical and high-tech ones, also made significant contributions. Additionally, this paper thoroughly discussed the evolution and research frontiers of AIDD, which were uncovered through co-occurrence analyses of keywords using VOSviewer. Our findings highlight that AIDD is an interdisciplinary and promising research field that has the potential to revolutionize drug discovery. The comprehensive overview provided here will be of significant interest to researchers, practitioners, and policy-makers in related fields. The results emphasize the need for continued investment and collaboration in AIDD to accelerate drug discovery, reduce costs, and improve patient outcomes.
Collapse
Affiliation(s)
- Baoyu He
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Jingjing Guo
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Henry H Y Tong
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Wai Ming To
- Faculty of Business, Macao Polytechnic University, Macao, China
| |
Collapse
|
33
|
Hamdani S, Allali H, Bouchentouf S. Exploring the Therapeutic Potential of Ginkgo biloba Polyphenols in Targeting Biomarkers of Colorectal Cancer: An In-silico Evaluation. Curr Drug Discov Technol 2024; 21:e020224226651. [PMID: 38318835 DOI: 10.2174/0115701638282497240124102345] [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: 10/12/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a major contributor to cancer-related deaths worldwide, driving the need for effective anticancer therapies with fewer side effects. The exploration of Ginkgo biloba, a natural source, offers a hopeful avenue for novel treatments targeting key colorectal biomarkers involved in CRC treatment. OBJECTIVE The aim of this study was to explore the binding affinity of natural molecules derived from G. biloba to essential biomarkers associated with CRC, including Kirsten rat sarcoma virus, neuroblastoma RAS mutations, serine/threonine-protein kinase B-Raf, phosphatidylinositol 3'-kinase, and deleted colorectal cancer, using molecular docking. The focus of this research was to evaluate how effectively these molecules bind to specified targets in order to identify potential inhibitors for the treatment of CRC. METHODS A total of 152 polyphenolic compounds from G. biloba were selected and subjected to molecular docking simulations to evaluate their interactions with CRC-related biomarkers. The docking results were analysed to identify ligands exhibiting strong affinities towards the targeted genes, suggesting potential inhibitory effects. RESULTS Docking simulations unveiled the strong binding affinities between selected polyphenolic compounds derived from G. biloba and genes associated with CRC. The complex glycoside structures that are found in flavonols are of significant importance. These compounds, including derivatives with distinctive arrangements, exhibited promising docking scores, signifying substantial interactions with the targeted biomarkers. CONCLUSION The study demonstrates the potential of G. biloba-derived molecules as effective anticancer agents for colorectal cancer. The identified ligands exhibit strong interactions with crucial CRC-related biomarkers, suggesting potential inhibition ability. Further in vitro and in vivo investigations are needed to validate and build upon these promising findings, advancing the development of novel and efficient CRC therapies.
Collapse
Affiliation(s)
- Sarra Hamdani
- Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
- Laboratory of Natural and Bioactive Substances (LASNABIO), Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
| | - Hocine Allali
- Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
- Laboratory of Natural and Bioactive Substances (LASNABIO), Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
| | - Salim Bouchentouf
- Laboratory of Natural and Bioactive Substances (LASNABIO), Department of Chemistry, Faculty of Sciences, Abou Bekr Belkaïd University, P.O. Box 119, Tlemcen 13000, Algeria
- Department of Process Engineering, Faculty of Technology, Doctor Tahar Moulay University of Saida, BP 138 cité EN-NASR, Saïda 20000, Algeria
| |
Collapse
|
34
|
Shen T, Liu F, Wang Z, Sun J, Bu Y, Meng J, Chen W, Yao K, Mu Y, Li W, Zhao G, Wang S, Wei Y, Zheng L. zPoseScore model for accurate and robust protein-ligand docking pose scoring in CASP15. Proteins 2023; 91:1837-1849. [PMID: 37606194 DOI: 10.1002/prot.26573] [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] [Received: 04/14/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/23/2023]
Abstract
We introduce a deep learning-based ligand pose scoring model called zPoseScore for predicting protein-ligand complexes in the 15th Critical Assessment of Protein Structure Prediction (CASP15). Our contributions are threefold: first, we generate six training and evaluation data sets by employing advanced data augmentation and sampling methods. Second, we redesign the "zFormer" module, inspired by AlphaFold2's Evoformer, to efficiently describe protein-ligand interactions. This module enables the extraction of protein-ligand paired features that lead to accurate predictions. Finally, we develop the zPoseScore framework with zFormer for scoring and ranking ligand poses, allowing for atomic-level protein-ligand feature encoding and fusion to output refined ligand poses and ligand per-atom deviations. Our results demonstrate excellent performance on various testing data sets, achieving Pearson's correlation R = 0.783 and 0.659 for ranking docking decoys generated based on experimental and predicted protein structures of CASF-2016 protein-ligand complexes. Additionally, we obtain an averaged local distance difference test (lDDT pli = 0.558) of AIchemy LIG2 in CASP15 for de novo protein-ligand complex structure predictions. Detailed analysis shows that accurate ligand binding site prediction and side-chain orientation are crucial for achieving better prediction performance. Our proposed model is one of the most accurate protein-ligand pose prediction models and could serve as a valuable tool in small molecule drug discovery.
Collapse
Affiliation(s)
- Tao Shen
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
| | - Fuxu Liu
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
| | - Zechen Wang
- School of Physics, Shandong University, Jinan, Shandong, China
| | - Jinyuan Sun
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yifan Bu
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
| | - Jintao Meng
- Shenzhen Key Laboratory of Intelligent Bioinformatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Weihua Chen
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
| | - Keyi Yao
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Weifeng Li
- School of Physics, Shandong University, Jinan, Shandong, China
| | - Guoping Zhao
- Shenzhen Key Laboratory of Intelligent Bioinformatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Sheng Wang
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
| | - Yanjie Wei
- Shenzhen Key Laboratory of Intelligent Bioinformatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Liangzhen Zheng
- Shanghai Zelixir Biotech Company Ltd., Shanghai, China
- Shenzhen Key Laboratory of Intelligent Bioinformatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| |
Collapse
|
35
|
Xia S, Chen E, Zhang Y. Integrated Molecular Modeling and Machine Learning for Drug Design. J Chem Theory Comput 2023; 19:7478-7495. [PMID: 37883810 PMCID: PMC10653122 DOI: 10.1021/acs.jctc.3c00814] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
Modern therapeutic development often involves several stages that are interconnected, and multiple iterations are usually required to bring a new drug to the market. Computational approaches have increasingly become an indispensable part of helping reduce the time and cost of the research and development of new drugs. In this Perspective, we summarize our recent efforts on integrating molecular modeling and machine learning to develop computational tools for modulator design, including a pocket-guided rational design approach based on AlphaSpace to target protein-protein interactions, delta machine learning scoring functions for protein-ligand docking as well as virtual screening, and state-of-the-art deep learning models to predict calculated and experimental molecular properties based on molecular mechanics optimized geometries. Meanwhile, we discuss remaining challenges and promising directions for further development and use a retrospective example of FDA approved kinase inhibitor Erlotinib to demonstrate the use of these newly developed computational tools.
Collapse
Affiliation(s)
- Song Xia
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Eric Chen
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Yingkai Zhang
- Department
of Chemistry, New York University, New York, New York 10003, United States
- Simons
Center for Computational Physical Chemistry at New York University, New York, New York 10003, United States
- NYU-ECNU
Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
| |
Collapse
|
36
|
Morales-Salazar I, Garduño-Albino CE, Montes-Enríquez FP, Nava-Tapia DA, Navarro-Tito N, Herrera-Zúñiga LD, González-Zamora E, Islas-Jácome A. Synthesis of Pyrrolo[3,4- b]pyridin-5-ones via Ugi-Zhu Reaction and In Vitro-In Silico Studies against Breast Carcinoma. Pharmaceuticals (Basel) 2023; 16:1562. [PMID: 38004428 PMCID: PMC10674953 DOI: 10.3390/ph16111562] [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: 10/12/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
An Ugi-Zhu three-component reaction (UZ-3CR) coupled in a one-pot manner to a cascade process (N-acylation/aza Diels-Alder cycloaddition/decarboxylation/dehydration) was performed to synthesize a series of pyrrolo[3,4-b]pyridin-5-ones in 20% to 92% overall yields using ytterbium triflate as a catalyst, toluene as a solvent, and microwaves as a heat source. The synthesized molecules were evaluated in vitro against breast cancer cell lines MDA-MB-231 and MCF-7, finding that compound 1f, at a concentration of 6.25 μM, exhibited a potential cytotoxic effect. Then, to understand the interactions between synthesized compounds and the main proteins related to the cancer cell lines, docking studies were performed on the serine/threonine kinase 1 (AKT1) and Orexetine type 2 receptor (Ox2R), finding moderate to strong binding energies, which matched accurately with the in vitro results. Additionally, molecular dynamics were performed between proteins related to the studied cell lines and the three best ligands.
Collapse
Affiliation(s)
- Ivette Morales-Salazar
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Carlos E. Garduño-Albino
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Flora P. Montes-Enríquez
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Dania A. Nava-Tapia
- Laboratorio de Biología Celular del Cáncer, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico;
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico;
| | - Leonardo David Herrera-Zúñiga
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Eduardo González-Zamora
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Alejandro Islas-Jácome
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| |
Collapse
|
37
|
Dong L, Shi S, Qu X, Luo D, Wang B. Ligand binding affinity prediction with fusion of graph neural networks and 3D structure-based complex graph. Phys Chem Chem Phys 2023; 25:24110-24120. [PMID: 37655493 DOI: 10.1039/d3cp03651k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Accurate prediction of protein-ligand binding affinity is pivotal for drug design and discovery. Here, we proposed a novel deep fusion graph neural networks framework named FGNN to learn the protein-ligand interactions from the 3D structures of protein-ligand complexes. Unlike 1D sequences for proteins or 2D graphs for ligands, the 3D graph of protein-ligand complex enables the more accurate representations of the protein-ligand interactions. Benchmark studies have shown that our fusion models FGNN can achieve more accurate prediction of binding affinity than any individual algorithm. The advantages of fusion strategies have been demonstrated in terms of expressive power of data, learning efficiency and model interpretability. Our fusion models show satisfactory performances on diverse data sets, demonstrating their generalization ability. Given the good performances in both binding affinity prediction and virtual screening, our fusion models are expected to be practically applied for drug screening and design. Our work highlights the potential of the fusion graph neural network algorithm in solving complex prediction problems in computational biology and chemistry. The fusion graph neural networks (FGNN) model is freely available in https://github.com/LinaDongXMU/FGNN.
Collapse
Affiliation(s)
- Lina Dong
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, iChEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Shuai Shi
- Department of Algorithm, TuringQ Co., Ltd., Shanghai, 200240, China
| | - Xiaoyang Qu
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, iChEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Ding Luo
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, iChEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Binju Wang
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, iChEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, China
| |
Collapse
|
38
|
Garcia PJB, Huang SKH, De Castro-Cruz KA, Leron RB, Tsai PW. In Silico Neuroprotective Effects of Specific Rheum palmatum Metabolites on Parkinson's Disease Targets. Int J Mol Sci 2023; 24:13929. [PMID: 37762232 PMCID: PMC10530814 DOI: 10.3390/ijms241813929] [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: 07/16/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Parkinson's disease (PD) is one of the large-scale health issues detrimental to human quality of life, and current treatments are only focused on neuroprotection and easing symptoms. This study evaluated in silico binding activity and estimated the stability of major metabolites in the roots of R. palmatum (RP) with main protein targets in Parkinson's disease and their ADMET properties. The major metabolites of RP were subjected to molecular docking and QSAR with α-synuclein, monoamine oxidase isoform B, catechol o-methyltransferase, and A2A adenosine receptor. From this, emodin had the greatest binding activity with Parkinson's disease targets. The chemical stability of the selected compounds was estimated using density functional theory analyses. The docked compounds showed good stability for inhibitory action compared to dopamine and levodopa. According to their structure-activity relationship, aloe-emodin, chrysophanol, emodin, and rhein exhibited good inhibitory activity to specific targets. Finally, mediocre pharmacokinetic properties were observed due to unexceptional blood-brain barrier penetration and safety profile. It was revealed that the major metabolites of RP may have good neuroprotective activity as an additional hit for PD drug development. Also, an association between redox-mediating and activities with PD-relevant protein targets was observed, potentially opening discussion on electrochemical mechanisms with biological functions.
Collapse
Affiliation(s)
- Patrick Jay B. Garcia
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines; (P.J.B.G.); (K.A.D.C.-C.); (R.B.L.)
- School of Graduate Studies, Mapúa University, Manila 1002, Philippines
| | - Steven Kuan-Hua Huang
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan;
- Division of Urology, Department of Surgery, Chi Mei Medical Center, Tainan 711, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kathlia A. De Castro-Cruz
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines; (P.J.B.G.); (K.A.D.C.-C.); (R.B.L.)
| | - Rhoda B. Leron
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines; (P.J.B.G.); (K.A.D.C.-C.); (R.B.L.)
| | - Po-Wei Tsai
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan;
| |
Collapse
|
39
|
Shen C, Zhang X, Hsieh CY, Deng Y, Wang D, Xu L, Wu J, Li D, Kang Y, Hou T, Pan P. A generalized protein-ligand scoring framework with balanced scoring, docking, ranking and screening powers. Chem Sci 2023; 14:8129-8146. [PMID: 37538816 PMCID: PMC10395315 DOI: 10.1039/d3sc02044d] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/03/2023] [Indexed: 08/05/2023] Open
Abstract
Applying machine learning algorithms to protein-ligand scoring functions has aroused widespread attention in recent years due to the high predictive accuracy and affordable computational cost. Nevertheless, most machine learning-based scoring functions are only applicable to a specific task, e.g., binding affinity prediction, binding pose prediction or virtual screening, suggesting that the development of a scoring function with balanced performance in all critical tasks remains a grand challenge. To this end, we propose a novel parameterization strategy by introducing an adjustable binding affinity term that represents the correlation between the predicted outcomes and experimental data into the training of mixture density network. The resulting residue-atom distance likelihood potential not only retains the superior docking and screening power over all the other state-of-the-art approaches, but also achieves a remarkable improvement in scoring and ranking performance. We emphatically explore the impacts of several key elements on prediction accuracy as well as the task preference, and demonstrate that the performance of scoring/ranking and docking/screening tasks of a certain model could be well balanced through an appropriate manner. Overall, our study highlights the potential utility of our innovative parameterization strategy as well as the resulting scoring framework in future structure-based drug design.
Collapse
Affiliation(s)
- Chao Shen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
- State Key Lab of CAD&CG, Zhejiang University Hangzhou 310058 Zhejiang China
- School of Public Health, Zhejiang University Hangzhou 310058 Zhejiang China
- CarbonSilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Xujun Zhang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Chang-Yu Hsieh
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yafeng Deng
- CarbonSilicon AI Technology Co., Ltd Hangzhou 310018 Zhejiang China
| | - Dong Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology Changzhou 213001 China
| | - Jian Wu
- School of Public Health, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Dan Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yu Kang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
- State Key Lab of CAD&CG, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Peichen Pan
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| |
Collapse
|
40
|
Scaini MC, Piccin L, Bassani D, Scapinello A, Pellegrini S, Poggiana C, Catoni C, Tonello D, Pigozzo J, Dall’Olmo L, Rosato A, Moro S, Chiarion-Sileni V, Menin C. Molecular Modeling Unveils the Effective Interaction of B-RAF Inhibitors with Rare B-RAF Insertion Variants. Int J Mol Sci 2023; 24:12285. [PMID: 37569660 PMCID: PMC10418914 DOI: 10.3390/ijms241512285] [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: 07/06/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
The Food and Drug Administration (FDA) has approved MAPK inhibitors as a treatment for melanoma patients carrying a mutation in codon V600 of the BRAF gene exclusively. However, BRAF mutations outside the V600 codon may occur in a small percentage of melanomas. Although these rare variants may cause B-RAF activation, their predictive response to B-RAF inhibitor treatments is still poorly understood. We exploited an integrated approach for mutation detection, tumor evolution tracking, and assessment of response to treatment in a metastatic melanoma patient carrying the rare p.T599dup B-RAF mutation. He was addressed to Dabrafenib/Trametinib targeted therapy, showing an initial dramatic response. In parallel, in-silico ligand-based homology modeling was set up and performed on this and an additional B-RAF rare variant (p.A598_T599insV) to unveil and justify the success of the B-RAF inhibitory activity of Dabrafenib, showing that it could adeptly bind both these variants in a similar manner to how it binds and inhibits the V600E mutant. These findings open up the possibility of broadening the spectrum of BRAF inhibitor-sensitive mutations beyond mutations at codon V600, suggesting that B-RAF V600 WT melanomas should undergo more specific investigations before ruling out the possibility of targeted therapy.
Collapse
Affiliation(s)
- Maria Chiara Scaini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.S.); (S.P.); (C.P.); (C.C.); (D.T.); (A.R.); (C.M.)
| | - Luisa Piccin
- Melanoma Unit, Oncology 2 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (L.P.); (J.P.); (V.C.-S.)
| | - Davide Bassani
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padua, Italy;
| | - Antonio Scapinello
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy;
| | - Stefania Pellegrini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.S.); (S.P.); (C.P.); (C.C.); (D.T.); (A.R.); (C.M.)
| | - Cristina Poggiana
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.S.); (S.P.); (C.P.); (C.C.); (D.T.); (A.R.); (C.M.)
| | - Cristina Catoni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.S.); (S.P.); (C.P.); (C.C.); (D.T.); (A.R.); (C.M.)
| | - Debora Tonello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.S.); (S.P.); (C.P.); (C.C.); (D.T.); (A.R.); (C.M.)
| | - Jacopo Pigozzo
- Melanoma Unit, Oncology 2 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (L.P.); (J.P.); (V.C.-S.)
| | - Luigi Dall’Olmo
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, 35128 Padua, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.S.); (S.P.); (C.P.); (C.C.); (D.T.); (A.R.); (C.M.)
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, 35128 Padua, Italy
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padua, Italy;
| | - Vanna Chiarion-Sileni
- Melanoma Unit, Oncology 2 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (L.P.); (J.P.); (V.C.-S.)
| | - Chiara Menin
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.S.); (S.P.); (C.P.); (C.C.); (D.T.); (A.R.); (C.M.)
| |
Collapse
|
41
|
Abdulredha FH, Mahdi MF, Khan AK. In silico evaluation of binding interaction and ADME study of new 1,3-diazetidin-2-one derivatives with high antiproliferative activity. J Adv Pharm Technol Res 2023; 14:176-184. [PMID: 37692021 PMCID: PMC10483897 DOI: 10.4103/japtr.japtr_116_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/01/2023] [Accepted: 06/07/2023] [Indexed: 09/12/2023] Open
Abstract
A series of eight novels' 1,3-diazetidin-2-ones have been proposed to assess their potential activities. They are intended to examine antiproliferative effects through inhibition of epidermal growth factor receptor (EGFR) expression. These eight compounds strongly interact with the EGFR protein, responsible for the activity. As part of a present study, these compounds were docked to the crystal structure of the EGFR (Protein Data Bank code: 1 M17) to determine their binding affinity at the active site. Based on computer predictions, two compounds were demonstrated high scores of 80.80 and 85.89. After analyzing ADME properties, these compounds were found to have significant potential for binding. Consequently, the abilities of gefitinib, erlotinib, imatinib, and sorafenib were selected for comparison as controls. Computational methods were performed to predict the critical disposition of eight novels' 1,3-diazetidin-2-one derivatives to the EGFR. Moreover, a docking technique employing the Genetic Optimization for Ligand Docking program was conducted. Compounds 2 and 7 demonstrate a high docking peace-wise scoring function (PLP) fitness of 85.89 and 80.80, respectively. They fulfilled the Lipinski's rule, topological descriptors, and fingerprints of drug-like molecular structure keys. These compounds can be used as lead compounds to develop novel antiproliferative agents. The outcome of applying this study is novel series of 1,3-diazetidin-2-one compounds as new analogs were designed and evaluated for their antiproliferative activity with a higher potency profile and binding affinity within the active sites of EGFR.
Collapse
Affiliation(s)
- Farah Haidar Abdulredha
- Department of Pharmaceutical Chemistry, College of Pharmacy, Al-Mustansiriyah University, Baghdad, Iraq
| | - Monther Faisal Mahdi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Al-Mustansiriyah University, Baghdad, Iraq
| | - Ayad Kareem Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Al-Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
42
|
Cui Z, Zhang N, Zhou T, Zhou X, Meng H, Yu Y, Zhang Z, Zhang Y, Wang W, Liu Y. Conserved Sites and Recognition Mechanisms of T1R1 and T2R14 Receptors Revealed by Ensemble Docking and Molecular Descriptors and Fingerprints Combined with Machine Learning. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:5630-5645. [PMID: 37005743 DOI: 10.1021/acs.jafc.3c00591] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Taste peptides, as an important component of protein-rich foodstuffs, potentiate the nutrition and taste of food. Thereinto, umami- and bitter-taste peptides have been ex tensively reported, while their taste mechanisms remain unclear. Meanwhile, the identification of taste peptides is still a time-consuming and costly task. In this study, 489 peptides with umami/bitter taste from TPDB (http://tastepeptides-meta.com/) were collected and used to train the classification models based on docking analysis, molecular descriptors (MDs), and molecular fingerprints (FPs). A consensus model, taste peptide docking machine (TPDM), was generated based on five learning algorithms (linear regression, random forest, gaussian naive bayes, gradient boosting tree, and stochastic gradient descent) and four molecular representation schemes. Model interpretive analysis showed that MDs (VSA_EState, MinEstateIndex, MolLogP) and FPs (598, 322, 952) had the greatest impact on the umami/bitter prediction of peptides. Based on the consensus docking results, we obtained the key recognition modes of umami/bitter receptors (T1Rs/T2Rs): (1) residues 107S-109S, 148S-154T, 247F-249A mainly form hydrogen bonding contacts and (2) residues 153A-158L, 163L, 181Q, 218D, 247F-249A in T1R1 and 56D, 106P, 107V, 152V-156F, 173K-180F in T2R14 constituted their hydrogen bond pockets. The model is available at http://www.tastepeptides-meta.com/yyds.
Collapse
Affiliation(s)
- Zhiyong Cui
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ninglong Zhang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tianxing Zhou
- Department of Bioinformatics, Faculty of Science, The University of Melbourne, Parkville 3010, Victoria, Australia
| | - Xueke Zhou
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hengli Meng
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanyang Yu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiwei Zhang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yin Zhang
- Key Laboratory of Meat Processing of Sichuan, Chengdu University, Chengdu 610106, China
| | - Wenli Wang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuan Liu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
43
|
Yang Z, Zhong W, Lv Q, Dong T, Yu-Chian Chen C. Geometric Interaction Graph Neural Network for Predicting Protein-Ligand Binding Affinities from 3D Structures (GIGN). J Phys Chem Lett 2023; 14:2020-2033. [PMID: 36794930 DOI: 10.1021/acs.jpclett.2c03906] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Predicting protein-ligand binding affinities (PLAs) is a core problem in drug discovery. Recent advances have shown great potential in applying machine learning (ML) for PLA prediction. However, most of them omit the 3D structures of complexes and physical interactions between proteins and ligands, which are considered essential to understanding the binding mechanism. This paper proposes a geometric interaction graph neural network (GIGN) that incorporates 3D structures and physical interactions for predicting protein-ligand binding affinities. Specifically, we design a heterogeneous interaction layer that unifies covalent and noncovalent interactions into the message passing phase to learn node representations more effectively. The heterogeneous interaction layer also follows fundamental biological laws, including invariance to translations and rotations of the complexes, thus avoiding expensive data augmentation strategies. GIGN achieves state-of-the-art performance on three external test sets. Moreover, by visualizing learned representations of protein-ligand complexes, we show that the predictions of GIGN are biologically meaningful.
Collapse
Affiliation(s)
- Ziduo Yang
- Intelligent Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, Guangdong 510275, China
| | - Weihe Zhong
- Intelligent Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, Guangdong 510275, China
| | - Qiujie Lv
- Intelligent Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, Guangdong 510275, China
| | - Tiejun Dong
- Intelligent Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, Guangdong 510275, China
| | - Calvin Yu-Chian Chen
- Intelligent Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, Guangdong 510275, China
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
44
|
Phengsakun G, Boonyarit B, Rungrotmongkol T, Suginta W. Structure-based virtual screening for potent inhibitors of GH-20 β-N-acetylglucosaminidase: classical and machine learning scoring functions, and molecular dynamics simulations. Comput Biol Chem 2023; 104:107856. [PMID: 37003097 DOI: 10.1016/j.compbiolchem.2023.107856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023]
Abstract
GH-20 β-N-acetylglucosaminidases (GlcNAcases) are promising targets in the development of antimicrobial agents against Vibrio infections in humans and aquatic animals. In this study, we set up structure-based virtual screening to identify potential GH-20 GlcNAcase inhibitors from the Reaxys commercial database, using VhGlcNAcase from V. campbellii type strain ATCC® BAA 1116 as the protein target and Redoxal as the reference ligand. Using ChemPLP and RF-Score-VS machine learning scoring functions, eight lead compounds were identified and further evaluated for protein interaction preference and pharmacological properties. Protein-ligand analysis demonstrated that all selected compounds interacted exclusively at subsite - 1 with five hydrophobic residues W487, W505, W546, W582 and V544 at site S1, and with two polar residues, D437 and E438, at site 3. For subsite + 1, the most common residues were R274 and E584 at site 2 and I397 and Q398 at site 4. Based on the data obtained from binding free energy changes (ΔG°binding), pharmacological property analysis and molecular dynamic simulations, two ChemPLP compounds, 338175 and 1146525, and one RF-Score-VS compound, 337447, were considered as the likely lead compounds. The most promising compound, 1146525, could serve as a scaffold for the future design of novel antimicrobial agents against Vibrio infections.
Collapse
|
45
|
Jiang K, Guo H, Zhai J. Interplay of phytohormones and epigenetic regulation: A recipe for plant development and plasticity. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2023; 65:381-398. [PMID: 36223083 DOI: 10.1111/jipb.13384] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Both phytohormone signaling and epigenetic mechanisms have long been known to play crucial roles in plant development and plasticity in response to ambient stimuli. Indeed, diverse signaling pathways mediated by phytohormones and epigenetic processes integrate multiple upstream signals to regulate various plant traits. Emerging evidence indicates that phytohormones and epigenetic processes interact at multiple levels. In this review, we summarize the current knowledge of the interplay between phytohormones and epigenetic processes from the perspective of phytohormone biology. We also review chemical regulators used in epigenetic studies and propose strategies for developing novel regulators using multidisciplinary approaches.
Collapse
Affiliation(s)
- Kai Jiang
- Institute of Plant and Food Science, Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Hongwei Guo
- Institute of Plant and Food Science, Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Jixian Zhai
- Institute of Plant and Food Science, Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| |
Collapse
|
46
|
Wang Z, Zheng L, Wang S, Lin M, Wang Z, Kong AWK, Mu Y, Wei Y, Li W. A fully differentiable ligand pose optimization framework guided by deep learning and a traditional scoring function. Brief Bioinform 2023; 24:6887112. [PMID: 36502369 DOI: 10.1093/bib/bbac520] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 12/14/2022] Open
Abstract
The recently reported machine learning- or deep learning-based scoring functions (SFs) have shown exciting performance in predicting protein-ligand binding affinities with fruitful application prospects. However, the differentiation between highly similar ligand conformations, including the native binding pose (the global energy minimum state), remains challenging that could greatly enhance the docking. In this work, we propose a fully differentiable, end-to-end framework for ligand pose optimization based on a hybrid SF called DeepRMSD+Vina combined with a multi-layer perceptron (DeepRMSD) and the traditional AutoDock Vina SF. The DeepRMSD+Vina, which combines (1) the root mean square deviation (RMSD) of the docking pose with respect to the native pose and (2) the AutoDock Vina score, is fully differentiable; thus is capable of optimizing the ligand binding pose to the energy-lowest conformation. Evaluated by the CASF-2016 docking power dataset, the DeepRMSD+Vina reaches a success rate of 94.4%, which outperforms most reported SFs to date. We evaluated the ligand conformation optimization framework in practical molecular docking scenarios (redocking and cross-docking tasks), revealing the high potentialities of this framework in drug design and discovery. Structural analysis shows that this framework has the ability to identify key physical interactions in protein-ligand binding, such as hydrogen-bonding. Our work provides a paradigm for optimizing ligand conformations based on deep learning algorithms. The DeepRMSD+Vina model and the optimization framework are available at GitHub repository https://github.com/zchwang/DeepRMSD-Vina_Optimization.
Collapse
Affiliation(s)
- Zechen Wang
- School of Physics, Shandong University, Jinan, Shandong 250100, China
| | - Liangzhen Zheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.,Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Sheng Wang
- Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Mingzhi Lin
- Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Zhihao Wang
- School of Physics, Shandong University, Jinan, Shandong 250100, China
| | - Adams Wai-Kin Kong
- Rolls-Royce Corporate Lab, Nanyang Technological University, Singapore 637551, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Yanjie Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Weifeng Li
- School of Physics, Shandong University, Jinan, Shandong 250100, China
| |
Collapse
|
47
|
Wu Q, Huang SY. HCovDock: an efficient docking method for modeling covalent protein-ligand interactions. Brief Bioinform 2023; 24:6961470. [PMID: 36573474 DOI: 10.1093/bib/bbac559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 12/28/2022] Open
Abstract
Covalent inhibitors have received extensive attentions in the past few decades because of their long residence time, high binding efficiency and strong selectivity. Therefore, it is valuable to develop computational tools like molecular docking for modeling of covalent protein-ligand interactions or screening of potential covalent drugs. Meeting the needs, we have proposed HCovDock, an efficient docking algorithm for covalent protein-ligand interactions by integrating a ligand sampling method of incremental construction and a scoring function with covalent bond-based energy. Tested on a benchmark containing 207 diverse protein-ligand complexes, HCovDock exhibits a significantly better performance than seven other state-of-the-art covalent docking programs (AutoDock, Cov_DOX, CovDock, FITTED, GOLD, ICM-Pro and MOE). With the criterion of ligand root-mean-squared distance < 2.0 Å, HCovDock obtains a high success rate of 70.5% and 93.2% in reproducing experimentally observed structures for top 1 and top 10 predictions. In addition, HCovDock is also validated in virtual screening against 10 receptors of three proteins. HCovDock is computationally efficient and the average running time for docking a ligand is only 5 min with as fast as 1 sec for ligands with one rotatable bond and about 18 min for ligands with 23 rotational bonds. HCovDock can be freely assessed at http://huanglab.phys.hust.edu.cn/hcovdock/.
Collapse
Affiliation(s)
- Qilong Wu
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P. R. China
| | - Sheng-You Huang
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P. R. China
| |
Collapse
|
48
|
Li M, Wang Y, Guo C, Wang S, Zheng L, Bu Y, Ding K. The claim of primacy of human gut Bacteroides ovatus in dietary cellobiose degradation. Gut Microbes 2023; 15:2227434. [PMID: 37349961 PMCID: PMC10291918 DOI: 10.1080/19490976.2023.2227434] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 06/14/2023] [Indexed: 06/24/2023] Open
Abstract
A demonstration of cellulose degrading bacterium from human gut changed our view that human cannot degrade the cellulose. However, investigation of cellulose degradation by human gut microbiota on molecular level has not been completed so far. We showed here, using cellobiose as a model that promoted the growth of human gut key members, such as Bacteroides ovatus (BO), to clarify the molecular mechanism. Our results showed that a new polysaccharide utilization locus (PUL) from BO was involved in the cellobiose capturing and degradation. Further, two new cellulases BACOVA_02626GH5 and BACOVA_02630GH5 on the cell surface performed the degradation of cellobiose into glucose were determined. The predicted structures of BACOVA_02626GH5 and BACOVA_02630GH5 were highly homologous with the cellulase from soil bacteria, and the catalytic residues were highly conservative with two glutamate residues. In murine experiment, we observed cellobiose reshaped the composition of gut microbiota and probably modified the metabolic function of bacteria. Taken together, our findings further highlight the evidence of cellulose can be degraded by human gut microbes and provide new insight in the field of investigation on cellulose.
Collapse
Affiliation(s)
- Meixia Li
- Glycochemistry and Glycobiology Lab, Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Yeqing Wang
- Glycochemistry and Glycobiology Lab, Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Ciliang Guo
- Glycochemistry and Glycobiology Lab, Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Science, Beijing, P. R. China
| | | | | | - Yifan Bu
- Zelixir Biotech, Shanghai, P. R. China
| | - Kan Ding
- Glycochemistry and Glycobiology Lab, Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P. R. China
- University of Chinese Academy of Science, Beijing, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan, P. R. China
| |
Collapse
|
49
|
Mirza FJ, Zahid S, Amber S, Sumera, Jabeen H, Asim N, Ali Shah SA. Multitargeted Molecular Docking and Dynamic Simulation Studies of Bioactive Compounds from Rosmarinus officinalis against Alzheimer's Disease. Molecules 2022; 27:7241. [PMID: 36364071 PMCID: PMC9653785 DOI: 10.3390/molecules27217241] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 02/05/2025] Open
Abstract
Alzheimer's disease (AD) has been associated with the hallmark features of cholinergic dysfunction, amyloid beta (Aβ) aggregation and impaired synaptic transmission, which makes the associated proteins, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE I), acetylcholine esterase (AChE) and synapsin I, II and III, major targets for therapeutic intervention. The present study investigated the therapeutic potential of three major phytochemicals of Rosmarinus officinalis, ursolic acid (UA), rosmarinic acid (RA) and carnosic acid (CA), based on their binding affinity with AD-associated proteins. Detailed docking studies were conducted using AutoDock vina followed by molecular dynamic (MD) simulations using Amber 20. The docking analysis of the selected molecules showed the binding energies of their interaction with the target proteins, while MD simulations comprising root mean square deviation (RMSD), root mean square fluctuation (RMSF) and molecular mechanics/generalized born surface area (MM/GBSA) binding free energy calculations were carried out to check the stability of bound complexes. The drug likeness and the pharmacokinetic properties of the selected molecules were also checked through the Lipinski filter and ADMETSAR analysis. All these bioactive compounds demonstrated strong binding affinity with AChE, BACE1 and synapsin I, II and III. The results showed UA and RA to be potential inhibitors of AChE and BACE1, exhibiting binding energies comparable to those of donepezil, used as a positive control. The drug likeness and pharmacokinetic properties of these compounds also demonstrated drug-like characteristics, indicating the need for further in vitro and in vivo investigations to ascertain their therapeutic potential for AD.
Collapse
Affiliation(s)
- Fatima Javed Mirza
- Neurobiology Laboratory, Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| | - Saadia Zahid
- Neurobiology Laboratory, Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| | - Sanila Amber
- Neurobiology Laboratory, Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| | - Sumera
- Neurobiology Laboratory, Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| | - Hira Jabeen
- Center for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TH, UK
| | - Noreen Asim
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar 25130, Pakistan
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam 42300, Malaysia
- Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam 42300, Malaysia
| |
Collapse
|
50
|
Aizezi Y, Xie Y, Guo H, Jiang K. New Wine in an Old Bottle: Utilizing Chemical Genetics to Dissect Apical Hook Development. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081285. [PMID: 36013464 PMCID: PMC9410295 DOI: 10.3390/life12081285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 02/08/2023]
Abstract
The apical hook is formed by dicot seedlings to protect the tender shoot apical meristem during soil emergence. Regulated by many phytohormones, the apical hook has been taken as a model to study the crosstalk between individual signaling pathways. Over recent decades, the roles of different phytohormones and environmental signals in apical hook development have been illustrated. However, key regulators downstream of canonical hormone signaling have rarely been identified via classical genetics screening, possibly due to genetic redundancy and/or lethal mutation. Chemical genetics that utilize small molecules to perturb and elucidate biological processes could provide a complementary strategy to overcome the limitations in classical genetics. In this review, we summarize current progress in hormonal regulation of the apical hook, and previously reported chemical tools that could assist the understanding of this complex developmental process. We also provide insight into novel strategies for chemical screening and target identification, which could possibly lead to discoveries of new regulatory components in apical hook development, or unidentified signaling crosstalk that is overlooked by classical genetics screening.
Collapse
Affiliation(s)
- Yalikunjiang Aizezi
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yinpeng Xie
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hongwei Guo
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Southern University of Science and Technology, Shenzhen 518055, China
- Correspondence: (H.G.); (K.J.)
| | - Kai Jiang
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen 518055, China
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Southern University of Science and Technology, Shenzhen 518055, China
- Correspondence: (H.G.); (K.J.)
| |
Collapse
|