1
|
Albanese V, Marini M, Tesi M, Landini L, Bellantoni E, Cosconati S, Roggia M, Tagliazucchi L, Gnudi L, Puscio V, Sturaro C, Ruzza C, Guerrini R, Geppetti P, Nassini R, Preti D, De Logu F, Pacifico S. Identification of isoxazole-based TRPA1 inhibitors with analgesic effects in vivo. Eur J Med Chem 2025; 294:117732. [PMID: 40378573 DOI: 10.1016/j.ejmech.2025.117732] [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: 02/20/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
The transient receptor potential ankyrin 1 (TRPA1) channel has been extensively studied as a potential therapeutic target for the treatment of different pain types, with better efficacy and safety profiles compared to current therapies. Because TRPA1 is implicated in different pathophysiological processes, selective antagonists of this channel could provide therapeutic benefits beyond pain relief. In this study, we report the design and synthesis of a novel series of carboxamide derivatives incorporating an isoxazole moiety, which were evaluated for their ability to inhibit TRPA1-mediated signalling. Among these, we identified the TRPA1 antagonists 12 and 13 displaying nanomolar potency in vitro and significant analgesic effects against the TRPA1 agonist, allyl isothiocyanate and in the formalin test in mice. Docking analyses were also conducted to explore the binding modes of the most representative compounds with the proposed pharmacological target.
Collapse
Affiliation(s)
- Valentina Albanese
- Department of Environmental and Prevention Sciences, University of Ferrara, Palazzo Turchi di Bagno, C.so Ercole I D'Este 32, 44121, Ferrara, Italy
| | - Matilde Marini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Viale Pieraccini 6, University of Florence, Florence, 50139, Italy
| | - Martina Tesi
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Viale Pieraccini 6, University of Florence, Florence, 50139, Italy
| | - Lorenzo Landini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Viale Pieraccini 6, University of Florence, Florence, 50139, Italy
| | - Elisa Bellantoni
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Viale Pieraccini 6, University of Florence, Florence, 50139, Italy
| | - Sandro Cosconati
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta, 81100, Italy
| | - Michele Roggia
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta, 81100, Italy
| | - Lorenzo Tagliazucchi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Lorenzo Gnudi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Valentina Puscio
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Chiara Sturaro
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, Ferrara, 44121, Italy
| | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via Luigi Borsari 46, Ferrara, 44121, Italy
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy; Technopole of Ferrara, Laboratory for Advanced Therapies (LTTA), via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Viale Pieraccini 6, University of Florence, Florence, 50139, Italy
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Viale Pieraccini 6, University of Florence, Florence, 50139, Italy.
| | - Delia Preti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy.
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Viale Pieraccini 6, University of Florence, Florence, 50139, Italy
| | - Salvatore Pacifico
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| |
Collapse
|
2
|
Chi X, Chen R, Chen R, Xu Y, Deng Y, Yang X, Pan Z, Xu X, Pan Y, Li Q, Zhou P, Huang W. Discovery and characterization of novel FAK inhibitors for breast cancer therapy via hybrid virtual screening, biological evaluation and molecular dynamics simulations. Bioorg Chem 2025; 159:108400. [PMID: 40163988 DOI: 10.1016/j.bioorg.2025.108400] [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: 01/10/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Focal adhesion kinase (FAK) is a critical drug target implicated in various disease pathways, including hematological malignancies and breast cancer. Therefore, identifying FAK inhibitors with novel scaffolds could offer new opportunities for developing effective therapeutic compounds. Herein, we disclosed the discovery of a new backbone inhibitor of FAK using an "internal" database, employing a structure-based high-transparency permeability virtual screening (HTVS) and a DeepDock algorithm based on geometric deep learning. Subsequently, molecular docking was conducted at different precisions to identify 10 compounds for further evaluation of biological activity. Ultimately, compound 4, a pyrimidin-4-amine derivative, demonstrated inhibitory activity against FAK and breast cancer cells, further supporting its potential as a FAK inhibitor. Moreover, molecular dynamics simulations were carried out to gain more detailed insights into the binding mechanism between compound 4 and FAK to guide subsequent structural optimization.
Collapse
Affiliation(s)
- Xinglong Chi
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Runmei Chen
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; School of Pharmacy, Hangzhou Medical College, Hangzhou 310058, PR China
| | - Roufen Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Yingxuan Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Yaru Deng
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Xinle Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China; College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Zhichao Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiangwei Xu
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Youlu Pan
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Qin Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310058, PR China.
| | - Peng Zhou
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China.
| | - Wenhai Huang
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310058, PR China; Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou 310053, PR China.
| |
Collapse
|
3
|
Poggialini F, Governa P, Vagaggini C, Maramai S, Lamponi S, Mugnaini C, Brizzi A, Purgatorio R, de Candia M, Catto M, Dreassi E, Manetti F, Corelli F, Altomare CD, Cappelli A, Paolino M. Light-mediated activation/deactivation control and in vitro ADME-Tox profiling of a donepezil-like Dual AChE/MAO-B Inhibitor. Eur J Pharm Sci 2025; 209:107066. [PMID: 40064401 DOI: 10.1016/j.ejps.2025.107066] [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: 01/07/2025] [Revised: 02/25/2025] [Accepted: 03/07/2025] [Indexed: 03/21/2025]
Abstract
The possibility to control the effects of drugs in time and space represents an ideal condition for developing safer and more personalized therapies against different disorders. In this context, photopharmacology has paved the way for the use of light in the modulation of drugs activity. Our interest is directed to photo-switchable molecules, capable of interconverting between two different isoforms upon light irradiation. We recently reported 1, a donepezil-like compound based on 2-benzylidenindan-1-one structure, as a dual AChE and MAO-B inhibitor, which can be converted into the E- (active form) and Z- (about tenfold less active form) diastereoisomers by irradiating with UV-vis light. Aiming at identifying compounds with remarkable activity in physiological conditions, we herein report a fine characterization of 1 in PBS solutions. First, we evaluated its ability to act as a photoswitch comparing PBS solution with organic solvents (e.g. methanol), demonstrating that a wavelength in the UV range (330 nm) can convert the E- into the Z-diastereoisomer, while the use of a visible light (400 nm) allows the interconversion from Z to E in both media. Along with its photoinducible behavior, we investigated the passive diffusion across cellular membrane with PAMPA experiments, plasma and microsomal stability, and binding to plasma proteins. Interestingly, the results of such studies suggested that 1 could persist in the blood circulation for a long time, which is desirable for application in photopharmacological therapies. Cytotoxicity studies highlighted the potential of our prototypic compound as a lead photodrug against neurodegenerative disorders, deserving to advance in molecular optimization studies and further in vitro and in vivo characterization.
Collapse
Affiliation(s)
- Federica Poggialini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Chiara Vagaggini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Samuele Maramai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Stefania Lamponi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Claudia Mugnaini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Antonella Brizzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Rosa Purgatorio
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Federico Corelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Marco Paolino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy.
| |
Collapse
|
4
|
Martínez León A, Ries B, Hub JS, Magarkar A. Moldrug algorithm for an automated ligand binding site exploration by 3D aware molecular enumerations. J Cheminform 2025; 17:85. [PMID: 40420238 DOI: 10.1186/s13321-025-01022-3] [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: 01/22/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
We present Moldrug, a computational tool for accelerating the hit-to-lead phase in structure-based drug design. Moldrug explores the chemical space using structural modifications suggested by the CReM library and by optimizing an adaptable fitness function with a genetic algorithm. Moldrug is complemented by Moldrug-Dashboard, a cross-platform and user-friendly graphical interface tailored for the analysis of Moldrug simulations. To illustrate Moldrug, we designed new potential inhibitors targeting the main protease (MPro) of SARS-CoV-2 by optimizing a consensus fitness function that balances binding affinity, drug-likeness, and synthetic accessibility. The designed molecules exhibited high chemical diversity. A subset of the designed molecules were ranked using MM/GBSA and alchemical binding free energy calculations, revealing predicted affinities as low as - 10 kcal mol - 1 . Moldrug is distributed as a Python package under the Apache 2.0 license. It offers pre-configured multi-parameter fitness functions for molecular design, while being highly adaptable for integrating functionalities from external software. Documentation and tutorials are available at https://moldrug.rtfd.io .
Collapse
Affiliation(s)
- Alejandro Martínez León
- Theoretical Physics and Center for Biophysics, Universität des Saarlandes, PharmaScienceHub (PSH), 66123, Saarbrücken, Germany
| | - Benjamin Ries
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany
- Open Molecular Software Foundation, Open Free Energy, Davis, CA, 95616, USA
| | - Jochen S Hub
- Theoretical Physics and Center for Biophysics, Universität des Saarlandes, PharmaScienceHub (PSH), 66123, Saarbrücken, Germany
| | - Aniket Magarkar
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany.
| |
Collapse
|
5
|
Gomes AAS, Giraldo J. Structural Determinants of Buprenorphine Partial Agonism at the μ-Opioid Receptor. J Chem Inf Model 2025; 65:5071-5085. [PMID: 40328437 PMCID: PMC12117568 DOI: 10.1021/acs.jcim.5c00078] [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: 01/17/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025]
Abstract
The μ-opioid receptor (μOR) is a class A G Protein-Coupled Receptor (GPCR) targeted by natural and synthetic ligands to provide analgesia to patients with pain of various etiologies. Available opioid medications present several unwanted side effects, stressing the need for safer pain therapies. Despite the attractive proposal that biasing μOR signaling toward G protein pathways would lead to fewer side effects, recent studies indicate that low-efficacy opioid drugs, such as buprenorphine, may represent a safer alternative. In the present work, we combine molecular docking, microsecond-time scale molecular dynamics (MD) simulations, and metadynamics to investigate the conformational dynamics of the μOR bound to morphine or buprenorphine. Our objective was to determine structural aspects associated with the unique pharmacological effects caused by the latter, taking morphine as a reference. MD simulations identified a salt bridge with D1493.32 as crucial for stabilizing both ligands into the μOR orthosteric site, with this interaction being weaker in buprenorphine. The morphinan-scaffold of both ligands shared contacts with transmembrane (TM) helix residues of the receptor, including TM3, TM5, TM6, and TM7. Conversely, while morphine showed stronger interactions with a few TM3 residues, additional chemical groups of buprenorphine showed stronger interactions with TM2, extracellular loop 2 (ECL2), and TM7 residues. We also observed distinct TM arrangements induced by these ligands, with buprenorphine causing an extracellular outward movement of TM7 and morphine provoking intracellular inward movements of TM5 and TM7 of the receptor. In addition, we found that buprenorphine tends to explore deeper regions in the μOR orthosteric site, further supported by funnel-metadynamics, resulting in diverse side chain orientations of W2956.48. Metadynamics also unveiled distinct intermediate states for morphine and buprenorphine, with the latter accessing a secondary binding site associated with partial μOR agonists. Our results indicate that the weakened salt bridge of buprenorphine with D1493.32, along with the strong TM7 interaction through its cyclopropyl group, may explain its low efficacy and consequent partial μOR agonism. Furthermore, ECL2 interactions may contribute to explaining the biased agonism of buprenorphine, a common feature shared with other opioid modulators with similar functional effects. Our study sheds light on the complex pharmacology of buprenorphine, identifying structural aspects associated with its partial and biased μOR agonism. These results can provide valuable information for the design of new effective and safer opioid drugs.
Collapse
Affiliation(s)
- Antoniel A. S. Gomes
- Laboratory
of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística
and Institut de Neurociències, Universitat
Autònoma de Barcelona, 08193Bellaterra, Spain
- Unitat
de Neurociència Traslacional, Parc Taulí Hospital Universitari,
Institut d’Investigació i Innovació Parc Taulí
(I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193Bellaterra, Spain
- Instituto
de Salud Carlos III, Centro de Investigación Biomédica
en Red de Salud Mental, CIBERSAM, 28029Madrid, Spain
| | - Jesús Giraldo
- Laboratory
of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística
and Institut de Neurociències, Universitat
Autònoma de Barcelona, 08193Bellaterra, Spain
- Unitat
de Neurociència Traslacional, Parc Taulí Hospital Universitari,
Institut d’Investigació i Innovació Parc Taulí
(I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193Bellaterra, Spain
- Instituto
de Salud Carlos III, Centro de Investigación Biomédica
en Red de Salud Mental, CIBERSAM, 28029Madrid, Spain
| |
Collapse
|
6
|
Li C, Wang Y, Xing H, Wang Y, Wang Y, Ye J. Vina-CUDA: An Efficient Program with in-Depth Utilization of GPU to Accelerate Molecular Docking. J Chem Inf Model 2025; 65:4751-4759. [PMID: 40377068 DOI: 10.1021/acs.jcim.4c01933] [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: 05/18/2025]
Abstract
As a mainstream technology in modern drug discovery, molecular docking methodologies enable precise and efficient identification of lead compounds within large chemical repositories to improve drug development efficiency and reduce costs. The exponential growth of chemical databases has substantially expanded drug discovery resources while improving the identification rates of true positives in lead compounds. However, this rapid expansion poses significant challenges for existing docking tools to efficiently screen lead compounds from these massive chemical libraries. In this study, we proposed Vina-CUDA, which leverages GPU hardware features to optimize and accelerate the core algorithm of the popular tool AutoDock Vina at three aspects, computational capability, memory access, and resource utilization, significantly improving docking efficiency. A hybrid parallel optimization strategy integrating task and computational parallelism was implemented, accompanied by systematic code and data structure optimization, to maximize GPU resource utilization and enhance computational efficiency. Building upon this, we developed its derivatives, QuickVina2-CUDA and QuickVina-W-CUDA, as well as a user-friendly multi-GPU docking framework to utilize multi-GPU resources to accelerate large-scale virtual screening tasks. The performance and docking accuracy of Vina-CUDA and its derivatives were evaluated under five chemical databases. Results showed that, compared to baseline programs, Vina-CUDA with RILC-BFGS optimization algorithm achieved average and maximum accelerations of 3.71× and 6.89× across five databases, while QuickVina2-CUDA and QuickVina-W-CUDA achieved average speedups of 6.19× and 1.46×, respectively, without compromising docking accuracy. Furthermore, Vina-CUDA and its derivatives demonstrated comparable performance to baseline docking programs in docking, scoring, and ranking power, with excellent scalability and portability.
Collapse
Affiliation(s)
- Chunfeng Li
- School of Computer Science and Technology, Beijing Institute of Technology, No. 5 Zhongguancun South Street, Haidian District, Beijing 100081, China
| | - Yizhuo Wang
- School of Computer Science and Technology, Beijing Institute of Technology, No. 5 Zhongguancun South Street, Haidian District, Beijing 100081, China
| | - Hongbo Xing
- School of Computer Science and Technology, Beijing Institute of Technology, No. 5 Zhongguancun South Street, Haidian District, Beijing 100081, China
| | - Yidan Wang
- School of Computer Science and Technology, Beijing Institute of Technology, No. 5 Zhongguancun South Street, Haidian District, Beijing 100081, China
| | - Yang Wang
- School of Computer Science and Technology, Beijing Institute of Technology, No. 5 Zhongguancun South Street, Haidian District, Beijing 100081, China
| | - Jiawei Ye
- School of Computer Science and Technology, Beijing Institute of Technology, No. 5 Zhongguancun South Street, Haidian District, Beijing 100081, China
| |
Collapse
|
7
|
Xu M, Wu C, Wang S, Zhan W, Guo L, Li Y, Vogel H, Yuan S. Identifying Potent Compounds Using Pairwise Consensus Methods. J Chem Inf Model 2025. [PMID: 40366258 DOI: 10.1021/acs.jcim.5c00942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Molecular docking is a widely used method within the in silico compound screening process of modern drug discovery. The accuracy of this method for predicting high-affinity small-molecule binders for a target protein from a large chemical library can be substantially improved by combining individual docking tools for cross-validation. This traditional consensus strategy typically relies on averaging scores or ranks obtained from molecular docking, which are, however, vulnerable to false positives and thus exploit shortcomings from scoring functions. To overcome this remarkable weakness, we developed here the pairwise consensus score (PCS) algorithm. PCS integrates structural similarity information on ligand-receptor complexes to evaluate predicted conformations and penalize highly dissimilar docked poses. To demonstrate the versatility of PCS, we developed a consensus docking protocol for targeting G protein-coupled receptors (GPCRs) that are among the most important targets for modern drug discovery. In particular, we screened a large compound library for highly potent antagonism ligands to an important GPCR therapeutic target, the neurokinin 1 receptor, and found several compounds targeting the receptor with ten-picomolar activity. Notably, these highly active compounds show a totally different chemical structure from that of previously reported NK1 binders. This opens exciting opportunities to develop drugs with unique alternative pharmacological features and therapeutic value.
Collapse
Affiliation(s)
- Marc Xu
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenyang Wu
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shiyu Wang
- AlphaMol Science Ltd, Shenzhen, Guangdong 518055, China
| | - Wenjin Zhan
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Liwei Guo
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yi Li
- AlphaMol Science Ltd, Shenzhen, Guangdong 518055, China
| | - Horst Vogel
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- AlphaMol Science Ltd, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
- Institut des Sciences et Ingénierie Chimiques (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland
| | - Shuguang Yuan
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- AlphaMol Science Ltd, Shenzhen, Guangdong 518055, China
| |
Collapse
|
8
|
Ha YJJ, Nisal A, Tang I, Lee C, Jhamb I, Wallace C, Howarth R, Schroeder S, Vong KI, Meave N, Jiwani F, Barrows C, Lee S, Jiang N, Patel A, Bagga K, Banka N, Friedman L, Blanco FA, Yu S, Rhee S, Jeong HS, Plutzer I, Major MB, Benoit B, Poüs C, Heffner C, Kibar Z, Bot GM, Northrup H, Au KS, Strain M, Ashley-Koch AE, Finnell RH, Le JT, Meltzer HS, Araujo C, Machado HR, Stevenson RE, Yurrita A, Mumtaz S, Ahmed A, Khara MH, Mutchinick OM, Medina-Bereciartu JR, Hildebrandt F, Melikishvili G, Marwan AI, Capra V, Noureldeen MM, Salem AMS, Issa MY, Zaki MS, Xu L, Lee JE, Shin D, Alkelai A, Shuldiner AR, Kingsmore SF, Murray SA, Gee HY, Miller WT, Tolias KF, Wallingford JB, Kim S, Gleeson JG. The contribution of de novo coding mutations to meningomyelocele. Nature 2025; 641:419-426. [PMID: 40140573 DOI: 10.1038/s41586-025-08676-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 01/20/2025] [Indexed: 03/28/2025]
Abstract
Meningomyelocele (also known as spina bifida) is considered to be a genetically complex disease resulting from a failure of the neural tube to close. Individuals with meningomyelocele display neuromotor disability and frequent hydrocephalus, requiring ventricular shunting. A few genes have been proposed to contribute to disease susceptibility, but beyond that it remains unexplained1. We postulated that de novo mutations under purifying selection contribute to the risk of developing meningomyelocele2. Here we recruited a cohort of 851 meningomyelocele trios who required shunting at birth and 732 control trios, and found that de novo likely gene disruption or damaging missense mutations occurred in approximately 22.3% of subjects, with 28% of such variants estimated to contribute to disease risk. The 187 genes with damaging de novo mutations collectively define networks including actin cytoskeleton and microtubule-based processes, Netrin-1 signalling and chromatin-modifying enzymes. Gene validation demonstrated partial or complete loss of function, impaired signalling and defective closure of the neural tube in Xenopus embryos. Our results indicate that de novo mutations make key contributions to meningomyelocele risk, and highlight critical pathways required for neural tube closure in human embryogenesis.
Collapse
Affiliation(s)
- Yoo-Jin Jiny Ha
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ashna Nisal
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Isaac Tang
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Chanjae Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Ishani Jhamb
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Cassidy Wallace
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Robyn Howarth
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Sarah Schroeder
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Keng Ioi Vong
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Naomi Meave
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Fiza Jiwani
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Chelsea Barrows
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Sangmoon Lee
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Nan Jiang
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Arzoo Patel
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Krisha Bagga
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Niyati Banka
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Liana Friedman
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Francisco A Blanco
- Department of Neuroscience, Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Seyoung Yu
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soeun Rhee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hui Su Jeong
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Isaac Plutzer
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, MO, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, MO, USA
| | - Béatrice Benoit
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| | - Christian Poüs
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
- Biochimie-Hormonologie, Assistance Publique - Hôpitaux de Paris Université Paris-Saclay, Clamart, France
| | | | - Zoha Kibar
- Department of Neurosciences, Research Center of CHU Sainte Justine, University of Montreal, Montreal, Quebec, Canada
| | - Gyang Markus Bot
- Neurosurgery Division, Department of Surgery, Jos University Teaching Hospital, Jos, Nigeria
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Kit Sing Au
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Madison Strain
- Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Richard H Finnell
- Center for Precision Environmental Health, Departments of Molecular and Human Genetics, Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Joan T Le
- Rady Children's Hospital, San Diego, CA, USA
| | | | - Camila Araujo
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Helio R Machado
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Roger E Stevenson
- J. C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, SC, USA
| | - Anna Yurrita
- Catedrática de Ciencias Ómicas, Facultad de Medicina, Universidad Francisco Marroquín, Guatemala City, Guatemala
| | - Sara Mumtaz
- National University of Medical Sciences, Rawalpindi, Pakistan
| | | | | | - Osvaldo M Mutchinick
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | | | - Gia Melikishvili
- Department of Pediatrics, MediClubGeorgia Medical Center, Tbilisi, Georgia
| | - Ahmed I Marwan
- Division of Pediatric General, Thoracic and Fetal Surgery, Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Valeria Capra
- Genomics and Clinical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Mahmoud M Noureldeen
- Department of Pediatrics, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Aida M S Salem
- Department of Pediatrics, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Mahmoud Y Issa
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Ji Eun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Donghyuk Shin
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | | - Heon Yung Gee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
- VA Medical Center, Northport, NY, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- POSTECH Biotechnology Center, Pohang University of Science and Technology, Pohang, Republic of Korea.
| | - Joseph G Gleeson
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA.
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
9
|
Wu H, Li Z, Zhong Z, Guo Y, He L, Xu X, Mao Y, Tang D, Zhang W, Jin F, Pang R. Insect Cytochrome P450 Database: An Integrated Resource of Genetic Diversity, Evolution and Function. Mol Ecol Resour 2025; 25:e14070. [PMID: 39776220 DOI: 10.1111/1755-0998.14070] [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: 06/27/2024] [Revised: 11/16/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Insects, the most numerous and diverse group of animal species on Earth, have important interactions with humans through providing resources, transmitting diseases and damaging agricultural cultivars. Cytochrome P450 monooxygenases (P450s) are one of the most important protein families in insects implicated in the endogenous metabolism and detoxification of xenobiotics, including allelochemicals, insecticides and environmental pollutants. To better understand the evolution and function of insect P450s and support the development and application of insecticides for pest control, an integrated bioinformatics platform is highly desirable. Here, we present the Insect Cytochrome P450 database (ICPD, http://www.insectp450.net/), which contains 66,477 P450s collected from public databases and predicted from the genomes of 682 insect species using a standardised bioinformatics protocol. Phylogenetic relationships between P450 genes are constructed for each species. The structures of all P450 proteins in the database are predicted using ESMFold, then visualised using WeView. Web services, such as BLAST, homogeneous modelling and molecular docking, are provided for determining the catalytic activities of P450 proteins. The ICPD will facilitate systematic investigations of the evolution and functions of the complete insect P450 complement, and represents a powerful tool for guiding insecticide design and application.
Collapse
Affiliation(s)
- Hongxin Wu
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Zhongsheng Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Zichun Zhong
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Yujing Guo
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Liuyan He
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Xiaoxia Xu
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Yijun Mao
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, China
| | - Deyu Tang
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, China
| | - Wenqing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Fengliang Jin
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Rui Pang
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou, China
| |
Collapse
|
10
|
He J, Tang H, Liao R, Lin H, Zhang W. Gemini surfactant stabilized zein nanoparticles: Preparation, characterization, interaction mechanism, and antibacterial activity. Int J Biol Macromol 2025; 305:141264. [PMID: 39984070 DOI: 10.1016/j.ijbiomac.2025.141264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
To enhance the physicochemical properties of zein nanoparticles, zein complexes with two Gemini surfactants (12-3-12 and 12-4-12) were prepared using the anti-solvent method and investigated the physicochemical properties, formation mechanism and antibacterial activity. Results indicated that the optimal mass ratio between zein and Gemini surfactants was at 1:1, and the incorporation of Gemini surfactants significantly improved the surface properties of zein, reducing its surface hydrophobicity and surface tension, thereby enhancing its dispersion in aqueous media. Fluorescence spectroscopy and molecular docking experiments further elucidated the interaction mechanisms between zein and Gemini surfactant, revealing a spontaneous binding process, mainly driven by hydrophobic and hydrogen interaction, and a strong binding affinity of 12-4-12 with zein. Additionally, the zein/Gemini surfactant complexes exhibited significant antibacterial activity against Staphylococcus aureus, with the zein/12-4-12 complex showing particularly prominent inhibitory effects. Therefore, this research not only provides a theoretical foundation for the construction of Gemini surfactant stabilized zein nanoparticles but also points the way for the subsequent embedding of bacteriostatic agents to achieve synergistic antibacterial effects.
Collapse
Affiliation(s)
- Junbo He
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, China; Engineering Research Center of Lipid-based Fine Chemicals of Hubei Province, Wuhan 43023, China.
| | - Hong Tang
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ruifeng Liao
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Hong Lin
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, China; Engineering Research Center of Lipid-based Fine Chemicals of Hubei Province, Wuhan 43023, China
| | - Weinong Zhang
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, College of Food Science & Engineering, Wuhan Polytechnic University, Wuhan 430023, China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, China; Engineering Research Center of Lipid-based Fine Chemicals of Hubei Province, Wuhan 43023, China
| |
Collapse
|
11
|
Martelli A, Abate F, Roggia M, Benedetti G, Caradonna E, Calderone V, Tenore GC, Cosconati S, Novellino E, Stornaiuolo M. Trimethylamine N-Oxide (TMAO) Acts as Inhibitor of Endothelial Nitric Oxide Synthase (eNOS) and Hampers NO Production and Acetylcholine-Mediated Vasorelaxation in Rat Aortas. Antioxidants (Basel) 2025; 14:517. [PMID: 40427399 PMCID: PMC12108457 DOI: 10.3390/antiox14050517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Trimethylamine N-oxide (TMAO) is an endogenous osmolyte produced by enzymatic reactions starting in the human gut, where microbiota release trimethylamine (TMA) from foods, and ending in the liver, where TMA is oxidized to TMAO by flavin-containing monooxygenase 3 (FMO3). While physiological concentrations of TMAO help proteins preserve their folding, high levels of this metabolite are harmful and promote oxidative stress, inflammation, and atherosclerosis. In humans, elevated levels of circulating TMAO predispose individuals to cardiovascular diseases and chronic kidney disease and increase mortality risk, especially in the elderly. How TMAO exerts its negative effects has been only partially elucidated. In hypertensive rats, the eNOS substrate L-arginine and Taurisolo®, a nutraceutical endowed with TMAO-reducing activity, act synergistically to reduce arterial blood pressure. Here, we investigate the molecular mechanisms underpinning this synergism and prove that TMAO, the target of Taurisolo®, acts as direct inhibitor of endothelial nitric oxide synthase (eNOS) and competes with L-arginine at its catalytic site, ultimately inhibiting NO production and acetylcholine (Ach)-induced relaxation in murine aortas.
Collapse
Affiliation(s)
- Alma Martelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56120 Pisa, Italy; (A.M.); (G.B.); (V.C.)
| | - Federico Abate
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy; (F.A.); (M.R.); (S.C.)
| | - Michele Roggia
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy; (F.A.); (M.R.); (S.C.)
| | - Giada Benedetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56120 Pisa, Italy; (A.M.); (G.B.); (V.C.)
| | - Eugenio Caradonna
- Centro Diagnostico Italiano, Department of Clinical Laboratory, 20100 Milan, Italy;
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56120 Pisa, Italy; (A.M.); (G.B.); (V.C.)
| | - Gian Carlo Tenore
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy;
| | - Sandro Cosconati
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy; (F.A.); (M.R.); (S.C.)
| | - Ettore Novellino
- Department of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy;
| |
Collapse
|
12
|
Kolokolov M, Sannikova N, Dementev S, Podarov R, Zhdanova K, Bragina N, Chubarov A, Fedin M, Krumkacheva O. Enhanced Binding Site Identification in Protein-Ligand Complexes with a Combined Blind Docking and Dipolar Electron Paramagnetic Resonance Approach. J Am Chem Soc 2025; 147:13677-13687. [PMID: 40214089 DOI: 10.1021/jacs.5c01274] [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: 04/24/2025]
Abstract
Understanding protein-drug complex structures is crucial for elucidating therapeutic mechanisms and side effects. Blind docking facilitates site identification but is hindered by computational complexity and imprecise scoring, causing ambiguity. Dipolar electron paramagnetic resonance (EPR) provides spin-spin distances but struggles to determine relative positions within complexes. We present a novel approach combining GPU-accelerated blind docking with EPR distance constraints to enhance binding site detection. Our algorithm uses a single EPR distance distribution to filter and validate docking results. Ligand poses from blind docking are clustered, filtered by expected distances, and refined through focused docking. To illustrate our approach, we investigated human serum albumin binding with porphyrin-based photosensitizers used in photodynamic therapy. Combining docking and EPR, we identified possible binding sites, demonstrating that EPR data significantly reduce possible configurations and provide experimentally validated information. This strategy produces a detailed map of photoligand binding sites, revealing that binding may occur away from standard albumin sites and often involves multiple locations. Furthermore, it overcomes key limitations of fluorescence-based methods, which are prone to misinterpretation in albumin studies due to non one-to-one donor-acceptor relationships. By resolving ambiguities in both blind docking and EPR, our framework provides a versatile platform for investigating EPR-active ligands.
Collapse
Affiliation(s)
- Mikhail Kolokolov
- EPR Laboratory, International Tomography Center SB RAS, 3 Institutskaya Street, Novosibirsk 630090, Russia
- Physics Department, Novosibirsk State University, 1 Pirogova Street, Novosibirsk 630090, Russia
| | - Natalya Sannikova
- EPR Laboratory, International Tomography Center SB RAS, 3 Institutskaya Street, Novosibirsk 630090, Russia
- Physics Department, Novosibirsk State University, 1 Pirogova Street, Novosibirsk 630090, Russia
| | - Sergei Dementev
- EPR Laboratory, International Tomography Center SB RAS, 3 Institutskaya Street, Novosibirsk 630090, Russia
- Physics Department, Novosibirsk State University, 1 Pirogova Street, Novosibirsk 630090, Russia
| | - Roman Podarov
- EPR Laboratory, International Tomography Center SB RAS, 3 Institutskaya Street, Novosibirsk 630090, Russia
- Physics Department, Novosibirsk State University, 1 Pirogova Street, Novosibirsk 630090, Russia
| | - Kseniya Zhdanova
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, 78 Vernadsky Avenue, Moscow 119454, Russia
| | - Natal'ya Bragina
- Institute of Fine Chemical Technology, MIREA-Russian Technological University, 78 Vernadsky Avenue, Moscow 119454, Russia
| | - Alexey Chubarov
- Department of Physics, Free University of Berlin, Arnimallee 14, Berlin 14195, Germany
| | - Matvey Fedin
- EPR Laboratory, International Tomography Center SB RAS, 3 Institutskaya Street, Novosibirsk 630090, Russia
- Physics Department, Novosibirsk State University, 1 Pirogova Street, Novosibirsk 630090, Russia
| | - Olesya Krumkacheva
- EPR Laboratory, International Tomography Center SB RAS, 3 Institutskaya Street, Novosibirsk 630090, Russia
- Physics Department, Novosibirsk State University, 1 Pirogova Street, Novosibirsk 630090, Russia
| |
Collapse
|
13
|
Zhao CX, Yan YF, Zhao LX, Tang X, Chen YG, Song WJ, Long LP, Chen J, Tan CL, Zhang QZ, Pu XL, Shen QQ, Fan YZ, Tao Y, Ye X, Li SH, Liu Y. Characterization of a 4'-O-rhamnosyltransferase and de novo biosynthesis of bioactive steroidal triglycosides from Paris polyphylla. PLANT COMMUNICATIONS 2025; 6:101257. [PMID: 39844466 PMCID: PMC12010398 DOI: 10.1016/j.xplc.2025.101257] [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: 10/14/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Steroidal saponins in Paris polyphylla featuring complicated sugar chains exhibit notable biological activities, but their sugar-chain biosynthesis is still not fully understood. Here, we identified a 4'-O-rhamnosyltransferase (UGT73DY2) from P. polyphylla, which catalyzes the 4'-O-rhamnosylation of polyphyllins V and VI, producing dioscin and pennogenin 3-O-β-chacotrioside, respectively. UGT73DY2 exhibits strict substrate specificity toward steroidal diglycosides and UDP-rhamnose, and a new steroidal triglycoside can be synthesized through enzyme catalysis. A mutation library was generated based on semi-rational design, identifying three mutants, I358T, A342V, and A132T, which displayed approximately two-fold enhanced enzyme activity. Molecular dynamics simulations revealed that shortened distances between the 4'-OH group of the sugar acceptor and either the crucial residue H20 or the donor UDP-Rha contribute to the enhanced enzyme activity. Moreover, subcellular localization analysis of UGT73DY2 and other biosynthetic enzymes indicated that dioscin biosynthesis predominantly occurs in the endoplasmic reticulum of plant cells. By co-expressing 14 biosynthetic genes in Nicotiana benthamiana, optimizing HMGR subcellular localization and cytochrome P450 gene sets, and engineering UGT73DY2, we successfully established a dioscin biosynthesis system with a yield of 3.12 ± 0.11 μg/g dry weight. This study not only clarifies the 4'-O-rhamnosylation process in steroidal saponin biosynthesis but also presents an alternative approach for the production of steroidal saponins in P. polyphylla through synthetic biology and metabolic engineering.
Collapse
Affiliation(s)
- Chen-Xiao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Yuan-Feng Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Li-Xiao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Xue Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Yue-Gui Chen
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P.R. China
| | - Wen-Jun Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Li-Ping Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Jing Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Chun-Lin Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Qiao-Zhuo Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Xiu-Lan Pu
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Qin-Qin Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Yu-Zhou Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Yang Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Xiao Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China
| | - Sheng-Hong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China; State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P.R. China.
| | - Yan Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P.R. China.
| |
Collapse
|
14
|
Chi X, Chen R, Yang X, He X, Pan Z, Yao C, Peng H, Yang H, Huang W, Chen Z. Discovery of Novel DDR1 Inhibitors through a Hybrid Virtual Screening Pipeline, Biological Evaluation and Molecular Dynamics Simulations. ACS Med Chem Lett 2025; 16:602-610. [PMID: 40236534 PMCID: PMC11995236 DOI: 10.1021/acsmedchemlett.4c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/18/2025] [Accepted: 02/20/2025] [Indexed: 04/17/2025] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy with limited therapeutic options for many patients. Discoidin domain receptor 1 (DDR1), a transmembrane tyrosine kinase receptor, has been implicated in AML progression and represents a promising therapeutic target. In this study, we employed a hybrid virtual screening workflow that integrates deep learning-based binding affinity predictions with molecular docking techniques to identify potential DDR1 inhibitors. A multistage screening process involving PSICHIC, KarmaDock, Vina-GPU, and similarity-based scoring was conducted, leading to the selection of seven candidate compounds. The biological evaluation identified Compound 4 as a novel DDR1 inhibitor, demonstrating significant DDR1 inhibitory activity with an IC50 of 46.16 nM and a 99.86% inhibition rate against Z-138 cells at 10 μM. Molecular dynamics simulations and binding free energy calculations further validated the stability and strong binding interactions of Compound 4 with DDR1. This study highlights the utility of combining deep learning models with traditional molecular docking techniques to accelerate the discovery of potent and selective DDR1 inhibitors. The identified compounds hold promise for further development as targeted therapies for AML.
Collapse
Affiliation(s)
- Xinglong Chi
- Department
of Hematology, Tongde Hospital of Zhejiang
Province, No. 234, Gucui Road, Hangzhou 310012, Zhejiang, P.R. China
- Affiliated
Yongkang First People’s Hospital and School of Pharmaceutical
Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
| | - Roufen Chen
- College
of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinle Yang
- College
of Pharmaceutical Sciences, Zhejiang University
of Technology, Hangzhou 310014, China
| | - Xinjun He
- College
of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhichao Pan
- College
of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chenpeng Yao
- College
of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huilin Peng
- Department
of Lymphoma, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Haiyan Yang
- Department
of Lymphoma, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Wenhai Huang
- Affiliated
Yongkang First People’s Hospital and School of Pharmaceutical
Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
| | - Zhilu Chen
- Department
of Hematology, Tongde Hospital of Zhejiang
Province, No. 234, Gucui Road, Hangzhou 310012, Zhejiang, P.R. China
| |
Collapse
|
15
|
Liu X, Sun S, Xia X, Shi H, Yang L, Mao Z, Xiao X, Zhou Y, Qing Z. Uncovering the Fluctuation of Peroxynitrite during Early Embryonic Development Using an Integrative Nanobeacon. Anal Chem 2025; 97:6192-6200. [PMID: 40085785 DOI: 10.1021/acs.analchem.4c06987] [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: 03/16/2025]
Abstract
Embryonic development is the beginning of life, and various kinds of bioactive molecules are implicated in this crucial process. Especially in its early stage, some important biochemical reactions regulating physiological balance may be resuscitated. Thus, revealing the dynamic changes of bioactive molecules during early embryonic development is crucial to the elucidation of biological phenomena. Peroxynitrite (ONOO-) is a typical signaling molecule in intercellular communication. However, up to now, no work has studied the fluctuation of ONOO- during early embryonic development due to its low content, especially in mammals. Herein, a polymeric nanobeacon that integrates an ONOO--responsive degradable scaffold and a fluorescence amplification module, named IFN, was designed to selectively sense embryonic ONOO- with high sensitivity. By virtue of the specific dye, ONOO- was sensitively detected in the range of 0-4.5 μM with a detection limit of 20.4 nM. From the attractive embryonic results, a sudden increase in ONOO- content after fertilization was observed in a mammalian model, while the level of ONOO- decreased slightly at the four-cell and eight-cell stages, finally reaching an equilibrium throughout the morula and blastocyst stages. This phenomenon is due to the resuscitation of ovotids, the activation of some life events by fertilization, and the subsequent establishment of physiological balance. This work not only suggests that ONOO- plays a positive role in normal embryonic development but also highlights the molecular events occurring at the initial phase of life. Furthermore it opens up new avenues for monitoring chemical changes during mammalian embryonic development.
Collapse
Affiliation(s)
- Xiaowen Liu
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Pharmaceutical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, PR China
| | - Shuanghong Sun
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Pharmaceutical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Xinchao Xia
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Pharmaceutical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Huiqiu Shi
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Pharmaceutical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Le Yang
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Pharmaceutical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Zenghui Mao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, PR China
| | - Xianjin Xiao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yibo Zhou
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Pharmaceutical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Zhihe Qing
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Pharmaceutical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| |
Collapse
|
16
|
Carvajal-Patiño JG, Mallet V, Becerra D, Niño Vasquez LF, Oliver C, Waldispühl J. RNAmigos2: accelerated structure-based RNA virtual screening with deep graph learning. Nat Commun 2025; 16:2799. [PMID: 40118849 PMCID: PMC11928640 DOI: 10.1038/s41467-025-57852-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/01/2025] [Indexed: 03/24/2025] Open
Abstract
RNAs are a vast reservoir of untapped drug targets. Structure-based virtual screening (VS) identifies candidate molecules by leveraging binding site information, traditionally using molecular docking simulations. However, docking struggles to scale with large compound libraries and RNA targets. Machine learning offers a solution but remains underdeveloped for RNA due to limited data and practical evaluations. We introduce a data-driven VS pipeline tailored for RNA, utilizing coarse-grained 3D modeling, synthetic data augmentation, and RNA-specific self-supervision. Our model achieves a 10,000x speedup over docking while ranking active compounds in the top 2.8% on structurally distinct test sets. It is robust to binding site variations and successfully screens unseen RNA riboswitches in a 20,000-compound in-vitro microarray, with a mean enrichment factor of 2.93 at 1%. This marks the first experimentally validated success of structure-based deep learning for RNA VS.
Collapse
Affiliation(s)
- Juan G Carvajal-Patiño
- School of Computer Science, McGill University, Montréal, QC, Canada
- Universidad Nacional de Colombia - Sede Bogotá - Facultad de Ingeniería - Depto. de Ingeniería de Sistemas e Industrial, Bogotá, Colombia
| | - Vincent Mallet
- LIX, Ecole Polytechnique, IP, Paris, France
- Mines Paris, PSL Research University, CBIO-Center of Computational Biology, Paris, France
- Institut Curie, PSL Research University, Paris, France
- INSERM, Paris, France
| | - David Becerra
- School of Computer Science, McGill University, Montréal, QC, Canada
- Universidad Nacional de Colombia - Sede Bogotá - Facultad de Ingeniería - Depto. de Ingeniería de Sistemas e Industrial, Bogotá, Colombia
| | - Luis Fernando Niño Vasquez
- Universidad Nacional de Colombia - Sede Bogotá - Facultad de Ingeniería - Depto. de Ingeniería de Sistemas e Industrial, Bogotá, Colombia
| | - Carlos Oliver
- Max Planck Institute of Biochemistry, Martinsried, Germany.
- Center for AI in Protein Dynamics, Vanderbilt University, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| | - Jérôme Waldispühl
- School of Computer Science, McGill University, Montréal, QC, Canada.
| |
Collapse
|
17
|
Xu Q, Authi KS, Kirpotina LN, Schepetkin IA, Quinn MT, Cilibrizzi A. Development of small-molecule fluorescent probes targeting neutrophils via N-formyl peptide receptors. RSC Med Chem 2025; 16:1397-1409. [PMID: 39886349 PMCID: PMC11775818 DOI: 10.1039/d4md00849a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025] Open
Abstract
N-Formyl peptide receptors (FPRs) are membrane receptors that are abundantly expressed in innate immune cells, including neutrophils and platelets, demonstrating potential new targets for immune system regulation and the treatment of inflammatory conditions. We report here the development and bio-physical validation of new FPR imaging agents as effective tools to track FPR distribution, localisation and functions, ultimately helping to establish FPR exact roles and functions in pathological and physiological conditions. The new series of probes feature a small molecule-based FPR address system conjugated to suitable fluorophores, resulting in highly specific FPR agents, including a partial agonist endowed with high affinity (i.e. low/sub-nanomolar potency) on FPR-transfected cells and human neutrophils. Preliminary imaging studies via multiphoton microscopy demonstrate that the probes enable the visualisation of FPRs in live cells, thus representing valid bio-imaging tools for the analysis of FPR-mediated signalling, such as the activation of neutrophils in inflammatory events.
Collapse
Affiliation(s)
- Qi Xu
- Institute of Pharmaceutical Science, King's College London Stamford Street London SE1 9NH UK +44 (0) 20 7848 9532
| | - Kalwant S Authi
- BHF Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London London SE1 9NH UK
| | - Liliya N Kirpotina
- Department of Microbiology and Cell Biology, Montana State University Bozeman Montana 59717 USA
| | - Igor A Schepetkin
- Department of Microbiology and Cell Biology, Montana State University Bozeman Montana 59717 USA
| | - Mark T Quinn
- Department of Microbiology and Cell Biology, Montana State University Bozeman Montana 59717 USA
| | - Agostino Cilibrizzi
- Institute of Pharmaceutical Science, King's College London Stamford Street London SE1 9NH UK +44 (0) 20 7848 9532
| |
Collapse
|
18
|
Tang L, Cheng H, Yang Q, Xie Y, Zhang Q. Umbelliferone as an effective component of Rhodiola for protecting the cerebral microvascular endothelial barrier in cSVD. Front Pharmacol 2025; 16:1552579. [PMID: 40166460 PMCID: PMC11955776 DOI: 10.3389/fphar.2025.1552579] [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: 12/28/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Objective Rhodiola is a common Chinese herb in the treatment of cerebral small vessel disease (cSVD). Umbelliferone, one of the effective components of Rhodiola, can protect the endothelial barrier. But its mechanisms are still unclear. Therefore, this study is aimed to explore mechanisms of umbelliferone of an effective component of Rhodiola in protecting the cerebral microvascular endothelial barrier in cSVD. Methods Firstly, ETCM, SwissTargetPrediction and literatures were used to screen components and targets of Rhodiola. GeneCards was used to obtain targets of cSVD. STRING and Cytoscape were utilized for building the PPI and C-T network. Metascape was utilized to construct GO and KEGG enrichment analysis. Then, molecular docking was employed to evaluate the binding ability of the compounds for their respective target molecules. Ultimately, the endothelial cell damage caused by OGD was employed to explore the protective impact of umbelliferone, a bioactive constituent of Rhodiola, on the endothelial barrier. Endothelial cell leakage and migration assays were used to assess the permeability and migration ability of endothelial cells. IF and WB techniques were employed to ascertain the expression of endothelial tight junction protein. The major target proteins and related pathways were validated by WB. Results Six effective components and 106 potential targets were identified and 1885 targets of cSVD were obtained. Nine key targets were selected. GO and KEGG enrichment analysis suggested that effects of Rhodiola in cSVD were associated with PI3K-Akt, Ras, Rap1 and MAPK signal pathways. Molecular docking results showed good binding ability between 28 pairs of key proteins and compounds. Umbelliferone of an effective component of Rhodiola can protect tight junction proteins and improve the permeability and migration ability of endothelial cells damaged by OGD through MMP9, MMP2, CCND1, PTGS2 and PI3K-Akt, Ras, Rap1 signaling pathways. Conclusion Our study systematically clarified mechanisms of Rhodiola in treating cSVD by network pharmacology and molecular docking, characterized by its multi-component, multi-target and multi-pathway effects. This finding was validated through in vitro tests, which demonstrated that umbelliferone of an effective component in Rhodiola can protect the brain microvascular endothelial barrier. It provided valuable ideas and references for additional research.
Collapse
Affiliation(s)
| | | | | | | | - Qiuxia Zhang
- College of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Mohammed I, Sagurthi SR. Current Approaches and Strategies Applied in First-in-class Drug Discovery. ChemMedChem 2025; 20:e202400639. [PMID: 39648151 DOI: 10.1002/cmdc.202400639] [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: 08/16/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
First-in-class drug discovery (FICDD) offers novel therapies, new biological targets and mechanisms of action (MOAs) toward targeting various diseases and provides opportunities to understand unexplored biology and to target unmet diseases. Current screening approaches followed in FICDD for discovery of hit and lead molecules can be broadly categorized and discussed under phenotypic drug discovery (PDD) and target-based drug discovery (TBDD). Each category has been further classified and described with suitable examples from the literature outlining the current trends in screening approaches applied in small molecule drug discovery (SMDD). Similarly, recent applications of functional genomics, structural biology, artificial intelligence (AI), machine learning (ML), and other such advanced approaches in FICDD have also been highlighted in the article. Further, some of the current medicinal chemistry strategies applied during discovery of hits and optimization studies such as hit-to-lead (HTL) and lead optimization (LO) have been simultaneously overviewed in this article.
Collapse
Affiliation(s)
- Idrees Mohammed
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
| | - Someswar Rao Sagurthi
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
20
|
Lv X, Kang Y, Chi X, Zhao J, Pan Z, Ying X, Li L, Pan Y, Huang W, Wang L. A Hybrid Energy-Based and AI-Based Screening Approach for the Discovery of Novel Inhibitors of AXL. ACS Med Chem Lett 2025; 16:410-419. [PMID: 40110119 PMCID: PMC11921171 DOI: 10.1021/acsmedchemlett.4c00511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 03/22/2025] Open
Abstract
AXL, part of the TAM receptor tyrosine kinase family, plays a significant role in the growth and survival of various tissues and tumors, making it a critical target for cancer therapy. This study introduces a novel high-throughput virtual screening (HTVS) methodology that merges an AI-enhanced graph neural network, PLANET, with a geometric deep learning algorithm, DeepDock. Using this approach, we identified potent AXL inhibitors from our database. Notably, compound 9, with an IC50 of 9.378 nM, showed excellent inhibitory activity, suggesting its potential as a candidate for further research. We also performed molecular dynamics simulations to explore the interactions between compound 9 and AXL, providing insights for future enhancements. This hybrid screening method proves effective in finding promising AXL inhibitors, and advancing the development of new cancer therapies.
Collapse
Affiliation(s)
- Xinting Lv
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Youkun Kang
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Xinglong Chi
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Jingyi Zhao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhichao Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaojun Ying
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
| | - Long Li
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
| | - Youlu Pan
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Wenhai Huang
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Linjun Wang
- Affiliated Yongkang First People's Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, P.R. China
| |
Collapse
|
21
|
Hansel‐Harris AT, Tillack AF, Santos‐Martins D, Holcomb M, Forli S. Docking guidance with experimental ligand structural density improves docking pose prediction and virtual screening performance. Protein Sci 2025; 34:e70082. [PMID: 39998966 PMCID: PMC11854350 DOI: 10.1002/pro.70082] [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: 10/31/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
Recent advances in structural biology have led to the publication of a wealth of high-resolution x-ray crystallography (XRC) and cryo-EM macromolecule structures, including many complexes with small molecules of interest for drug design. While it is common to incorporate information from the atomic coordinates of these complexes into docking (e.g., pharmacophore models or scaffold hopping), there are limited methods to directly leverage the underlying density information. This is desirable because it does not rely on the determination of relevant coordinates, which may require expert intervention, but instead interprets all density as indicative of regions to which a ligand may be bound. To do so, we have developed CryoXKit, a tool to incorporate experimental densities from either cryo-EM or XRC as a biasing potential on heavy atoms during docking. Using this structural density guidance with AutoDock-GPU, we found significant improvements in re-docking and cross-docking, important pose prediction tasks, compared with the unmodified AutoDock4 force field. Failures in cross-docking tasks are additionally reflective of changes in the positioning of pharmacophores in the site, suggesting it is a fundamental limitation of transferring information between complexes. We additionally found, against a set of targets selected from the LIT-PCBA dataset, that rescoring of these improved poses leads to better discriminatory power in virtual screenings for selected targets. Overall, CryoXKit provides a user-friendly method for improving docking performance with experimental data while requiring no a priori pharmacophore definition and at virtually no computational expense. Map-modification code available at: https://github.com/forlilab/CryoXKit.
Collapse
Affiliation(s)
- Althea T. Hansel‐Harris
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Andreas F. Tillack
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Diogo Santos‐Martins
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Matthew Holcomb
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| | - Stefano Forli
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
| |
Collapse
|
22
|
Li MY, Deng K, Cheng XH, Siu LYL, Gao ZR, Naik TS, Stancheva VG, Cheung PPH, Teo QW, van Leur SW, Wong HH, Lan Y, Lam TTY, Sun MX, Zhang NN, Zhang Y, Cao TS, Yang F, Deng YQ, Sanyal S, Qin CF. ARF4-mediated intracellular transport as a broad-spectrum antiviral target. Nat Microbiol 2025; 10:710-723. [PMID: 39972062 DOI: 10.1038/s41564-025-01940-w] [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: 03/05/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025]
Abstract
Host factors that are involved in modulating cellular vesicular trafficking of virus progeny could be potential antiviral drug targets. ADP-ribosylation factors (ARFs) are GTPases that regulate intracellular vesicular transport upon GTP binding. Here we demonstrate that genetic depletion of ARF4 suppresses viral infection by multiple pathogenic RNA viruses including Zika virus (ZIKV), influenza A virus (IAV) and SARS-CoV-2. Viral infection leads to ARF4 activation and virus production is rescued upon complementation with active ARF4, but not with inactive mutants. Mechanistically, ARF4 deletion disrupts translocation of virus progeny into the Golgi complex and redirects them for lysosomal degradation, thereby blocking virus release. More importantly, peptides targeting ARF4 show therapeutic efficacy against ZIKV and IAV challenge in mice by inhibiting ARF4 activation. Our findings highlight the role of ARF4 during viral infection and its potential as a broad-spectrum antiviral target for further development.
Collapse
Affiliation(s)
- Ming-Yuan Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kao Deng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-He Cheng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Lewis Yu-Lam Siu
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhuo-Ran Gao
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Trupti Shivaprasad Naik
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Peter Pak-Hang Cheung
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi-Wen Teo
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie W van Leur
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Ho-Him Wong
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yun Lan
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Tommy Tsan-Yuk Lam
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Meng-Xu Sun
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Na-Na Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yue Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Tian-Shu Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Fan Yang
- Institute of Pathogenic Biology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China.
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
23
|
Oestreich M, Merdivan E, Lee M, Schultze JL, Piraud M, Becker M. DrugDiff: small molecule diffusion model with flexible guidance towards molecular properties. J Cheminform 2025; 17:23. [PMID: 40001177 PMCID: PMC11854002 DOI: 10.1186/s13321-025-00965-x] [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: 10/24/2024] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
With the cost/yield-ratio of drug development becoming increasingly unfavourable, recent work has explored machine learning to accelerate early stages of the development process. Given the current success of deep generative models across domains, we here investigated their application to the property-based proposal of new small molecules for drug development. Specifically, we trained a latent diffusion model-DrugDiff-paired with predictor guidance to generate novel compounds with a variety of desired molecular properties. The architecture was designed to be highly flexible and easily adaptable to future scenarios. Our experiments showed successful generation of unique, diverse and novel small molecules with targeted properties. The code is available at https://github.com/MarieOestreich/DrugDiff . SCIENTIFIC CONTRIBUTION: This work expands the use of generative modelling in the field of drug development from previously introduced models for proteins and RNA to the here presented application to small molecules. With small molecules making up the majority of drugs, but simultaneously being difficult to model due to their elaborate chemical rules, this work tackles a new level of difficulty in comparison to sequence-based molecule generation as is the case for proteins and RNA. Additionally, the demonstrated framework is highly flexible, allowing easy addition or removal of considered molecular properties without the need to retrain the model, making it highly adaptable to diverse research settings and it shows compelling performance for a wide variety of targeted molecular properties.
Collapse
Affiliation(s)
- Marie Oestreich
- Modular High-Performance Computing and Artificial Intelligence, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Erinc Merdivan
- Helmholtz AI, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Michael Lee
- Modular High-Performance Computing and Artificial Intelligence, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Joachim L Schultze
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany
| | - Marie Piraud
- Helmholtz AI, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Matthias Becker
- Modular High-Performance Computing and Artificial Intelligence, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
24
|
Sierra-Hernandez O, Saurith-Coronell O, Rodríguez-Macías J, Márquez E, Mora JR, Paz JL, Flores-Sumoza M, Mendoza-Mendoza A, Flores-Morales V, Marrero-Ponce Y, Barigye SJ, Martinez-Rios F. In Silico Identification of Potential Clovibactin-like Antibiotics Binding to Unique Cell Wall Precursors in Diverse Gram-Positive Bacterial Strains. Int J Mol Sci 2025; 26:1724. [PMID: 40004190 PMCID: PMC11855776 DOI: 10.3390/ijms26041724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The rise in multidrug-resistant bacteria highlights the critical need for novel antibiotics. This study explores clovibactin-like compounds as potential therapeutic agents targeting lipid II, a crucial component in bacterial cell wall synthesis, using in silico techniques. A total of 2624 clovibactin analogs were sourced from the PubChem database and screened using ProTox 3.0 software based on their ADME-Tox properties, prioritizing candidates with favorable pharmacokinetic profiles and minimal toxicity. Molecular docking protocols were then employed to assess the binding interactions of the selected compounds with lipid II. Our analysis identified Compound 22 as a particularly promising candidate, exhibiting strong binding affinity, stable complex formation, and high selectivity for the target. Binding energy analysis, conducted via molecular dynamics simulations, revealed a highly negative value of -25.50 kcal/mol for Compound 22, surpassing that of clovibactin and underscoring its potential efficacy. In addition, Compound 22 was prioritized due to its exceptional binding affinity to lipid II and its favorable ADME-Tox properties, suggesting a lower likelihood of adverse effects. These characteristics position Compound 22 as a promising candidate for further pharmacological development. While our computational results are encouraging, experimental validation is essential to confirm the efficacy and safety of these compounds. This study not only advances our understanding of clovibactin analogs but also contributes to the ongoing efforts to combat antimicrobial resistance through innovative antibiotic development.
Collapse
Affiliation(s)
- Olimpo Sierra-Hernandez
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Km 5, Vía Puerto Colombia, Puerto Colombia 081007, Colombia; (O.S.-H.); (O.S.-C.)
- Grupo de Investigaciones en Química y Biología, Departamento de Química y Biología, Facultad de Ciencias Básicas, Universidad del Norte, Carrera 51B, Km 5, Vía Puerto Colombia, Barranquilla 081007, Colombia
| | - Oscar Saurith-Coronell
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Km 5, Vía Puerto Colombia, Puerto Colombia 081007, Colombia; (O.S.-H.); (O.S.-C.)
- Grupo de Investigaciones en Química y Biología, Departamento de Química y Biología, Facultad de Ciencias Básicas, Universidad del Norte, Carrera 51B, Km 5, Vía Puerto Colombia, Barranquilla 081007, Colombia
| | - Juan Rodríguez-Macías
- Facultad de Ciencias de la Salud, Exactas y Naturales, Universidad Libre, Barranquilla 080001, Colombia;
| | - Edgar Márquez
- Grupo de Investigaciones en Química y Biología, Departamento de Química y Biología, Facultad de Ciencias Básicas, Universidad del Norte, Carrera 51B, Km 5, Vía Puerto Colombia, Barranquilla 081007, Colombia
| | - José Ramón Mora
- Grupo de Química Computacional y Teórica (QCT-USFQ), Departamento de Ingeniería Química, Universidad San Francisco de Quito, Diego de Robles y Vía Interoceánica, Quito 170901, Ecuador;
| | - José L. Paz
- Departamento Académico de Química Inorgánica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru;
| | - Maryury Flores-Sumoza
- Programa de Química y Farmacia, Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Carrera 59 N° 59-65, Barranquilla 080002, Colombia;
| | - Adel Mendoza-Mendoza
- Programa de Ingeniería Industrial, Universidad del Atlántico, Barranquilla 080001, Colombia;
| | - Virginia Flores-Morales
- Laboratorio de Síntesis Asimétrica y Bioenergética (LSAyB), Ingeniería Química (UACQ), Universidad Autónoma de Zacatecas, Campus XXI Km 6 Carr. Zac-Gdl, Zacatecas 98160, Mexico;
| | - Yovani Marrero-Ponce
- Facultad de Ingeniería, Universidad Panamericana, Augusto Rodin No. 498, Insurgentes Mixcoac, Benito Juárez, Ciudad de México 03920, Mexico; (Y.M.-P.); (F.M.-R.)
- Grupo de Medicina Molecular y Traslacional (MeM&T), Colegio de Ciencias de la Salud (COCSA), Escuela de Medicina, Edificio de Especialidades Médicas, Diego de Robles y Vía Interoceánica, Universidad San Francisco de Quito (USFQ), Quito 170157, Ecuador
| | - Stephen J. Barigye
- Departamento de Química Física Aplicada, Facultad de Ciencias, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain;
| | - Felix Martinez-Rios
- Facultad de Ingeniería, Universidad Panamericana, Augusto Rodin No. 498, Insurgentes Mixcoac, Benito Juárez, Ciudad de México 03920, Mexico; (Y.M.-P.); (F.M.-R.)
| |
Collapse
|
25
|
Ivković Đ, Andrić F, Senćanski M, Stević T, Krstić Ristivojević M, Ristivojević P. Innovative analytical methodology for skin anti-aging compounds discovery from plant extracts: Integration of High-Performance Thin-Layer Chromatography-in vitro spectrophotometry bioassays with multivariate modeling and molecular docking. J Chromatogr A 2025; 1742:465640. [PMID: 39752893 DOI: 10.1016/j.chroma.2024.465640] [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: 09/20/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025]
Abstract
Skin aging, characterized by reduced elasticity, wrinkles, and changes in pigmentation, presents significant challenges in the cosmetics industry. Identifying compounds that can help mitigate these effects is crucial to developing effective anti-aging treatments and improving skin health. An advanced analytical approach for identifying skin anti-aging compounds within complex natural mixtures must be developed to achieve this. This study introduces a state-of-the-art methodology that combines High-Performance Thin-Layer Chromatography (HPTLC) and in vitro skin anti-aging spectrophotometry bioassays with regression multivariate analysis and molecular docking. The proposed methodology integrates spectrophotometric assays for tyrosinase inhibition (anti-pigmentation), elastase inhibition (anti-wrinkle), and radical scavenging capacity (DPPH•/ ABTS• assays) with analytical signals obtained from HPTLC chromatograms using Partial Least Squares models (PLS). The PLS models for predicting elastase inhibition and antioxidative capacity showed high accuracy with minimal errors. This study introduces an innovative approach combining HPTLC profiling and PLS regression to identify single phenolic compounds/bands responsible for anti-aging effects. In addition, identified bioactives were submitted to molecular docking studies to elucidate the enzyme inhibition mechanisms of elastase and confirm our approach. This integrated, simple, cost-effective, and high-throughput approach represents a significant advancement in the discovery of anti-aging compounds, with promising implications for future skincare and therapeutic applications.
Collapse
Affiliation(s)
- Đurđa Ivković
- Innovative Centre of Faculty of Chemistry Ltd., Studentski Trg 12-16, 11158 Belgrade, Serbia.
| | - Filip Andrić
- Department of Analytical Chemistry, University of Belgrade-Faculty of Chemistry, Studentski trg 12-16, 11158 Belgrade, Serbia
| | - Milan Senćanski
- Laboratory of Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences Vinca, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
| | - Tatjana Stević
- Institute for Medicinal Plants Research "Dr. Josif Pancic", Tadeuša Košćuška 1, 11000 Belgrade, Serbia
| | - Maja Krstić Ristivojević
- Department of Biochemistry, University of Belgrade-Faculty of Chemistry, Studentski trg 12-16, 11158 Belgrade, Serbia
| | - Petar Ristivojević
- Department of Analytical Chemistry, University of Belgrade-Faculty of Chemistry, Studentski trg 12-16, 11158 Belgrade, Serbia.
| |
Collapse
|
26
|
Nie H, Zhang S, Wang L, Wang M, Qiu J, Jia F, Li X, Tian G, An B. Synthesis of novel deuterated EGFR/ALK dual-target inhibitors and their activity against non-small cell lung cancer. Eur J Med Chem 2025; 283:117146. [PMID: 39657459 DOI: 10.1016/j.ejmech.2024.117146] [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: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
EGFR and ALK are common driver genes in NSCLC, and more patients with these mutations are being identified due to medical advances. Thus, developing dual-target EGFR/ALK inhibitors is crucial. In this study, 10 novel small molecules were designed and synthesized. CCK8 experiments revealed that compound (-)-9a exhibited the best anti-tumor activity, with IC50 values of 1.08 ± 0.07 nM for EGFR and 2.395 ± 0.023 nM for ALK mutant tumor cells. Studies show that compound (-)-9a can inhibit phosphorylated proteins in EGFR, ALK, and BRK signaling pathways and halt the cell cycle, leading to reduced mitochondrial membrane potential and apoptosis in tumor cells. Additionally, (-)-9a not only directly targets tumor cells but also exhibits potential immune-enhancing effects. Furthermore, evaluations conducted in animal models have demonstrated that this drug effectively reduces tumor growth in vivo. In summary, (-)-9a boasts dual-targeting, potent antitumor activity, and immune-enhancing potential, presenting vast potential as a next-gen anticancer drug.
Collapse
Affiliation(s)
- Haoran Nie
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China
| | - Shuai Zhang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China
| | - Lihan Wang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China
| | - Mengxuan Wang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China
| | - Jiaqi Qiu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China
| | - Fangyi Jia
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China
| | - Baijiao An
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, PR China.
| |
Collapse
|
27
|
Shen Z, Chen R, Gao J, Chi X, Zhang Q, Bian Q, Zhou B, Che J, Dai H, Dong X. EvaluationMaster: A GUI Tool for Structure-Based Virtual Screening Evaluation Analysis and Decision-Making Support. J Chem Inf Model 2025; 65:7-14. [PMID: 39692527 DOI: 10.1021/acs.jcim.4c01818] [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: 12/19/2024]
Abstract
Structure-based virtual screening (SBVS) plays an indispensable role in the early phases of drug discovery, utilizing computational docking techniques to predict interactions between molecules and biological targets. During the SBVS process, selecting appropriate target structures and screening algorithms is crucial, as these choices significantly shape the outcomes. Typically, such selections require researchers to be proficient with multiple algorithms and familiar with evaluation and analysis processes, complicating their tasks. These algorithms' lack of graphical user interfaces (GUIs) further complicates it. To address these challenges, we introduced EvaluationMaster, the first GUI tool designed specifically to streamline and standardize the evaluation and decision-making processes in SBVS. It supports four docking algorithms' evaluation under multiple target structures and offers a comprehensive platform that manages the entire workflow─including the downloading of molecules, construction of decoy datasets, prediction of protein pockets, batch docking, and extensive data analysis. By automating complex evaluation tasks and providing clear visualizations of analysis results, EvaluationMaster significantly reduces the learning curve for researchers and boosts the efficiency of evaluations, potentially improving SBVS hit rates and accelerating the discovery and development of new therapeutic agents.
Collapse
Affiliation(s)
- Zheyuan Shen
- College of Pharmaceutical Sciences, Zhejiang University, HangzhouZhejiang310058, China
| | - Roufen Chen
- College of Pharmaceutical Sciences, Zhejiang University, HangzhouZhejiang310058, China
| | - Jian Gao
- College of Pharmaceutical Sciences, Zhejiang University, HangzhouZhejiang310058, China
| | - Xinglong Chi
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, HangzhouZhejiang310058, China
| | - Qingnan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, HangzhouZhejiang310058, China
| | - Qingyu Bian
- College of Pharmaceutical Sciences, Zhejiang University, HangzhouZhejiang310058, China
| | - Binbin Zhou
- Department of Computer Science and Computing, Zhejiang University City College, HangzhouZhejiang310058, China
| | - Jinxin Che
- College of Pharmaceutical Sciences, Zhejiang University, HangzhouZhejiang310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, HangzhouZhejiang310058, China
| | - Haibin Dai
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, HangzhouZhejiang310058, China
| | - Xiaowu Dong
- College of Pharmaceutical Sciences, Zhejiang University, HangzhouZhejiang310058, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, HangzhouZhejiang310058, China
| |
Collapse
|
28
|
Mozaffari S, Moen A, Ng CY, Nicolaes GA, Wichapong K. Structural bioinformatics for rational drug design. Res Pract Thromb Haemost 2025; 9:102691. [PMID: 40027444 PMCID: PMC11869865 DOI: 10.1016/j.rpth.2025.102691] [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: 10/24/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 03/05/2025] Open
Abstract
A State of the Art lecture titled "structural bioinformatics technologies for rational drug design: from in silico to in vivo" was presented at the International Society on Thrombosis and Haemostasis (ISTH) Congress in 2024. Drug discovery remains a resource-intensive and complex endeavor, which usually takes over a decade and costs billions to bring a new therapeutic agent to market. However, the landscape of drug discovery has been transformed by the recent advancements in bioinformatics and cheminformatics. Key techniques, including structure- and ligand-based virtual screening, molecular dynamics simulations, and artificial intelligence-driven models are allowing researchers to explore vast chemical spaces, investigate molecular interactions, predict binding affinity, and optimize drug candidates with unprecedented accuracy and efficiency. These computational methods complement experimental techniques by accelerating the identification of viable drug candidates and refining lead compounds. Artificial intelligence models, alongside traditional physics-based simulations, now play an important role in predicting key properties such as binding affinity and toxicity, contributing to more informed decision-making, particularly early in the drug discovery process. Despite these advancements, challenges remain in terms of accuracy, interpretability, and the needed computational power. This review explores the state of the art in computational drug discovery, examining the latest methods and technologies, their transformative impact on the drug development pipeline, and the future directions needed to overcome remaining limitations. Finally, we summarize relevant data and highlight cases where various computational approaches were successfully applied to develop novel inhibitors, as presented during the ISTH 2024 Congress.
Collapse
Affiliation(s)
- Soroush Mozaffari
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Agnethe Moen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Che Yee Ng
- Hillmark B.V., Maastricht, the Netherlands
| | - Gerry A.F. Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Hillmark B.V., Maastricht, the Netherlands
| | - Kanin Wichapong
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Hillmark B.V., Maastricht, the Netherlands
| |
Collapse
|
29
|
Corrêa Veríssimo G, Salgado Ferreira R, Gonçalves Maltarollo V. Ultra-Large Virtual Screening: Definition, Recent Advances, and Challenges in Drug Design. Mol Inform 2025; 44:e202400305. [PMID: 39635776 DOI: 10.1002/minf.202400305] [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: 10/15/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
Virtual screening (VS) in drug design employs computational methodologies to systematically rank molecules from a virtual compound library based on predicted features related to their biological activities or chemical properties. The recent expansion in commercially accessible compound libraries and the advancements in artificial intelligence (AI) and computational power - including enhanced central processing units (CPUs), graphics processing units (GPUs), high-performance computing (HPC), and cloud computing - have significantly expanded our capacity to screen libraries containing over 109 molecules. Herein, we review the concept of ultra-large virtual screening (ULVS), focusing on the various algorithms and methodologies employed for virtual screening at this scale. In this context, we present the software utilized, applications, and results of different approaches, such as brute force docking, reaction-based docking approaches, machine learning (ML) strategies applied to docking or other VS methods, and similarity/pharmacophore search-based techniques. These examples represent a paradigm shift in the drug discovery process, demonstrating not only the feasibility of billion-scale compound screening but also their potential to identify hit candidates and increase the structural diversity of novel compounds with biological activities.
Collapse
Affiliation(s)
- Gabriel Corrêa Veríssimo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
- Programa de Pós-Graduação em Bioinformática, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Vinícius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| |
Collapse
|
30
|
Poggetti V, Angeloni E, Germelli L, Natale B, Waqas M, Sarno G, Angeli A, Daniele S, Salerno S, Barresi E, Cosconati S, Castellano S, Da Pozzo E, Costa B, Supuran CT, Da Settimo F, Taliani S. Discovery of the First-in-Class Dual TSPO/Carbonic Anhydrase Modulators with Promising Neurotrophic Activity. ACS Chem Neurosci 2025; 16:1-15. [PMID: 39545683 DOI: 10.1021/acschemneuro.4c00477] [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] [Indexed: 11/17/2024] Open
Abstract
In searching for putative new therapeutic strategies to treat neurodegenerative diseases, the mitochondrial 18 kDa translocator protein (TSPO) and cerebral isoforms of carbonic anhydrase (CA) were exploited as potential targets. Based on the structures of a class of highly affine and selective TSPO ligands and a class of CA activators, both developed by us in recent years, a small library of 2-phenylindole-based dual TSPO/CA modulators was developed, able to bind TSPO and activate CA VII in the low micromolar/submicromolar range. The interaction with the two targets was corroborated by computational studies. Biological investigation on human microglia C20 cells identified derivative 3 as a promising lead compound worthy of future optimization due to its (i) lack of cytotoxicity, (ii) ability to stimulate TSPO steroidogenic function and activate CA VII, and (iii) ability to effectively upregulate gene expression of the brain-derived neurotrophic factor.
Collapse
Affiliation(s)
- Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Elisa Angeloni
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Lorenzo Germelli
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Benito Natale
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100 Caserta, Italy
| | - Muhammad Waqas
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100 Caserta, Italy
| | - Giuliana Sarno
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Andrea Angeli
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, Polo Scientifico, University of Florence, Via U. Schiff, 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Sandro Cosconati
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100 Caserta, Italy
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Eleonora Da Pozzo
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, Polo Scientifico, University of Florence, Via U. Schiff, 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| |
Collapse
|
31
|
Kim DN, Yin T, Zhang T, Im AK, Cort JR, Rozum JC, Pollock D, Qian WJ, Feng S. Artificial Intelligence Transforming Post-Translational Modification Research. Bioengineering (Basel) 2024; 12:26. [PMID: 39851300 PMCID: PMC11762806 DOI: 10.3390/bioengineering12010026] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/29/2024] [Indexed: 01/26/2025] Open
Abstract
Post-Translational Modifications (PTMs) are covalent changes to amino acids that occur after protein synthesis, including covalent modifications on side chains and peptide backbones. Many PTMs profoundly impact cellular and molecular functions and structures, and their significance extends to evolutionary studies as well. In light of these implications, we have explored how artificial intelligence (AI) can be utilized in researching PTMs. Initially, rationales for adopting AI and its advantages in understanding the functions of PTMs are discussed. Then, various deep learning architectures and programs, including recent applications of language models, for predicting PTM sites on proteins and the regulatory functions of these PTMs are compared. Finally, our high-throughput PTM-data-generation pipeline, which formats data suitably for AI training and predictions is described. We hope this review illuminates areas where future AI models on PTMs can be improved, thereby contributing to the field of PTM bioengineering.
Collapse
Affiliation(s)
- Doo Nam Kim
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
| | - Tianzhixi Yin
- National Security Directorate, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA
| | - Tong Zhang
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
| | - Alexandria K. Im
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
| | - John R. Cort
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
| | - Jordan C. Rozum
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
| | - David Pollock
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
| | - Song Feng
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA (J.C.R.); (D.P.); (W.-J.Q.)
| |
Collapse
|
32
|
Gawande V, Kushwaha R, Mandal AA, Banerjee S. Targeting SARS-CoV-2 Proteins: In Silico Investigation with Polypyridyl-Based Zn(II)-Curcumin Complexes. Chembiochem 2024; 25:e202400612. [PMID: 39264259 DOI: 10.1002/cbic.202400612] [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: 07/21/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/13/2024]
Abstract
Herein, we have selected eight Zn(II)-based complexes viz., [Zn(bpy)(acac)Cl] (1), [Zn(phen)(acac)Cl] (2), [Zn(dppz)(acac)Cl] (3), [Zn(dppn)(acac)Cl] (4), [Zn(bpy)(cur)Cl] (5), [Zn(phen)(cur)Cl] (6), [Zn(dppz)(cur)Cl] (7), [Zn(dppn)(cur)Cl] (8), where bpy=2,2'-bipyridine, phen=1,10-phenanthroline, dppz=benzo[i]dipyrido[3,2-a:2',3'-c]phenazine, dppn=naphtho[2,3-i]dipyrido[3,2-a:2',3'-c]phenazine, acac=acetylacetonate, cur=curcumin and performed in silico molecular docking studies with the viral proteins, i. e., spike protein (S), Angiotensin-converting enzyme II Receptor protein (ACE2), nucleocapsid protein (N), main protease protein (Mpro), and RNA-dependent RNA polymerase protein (RdRp) of SARS-CoV-2. The binding energy calculations, visualization of the docking orientation, and analysis of the interactions revealed that these complexes could be potential inhibitors of the viral proteins. Among complexes 1-8, complex 6 showed the strongest binding affinity with S and ACE2 proteins. 4 exerted better binding affinity in the case of the N protein, whereas 8 presented the highest binding affinities with Mpro and RdRp among all the complexes. Overall, the study indicated that Zn(II) complexes have the potential as alternative and viable therapeutic solutions for COVID-19.
Collapse
Affiliation(s)
- Vedant Gawande
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
33
|
Mihalovits L, Szalai TV, Bajusz D, Keserű GM. Exploring Chemical Spaces in the Billion Range: Is Docking a Computational Alternative to DNA-Encoded Libraries? J Chem Inf Model 2024; 64:8963-8979. [PMID: 39305268 PMCID: PMC11632764 DOI: 10.1021/acs.jcim.4c00803] [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: 05/10/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 12/10/2024]
Abstract
The concept of DNA-encoded libraries (DELs) enables the experimental screening of billions of compounds simultaneously, offering an unprecedented boost in the coverage of chemical space. In parallel, however, dramatically increased access to supercomputers and a number of ultrahigh throughput virtual screening (uHTVS) tools have made screening of billion-membered virtual libraries available. Here, we investigate whether current, brute-force, or AI-enabled uHTVS approaches might constitute a computational alternative to DEL screening. While it is tempting to look at uHTVS as a computational analogue of DEL screening, we found specific advantages and limitations of both methodologies that suggest them being complementary rather than competitive.
Collapse
Affiliation(s)
- Levente
M. Mihalovits
- Medicinal
Chemistry Research Group and Drug Innovation Centre, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117 Budapest, Hungary
| | - Tibor V. Szalai
- Medicinal
Chemistry Research Group and Drug Innovation Centre, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117 Budapest, Hungary
- Department
of Inorganic and Analytical Chemistry, Faculty of Chemical Technology
and Biotechnology, Budapest University of
Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Dávid Bajusz
- Medicinal
Chemistry Research Group and Drug Innovation Centre, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117 Budapest, Hungary
| | - György M. Keserű
- Medicinal
Chemistry Research Group and Drug Innovation Centre, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117 Budapest, Hungary
- Department
of Organic Chemistry and Technology, Faculty of Chemical Technology
and Biotechnology Budapest University of
Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| |
Collapse
|
34
|
Dong C, Huang YP, Lin X, Zhang H, Gao YQ. DSDPFlex: Flexible-Receptor Docking with GPU Acceleration. J Chem Inf Model 2024; 64:8537-8548. [PMID: 39514506 DOI: 10.1021/acs.jcim.4c01715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Molecular docking is an essential tool in structure-based drug discovery, widely utilized to model ligand-protein interactions and enrich potential hits. Among the different docking strategies, semiflexible docking (rigid-receptor and flexible-ligand model) is the most popular, benefiting from its balance of docking accuracy and speed. However, this approach ignores the conformational changes of proteins and hence demands suitable protein conformations as input. When the binding interaction adheres to an induced-fit model, flexible methods such as molecular dynamics simulation can be utilized, but they are computationally demanding. To balance between speed and accuracy, the flexible docking approach is an effective choice, as exemplified by AutoDock Vina and AutoDockFR, which treat selected protein side chains as flexible parts. However, the efficiency of flexible docking methods is yet to be improved for virtual screening usage. In this article, we introduce DSDPFlex, an improved flexible-receptor docking method accelerated by GPU parallelization. Beyond acceleration, optimizations with respect to sampling, scoring, and search space are implemented in DSDPFlex to further improve its capability in flexible tasks. In cross-docking evaluation, DSDPFlex demonstrates superior accuracy compared to AutoDock Vina and is 100 times faster than Vina in flexible-receptor tasks. We also show the advantage of flexible-receptor methods on suboptimal pockets and validate the advantage of DSDPFlex in screening on apo and AlphaFold2-predicted structures. With improvements in both efficiency and accuracy, DSDPFlex is expected to hold potential in future docking-based studies.
Collapse
Affiliation(s)
- Chengwei Dong
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yu-Peng Huang
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xiaohan Lin
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Hong Zhang
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102200, China
| | - Yi Qin Gao
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102200, China
- Biomedical Pioneering Innovation Center, Peking University, Beijing 100871, China
| |
Collapse
|
35
|
Schwalm MP, Dopfer J, Kumar A, Greco FA, Bauer N, Löhr F, Heering J, Cano-Franco S, Lechner S, Hanke T, Jaser I, Morasch V, Lenz C, Fearon D, Marples PG, Tomlinson CWE, Brunello L, Saxena K, Adams NBP, von Delft F, Müller S, Stolz A, Proschak E, Kuster B, Knapp S, Rogov VV. Critical assessment of LC3/GABARAP ligands used for degrader development and ligandability of LC3/GABARAP binding pockets. Nat Commun 2024; 15:10204. [PMID: 39587067 PMCID: PMC11589570 DOI: 10.1038/s41467-024-54409-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/08/2024] [Indexed: 11/27/2024] Open
Abstract
Recent successes in developing small molecule degraders that act through the ubiquitin system have spurred efforts to extend this technology to other mechanisms, including the autophagosomal-lysosomal pathway. Therefore, reports of autophagosome tethering compounds (ATTECs) have received considerable attention from the drug development community. ATTECs are based on the recruitment of targets to LC3/GABARAP, a family of ubiquitin-like proteins that presumably bind to the autophagosome membrane and tether cargo-loaded autophagy receptors into the autophagosome. In this work, we rigorously tested the target engagement of the reported ATTECs to validate the existing LC3/GABARAP ligands. Surprisingly, we were unable to detect interaction with their designated target LC3 using a diversity of biophysical methods. Intrigued by the idea of developing ATTECs, we evaluated the ligandability of LC3/GABARAP by in silico docking and large-scale crystallographic fragment screening. Data based on approximately 1000 crystal structures revealed that most fragments bound to the HP2 but not to the HP1 pocket within the LIR docking site, suggesting a favorable ligandability of HP2. Through this study, we identified diverse validated LC3/GABARAP ligands and fragments as starting points for chemical probe and ATTEC development.
Collapse
Affiliation(s)
- Martin P Schwalm
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
- German Cancer Consortium (DKTK) / German Cancer Research Center (DKFZ), DKTK site Frankfurt-Mainz, 69120, Heidelberg, Germany
| | - Johannes Dopfer
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Adarsh Kumar
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Francesco A Greco
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Nicolas Bauer
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Frank Löhr
- Institute for Biophysical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt, Germany
| | - Sara Cano-Franco
- Institute of Biochemistry II (IBC2), Faculty of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Max-von-Laue-Straße 15, 60438, Frankfurt am Main, Germany
| | - Severin Lechner
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Thomas Hanke
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Ivana Jaser
- NanoTemper Technologies GmbH, Flößergasse 4, 81369, Munich, Germany
| | - Viktoria Morasch
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Christopher Lenz
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Daren Fearon
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0QX, UK
| | - Peter G Marples
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0QX, UK
| | - Charles W E Tomlinson
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0QX, UK
| | - Lorene Brunello
- Institute of Biochemistry II (IBC2), Faculty of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Max-von-Laue-Straße 15, 60438, Frankfurt am Main, Germany
| | - Krishna Saxena
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Nathan B P Adams
- NanoTemper Technologies GmbH, Flößergasse 4, 81369, Munich, Germany
| | - Frank von Delft
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0QX, UK
| | - Susanne Müller
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany
| | - Alexandra Stolz
- Institute of Biochemistry II (IBC2), Faculty of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Max-von-Laue-Straße 15, 60438, Frankfurt am Main, Germany
| | - Ewgenij Proschak
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany.
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany.
- German Cancer Consortium (DKTK) / German Cancer Research Center (DKFZ), DKTK site Frankfurt-Mainz, 69120, Heidelberg, Germany.
| | - Vladimir V Rogov
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany.
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438, Frankfurt, Germany.
| |
Collapse
|
36
|
Patra J, Arora S, Debnath U, Mahindroo N. In silico studies for improving target selectivity of anti-malarial dual falcipain inhibitors vis-à-vis human cathepsins. J Biomol Struct Dyn 2024:1-20. [PMID: 39552300 DOI: 10.1080/07391102.2024.2427372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/21/2024] [Indexed: 11/19/2024]
Abstract
Dual falcipain-2 (FP-2) and falcipain-3 (FP-3) inhibitors, NM12 and NM15, displayed micromolar inhibitions but they exhibit similar binding affinities for the human cathepsins, thus indicating potential toxicity. The current study aims to develop a model to enhance the selectivity of the falcipain inhibitors vis-à-vis human cathepsins using previously identified dual falcipain 2 and 3 inhibitors, NM12 and NM15. To improve the selectivity of NM12 and NM15, analogs with weaker interactions with the conserved residues in the FPs and hCatK were designed while enhancing the unique interactions for the FPs. In silico analysis was carried out in the S2 subsite of both plasmodium and human proteases which is considered the preferred selective site due to the presence of less conserved residues. The Fasta sequence alignment and active/conserved binding site superimposition show that FPs contain acidic polar residues (Asp234 for FP2 and Glu243 for FP3) while hCatK has a neutral hydrophobic residue (Leu209) at the S2 subsite. Therefore, analogs of NM12 and NM15 were designed to enhance affinity and selectivity by improving interactions with these acidic residues while avoiding interactions with hydrophobic residues in hCatK. Newly designed analogs (NM12H and NM15G) show better selectivity as well as binding affinity towards FPs (ΔG of NM12H: -74.49 kcal/mol for FP2, -70.97 kcal/mol for FP3; ΔG of NM15G: -70.09 kcal/mol for FP2, -74.52 kcal/mol for FP3) as compared to NM12 and NM15. Thus, the selectivity and binding affinity against dual falcipains vis-à-vis human cathepsin were improved using molecular dynamic simulations.
Collapse
Affiliation(s)
- Jeevan Patra
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
| | - Smriti Arora
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
| | - Utsab Debnath
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
| | - Neeraj Mahindroo
- School of Health Sciences and Technology, UPES, Energy Acres, Bidholi, India
- School of Health Sciences and Technology, Vishwanath Karad MIT World Peace University, Kothrud, Pune, India
| |
Collapse
|
37
|
Moesgaard L, Kongsted J. Introducing SpaceGA: A Search Tool to Accelerate Large Virtual Screenings of Combinatorial Libraries. J Chem Inf Model 2024; 64:8123-8130. [PMID: 39475501 DOI: 10.1021/acs.jcim.4c01308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
The growth of make-on-demand libraries in recent years has provided completely new possibilities for virtual screening for discovering new hit compounds with specific and favorable properties. However, since these libraries now contain billions of compounds, screening them using traditional methods such as molecular docking has become challenging and requires substantial computational resources. Thus, to take real advantage of the new possibilities introduced by the make-on-demand libraries, different methods have been proposed to accelerate the screening process and prioritize molecules for evaluation. Here, we introduce SpaceGA, a genetic algorithm that leverages the rapid similarity search tool SpaceLight (Bellmann, L.; Penner, P.; Rarey, M. Topological similarity search in large combinatorial fragment spaces. J. Chem. Inf. Model. 2021, 61, 238-251). to constrain the optimization process to accessible compounds within desired combinatorial libraries. As shown herein, SpaceGA is able to efficiently identify molecules with desired properties from trillions of synthesizable compounds by enumerating and evaluating only a small fraction of them. On this basis, SpaceGA represents a promising new tool for accelerating and simplifying virtual screens of ultralarge combinatorial databases.
Collapse
Affiliation(s)
- Laust Moesgaard
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense DK-5230, Denmark
| | - Jacob Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense DK-5230, Denmark
| |
Collapse
|
38
|
Roggia M, Natale B, Amendola G, Grasso N, Di Maro S, Taliani S, Castellano S, Reina SCR, Salvati E, Amato J, Cosconati S. Discovering Dually Active Anti-cancer Compounds with a Hybrid AI-structure-based Approach. J Chem Inf Model 2024; 64:8299-8309. [PMID: 39276072 DOI: 10.1021/acs.jcim.4c01132] [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: 09/16/2024]
Abstract
Cancer's persistent growth often relies on its ability to maintain telomere length and tolerate the accumulation of DNA damage. This study explores a computational approach to identify compounds that can simultaneously target both G-quadruplex (G4) structures and poly(ADP-ribose) polymerase (PARP)1 enzyme, offering a potential multipronged attack on cancer cells. We employed a hybrid virtual screening (VS) protocol, combining the power of machine learning with traditional structure-based methods. PyRMD, our AI-powered tool, was first used to analyze vast chemical libraries and to identify potential PARP1 inhibitors based on known bioactivity data. Subsequently, a structure-based VS approach selected compounds from these identified inhibitors for their G4 stabilization potential. This two-step process yielded 50 promising candidates, which were then experimentally validated for their ability to inhibit PARP1 and stabilize G4 structures. Ultimately, four lead compounds emerged as promising candidates with the desired dual activity and demonstrated antiproliferative effects against specific cancer cell lines. This study highlights the potential of combining Artificial Intelligence and structure-based methods for the discovery of multitarget anticancer compounds, offering a valuable approach for future drug development efforts.
Collapse
Affiliation(s)
- Michele Roggia
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Benito Natale
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Giorgio Amendola
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Nicola Grasso
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, Naples 80131, Italy
| | - Salvatore Di Maro
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Sabrina Castellano
- Dipartimento di Farmacia, Università di Salerno, Via Giovanni Paolo II 132, 84084 Fisciano Salerno, Italy
| | | | - Erica Salvati
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, Naples 80131, Italy
| | - Sandro Cosconati
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| |
Collapse
|
39
|
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
|
40
|
Tang S, Ding J, Zhu X, Wang Z, Zhao H, Wu J. Vina-GPU 2.1: Towards Further Optimizing Docking Speed and Precision of AutoDock Vina and Its Derivatives. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2382-2393. [PMID: 39320991 DOI: 10.1109/tcbb.2024.3467127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
AutoDock Vina and its derivatives have established themselves as a prevailing pipeline for virtual screening in contemporary drug discovery. Our Vina-GPU method leverages the parallel computing power of GPUs to accelerate AutoDock Vina, and Vina-GPU 2.0 further enhances the speed of AutoDock Vina and its derivatives. Given the prevalence of large virtual screens in modern drug discovery, the improvement of speed and accuracy in virtual screening has become a longstanding challenge. In this study, we propose Vina-GPU 2.1, aimed at enhancing the docking speed and precision of AutoDock Vina and its derivatives through the integration of novel algorithms to facilitate improved docking and virtual screening outcomes. Building upon the foundations laid by Vina-GPU 2.0, we introduce a novel algorithm, namely Reduced Iteration and Low Complexity BFGS (RILC-BFGS), designed to expedite the most time-consuming operation. Additionally, we implement grid cache optimization to further enhance the docking speed. Furthermore, we employ optimal strategies to individually optimize the structures of ligands, receptors, and binding pockets, thereby enhancing the docking precision. To assess the performance of Vina-GPU 2.1, we conduct extensive virtual screening experiments on three prominent targets, utilizing two fundamental compound libraries and seven docking tools. Our results demonstrate that Vina-GPU 2.1 achieves an average 4.97-fold acceleration in docking speed and an average 342% improvement in EF1% compared to Vina-GPU 2.0.
Collapse
|
41
|
Boya P. CA, Rodriguez C, Mojica-Flores R, Urrutia JC, Cantilo-Diaz V, Barrios-Jaén M, Ng MG, Pineda L, Llanes A, Spadafora C, Mejía LC, Gutiérrez M. Antiprotozoal Natural Products from Endophytic Fungi Associated with Cacao and Coffee. Metabolites 2024; 14:575. [PMID: 39590811 PMCID: PMC11596112 DOI: 10.3390/metabo14110575] [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: 09/20/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Collectively, leishmaniasis and Chagas disease cause approximately 8 million cases and more than 40,000 deaths annually, mostly in tropical and subtropical regions. The current drugs used to treat these diseases have limitations and many undesirable side effects; hence, new drugs with better clinical profiles are needed. Fungal endophytes associated with plants are known to produce a wide array of bioactive secondary metabolites, including antiprotozoal compounds. In this study, we analyzed endophytic fungal isolates associated with Theobroma cacao and Coffea arabica crop plants, which yielded extracts with antitrypanosomatid activity. METHODS Crude extracts were subjected to bioassay-guided isolation by HPLC, followed by spectrometric and spectroscopic analyses via mass spectrometry (MS) and nuclear magnetic resonance (NMR), Results: Compounds 1-9 were isolated and displayed novel antitrypanosomal and antileishmanial activities ranging from 0.92 to 32 μM. Tandem liquid chromatography-mass spectrometry (LC-MS) analysis of the organic extracts from different strains via the feature-based Global Natural Products Social (GNPS) molecular networking platform allowed us to dereplicate a series of metabolites (10-23) in the extracts. Molecular docking simulations of the active compounds, using the 3-mercaptopyruvate sulfurtransferase protein from L. donovani (Ld3MST) and the cruzipain enzyme from T. cruzi as putative molecular targets, allowed us to suggest possible mechanisms for the action of these compounds. CONCLUSIONS The isolation of these antiprotozoal compounds confirms that crop plants like coffee and cacao harbor populations of endophytes with biomedical potential that confer added value to these crops.
Collapse
Affiliation(s)
- Cristopher A. Boya P.
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Candelario Rodriguez
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
- Estación Científica COIBA AIP, Ciudad del Saber, Panamá 0816-02852, Panama
| | - Randy Mojica-Flores
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Jean Carlo Urrutia
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Víctor Cantilo-Diaz
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Masiel Barrios-Jaén
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Michelle G. Ng
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Laura Pineda
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Alejandro Llanes
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Carmenza Spadafora
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Luis C. Mejía
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
- Smithsonian Tropical Research Institute, Ancón 0843-03092, Panama
| | - Marcelino Gutiérrez
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| |
Collapse
|
42
|
Song J, Ha J, Lee J, Ko J, Shin WH. Improving docking and virtual screening performance using AlphaFold2 multi-state modeling for kinases. Sci Rep 2024; 14:25167. [PMID: 39448664 PMCID: PMC11502823 DOI: 10.1038/s41598-024-75400-6] [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: 08/13/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
Structure-based virtual screening (SBVS) is a crucial computational approach in drug discovery, but its performance is sensitive to structural variations. Kinases, which are major drug targets, exemplify this challenge due to active site conformational changes caused by different inhibitor types. Most experimentally determined kinase structures have the DFGin state, potentially biasing SBVS towards type I inhibitors and limiting the discovery of diverse scaffolds. We introduce a multi-state modeling (MSM) protocol for AlphaFold2 (AF2) kinase structures using state-specific templates to address these challenges. Our comprehensive benchmarks evaluate predicted model qualities, binding pose prediction accuracy, and hit compound identification through ensemble SBVS. Results demonstrate that MSM models exhibit comparable or improved structural accuracy compared to standard AF2 models, enhancing pose prediction accuracy and effectively capturing kinase-ligand interactions. In virtual screening experiments, our MSM approach consistently outperforms standard AF2 and AF3 modeling, particularly in identifying diverse hit compounds. This study highlights the potential of MSM in broadening kinase inhibitor discovery by facilitating the identification of chemically diverse inhibitors, offering a promising solution to the structural bias problem in kinase-targeted drug discovery.
Collapse
Affiliation(s)
- Jinung Song
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Junsu Ha
- Arontier Co., Seoul, Republic of Korea
| | - Juyong Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Arontier Co., Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul, Republic of Korea
| | - Junsu Ko
- Arontier Co., Seoul, Republic of Korea
| | - Woong-Hee Shin
- Arontier Co., Seoul, Republic of Korea.
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Aguilera-Durán G, Hernández-Castro S, Loera-García BV, Rivera-Vargas A, Alvarez-Baltazar JM, Cuevas-Flores MDR, Romo-Mancillas A. Ursolic acid interaction with transcription factors BRAF, V600E, and V600K: a computational approach towards new potential melanoma treatments. J Mol Model 2024; 30:373. [PMID: 39387972 DOI: 10.1007/s00894-024-06165-y] [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: 05/21/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
CONTEXT Melanoma is one of the cancers with the highest mortality rate for its ability to metastasize. Several targets have undergone investigation for the development of drugs against this pathology. One of the main targets is the kinase BRAF (RAF, rapidly accelerated fibrosarcoma). The most common mutation in melanoma is BRAFV600E and has been reported in 50-90% of patients with melanoma. Due to the relevance of the BRAFV600E mutation, inhibitors to this kinase have been developed, vemurafenib-OMe and dabrafenib. Ursolic acid (UA) is a pentacyclic triterpene with a privileged structure, the pentacycle scaffold, which allows to have a broad variety of biological activity; the most studied is its anticancer capacity. In this work, we reported the interaction profile of vemurafenib-OMe, dabrafenib, and UA, to define whether UA has binding capacity to BRAFWT, BRAFV600E, and BRAFV600K. Homology modeling of BRAFWT, V600E, and V600K; molecular docking; and molecular dynamics simulations were carried out and interactions and residues relevant to the binding of the inhibitors were obtained. We found that UA, like the inhibitors, presents hydrogen bond interactions, and hydrophobic interactions of van der Waals, and π-stacking with I463, Q530, C532, and F583. The ΔG of ursolic acid in complex with BRAFV600K (- 63.31 kcal/mol) is comparable to the ΔG of the selective inhibitor dabrafenib (- 63.32 kcal/mol) in complex to BRAFV600K and presents a ΔG like vemurafenib-OMe with BRAFWT and V600E. With this information, ursolic acid could be considered as a lead compound for design cycles and to optimize the binding profile and the selectivity towards mutations for the development of new selective inhibitors for BRAFV600E and V600K to new potential melanoma treatments. METHODS The homology modeling calculations were executed on the public servers I-TASSER and ROBETTA, followed by molecular docking calculations using AutoGrid 4.2.6, AutoDockGPU 1.5.3, and AutoDockTools 1.5.6. Molecular dynamics and metadynamics simulations were performed in the Desmond module of the academic version of the Schrödinger-Maestro 2020-4 program, utilizing the OPLS-2005 force field. Ligand-protein interactions were evaluated using Schrödinger-Maestro program, LigPlot + , and PLIP (protein-ligand interaction profiler). Finally, all of the protein figures presented in this article were made in the PyMOL program.
Collapse
Affiliation(s)
- Giovanny Aguilera-Durán
- Laboratorio de Química Cuántica y Modelado Molecular, Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, 98160, Zacatecas, Mexico.
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico.
| | - Stephanie Hernández-Castro
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico
| | - Brenda V Loera-García
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Zona Universitaria, 78210, San Luis Potosí, Mexico
| | - Alex Rivera-Vargas
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico
| | - J M Alvarez-Baltazar
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico
| | - Ma Del Refugio Cuevas-Flores
- Laboratorio de Química Cuántica y Modelado Molecular, Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, 98160, Zacatecas, Mexico
| | - Antonio Romo-Mancillas
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de Las Campanas S/N, 76010, Querétaro, Mexico.
- Grupo de Diseño Asistido Por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, 76010, Querétaro, Mexico.
| |
Collapse
|
44
|
Tomassi S, Natale B, Roggia M, Amato L, De Rosa C, Della Corte CM, Baglini E, Amendola G, Messere A, Di Maro S, Barresi E, Da Settimo F, Trincavelli ML, Ciardiello F, Taliani S, Morgillo F, Cosconati S. Discovery of N-substituted-2-oxoindolin benzoylhydrazines as c-MET/SMO modulators in EGFRi-resistant non-small cell lung cancer. RSC Med Chem 2024; 16:d4md00553h. [PMID: 39512947 PMCID: PMC11539002 DOI: 10.1039/d4md00553h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/29/2024] [Indexed: 11/15/2024] Open
Abstract
Non-small cell lung cancer (NSCLC), the leading cause of cancer-related mortality worldwide, poses a formidable challenge due to its heterogeneity and the emergence of resistance to targeted therapies. While initially effective, first- and third-generation EGFR-tyrosine kinase inhibitors (TKIs) often fail to control disease progression, leaving patients with limited treatment options. To address this unmet medical need, we explored the therapeutic potential of multitargeting agents that simultaneously inhibit two key signalling pathways, the mesenchymal-epithelial transition factor (c-MET) and the G protein-coupled receptor Smoothened (SMO), frequently dysregulated in NSCLC. By employing a combination of in silico drug repurposing and structure-based structure-activity relationship (SAR) studies, we identified and developed novel c-MET/SMO-targeting agents with antiproliferative activity against first- as well as third-generation EGFR-TKI-resistant NSCLC cells suggesting a synergistic effect arising from the simultaneous inhibition of c-MET and SMO.
Collapse
Affiliation(s)
- Stefano Tomassi
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
- Department of Life Science, Health, and Health Professions, LINK Campus University Via del Casale di San Pio V, 44 00165 Rome Italy
| | - Benito Natale
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Michele Roggia
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Luisa Amato
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Caterina De Rosa
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Carminia Maria Della Corte
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Emma Baglini
- CNR IFC, Institute of Clinical Physiology, National Research Council of Italy CNR Research Area, Via G. Moruzzi 1 Pisa 56124 Italy
| | - Giorgio Amendola
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Anna Messere
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Salvatore Di Maro
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | | | | | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | - Floriana Morgillo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Sandro Cosconati
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| |
Collapse
|
45
|
Gil MV, Fernández-Rivera N, Gutiérrez-Díaz G, Parrón-Ballesteros J, Pastor-Vargas C, Betancor D, Nieto C, Cintas P. Antioxidant Activity and Hypoallergenicity of Egg Protein Matrices Containing Polyphenols from Citrus Waste. Antioxidants (Basel) 2024; 13:1154. [PMID: 39456407 PMCID: PMC11504875 DOI: 10.3390/antiox13101154] [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: 08/05/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
This study reports on the interactions of egg proteins, which represent a major health concern in food allergy, with polyphenols obtained from orange and lemon peels. The antioxidant properties of such citrus peel extracts prior to protein binding were evaluated. The resulting edible, and therefore inherently safe, matrices exhibit reduced IgE binding compared to pure proteins in indirect immunological assays (ELISA) using individual sera from patients allergic to ovalbumin and lysozyme. The reduced allergenicity could arise from the interactions with polyphenols, which alter the structure and functionality of the native proteins. It is hypothesized that the anti-inflammatory and antioxidant properties of the polyphenols, described as inhibitors of the allergic response, could add immunomodulatory features to the hypoallergenic complexes. A docking analysis using lysozyme was conducted to scrutinize the nature of the protein-polyphenol interactions. An in silico study unravelled the complexity of binding modes depending on the isoforms considered. Altogether, the presented results validate the antioxidant properties and reduced allergenicity of polyphenol-fortified proteins. Lastly, this study highlights the upgrading of vegetable wastes as a source of natural antioxidants, thus showing the benefits of a circular economy in agri-food science.
Collapse
Affiliation(s)
- María Victoria Gil
- Department of Organic and Inorganic Chemistry, IACYS-Green Chemistry and Sustainable Development Unit, Faculty of Sciences, University of Extremadura, 06006 Badajoz, Spain; (N.F.-R.); (P.C.)
| | - Nuria Fernández-Rivera
- Department of Organic and Inorganic Chemistry, IACYS-Green Chemistry and Sustainable Development Unit, Faculty of Sciences, University of Extremadura, 06006 Badajoz, Spain; (N.F.-R.); (P.C.)
| | - Gloria Gutiérrez-Díaz
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain; (G.G.-D.); (J.P.-B.); (C.P.-V.)
| | - Jorge Parrón-Ballesteros
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain; (G.G.-D.); (J.P.-B.); (C.P.-V.)
| | - Carlos Pastor-Vargas
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain; (G.G.-D.); (J.P.-B.); (C.P.-V.)
| | - Diana Betancor
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | - Carlos Nieto
- Department of Organic Chemistry, Faculty of Chemical Sciences, University of Salamanca, Pl. Caídos s/n, 37008 Salamanca, Spain;
| | - Pedro Cintas
- Department of Organic and Inorganic Chemistry, IACYS-Green Chemistry and Sustainable Development Unit, Faculty of Sciences, University of Extremadura, 06006 Badajoz, Spain; (N.F.-R.); (P.C.)
| |
Collapse
|
46
|
García-Cuesta EM, Martínez P, Selvaraju K, Ulltjärn G, Gómez Pozo AM, D'Agostino G, Gardeta S, Quijada-Freire A, Blanco Gabella P, Roca C, Hoyo DD, Jiménez-Saiz R, García-Rubia A, Soler Palacios B, Lucas P, Ayala-Bueno R, Santander Acerete N, Carrasco Y, Oscar Sorzano C, Martinez A, Campillo NE, Jensen LD, Rodriguez Frade JM, Santiago C, Mellado M. Allosteric modulation of the CXCR4:CXCL12 axis by targeting receptor nanoclustering via the TMV-TMVI domain. eLife 2024; 13:RP93968. [PMID: 39248648 PMCID: PMC11383527 DOI: 10.7554/elife.93968] [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] [Indexed: 09/10/2024] Open
Abstract
CXCR4 is a ubiquitously expressed chemokine receptor that regulates leukocyte trafficking and arrest in both homeostatic and pathological states. It also participates in organogenesis, HIV-1 infection, and tumor development. Despite the potential therapeutic benefit of CXCR4 antagonists, only one, plerixafor (AMD3100), which blocks the ligand-binding site, has reached the clinic. Recent advances in imaging and biophysical techniques have provided a richer understanding of the membrane organization and dynamics of this receptor. Activation of CXCR4 by CXCL12 reduces the number of CXCR4 monomers/dimers at the cell membrane and increases the formation of large nanoclusters, which are largely immobile and are required for correct cell orientation to chemoattractant gradients. Mechanistically, CXCR4 activation involves a structural motif defined by residues in TMV and TMVI. Using this structural motif as a template, we performed in silico molecular modeling followed by in vitro screening of a small compound library to identify negative allosteric modulators of CXCR4 that do not affect CXCL12 binding. We identified AGR1.137, a small molecule that abolishes CXCL12-mediated receptor nanoclustering and dynamics and blocks the ability of cells to sense CXCL12 gradients both in vitro and in vivo while preserving ligand binding and receptor internalization.
Collapse
Affiliation(s)
- Eva M García-Cuesta
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Pablo Martínez
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Karthik Selvaraju
- Division of Diagnostics and Specialist Medicine, Department of Health, Medical and Caring Sciences, Linköping University, Linköping, Sweden
| | - Gabriel Ulltjärn
- Division of Diagnostics and Specialist Medicine, Department of Health, Medical and Caring Sciences, Linköping University, Linköping, Sweden
| | | | - Gianluca D'Agostino
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Sofia Gardeta
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Adriana Quijada-Freire
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | | | - Carlos Roca
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Daniel Del Hoyo
- Biocomputing Unit, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain
| | - Rodrigo Jiménez-Saiz
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute, McMaster University, Hamilton, Canada
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | | | - Blanca Soler Palacios
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Pilar Lucas
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Rosa Ayala-Bueno
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Noelia Santander Acerete
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Yolanda Carrasco
- B Lymphocyte Dynamics, Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Carlos Oscar Sorzano
- Biocomputing Unit, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
- Neurodegenerative Diseases Biomedical Research Network Center (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Nuria E Campillo
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Lasse D Jensen
- Division of Diagnostics and Specialist Medicine, Department of Health, Medical and Caring Sciences, Linköping University, Linköping, Sweden
| | - Jose Miguel Rodriguez Frade
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - César Santiago
- X-ray Crystallography Unit, Department of Macromolecules Structure, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Mario Mellado
- Chemokine Signaling group, Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, Madrid, Spain
| |
Collapse
|
47
|
Cerutti JP, Diniz LA, Santos VC, Vilchez Larrea SC, Alonso GD, Ferreira RS, Dehaen W, Quevedo MA. Structure-Aided Computational Design of Triazole-Based Targeted Covalent Inhibitors of Cruzipain. Molecules 2024; 29:4224. [PMID: 39275072 PMCID: PMC11396839 DOI: 10.3390/molecules29174224] [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: 07/03/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
Cruzipain (CZP), the major cysteine protease present in T. cruzi, the ethiological agent of Chagas disease, has attracted particular attention as a therapeutic target for the development of targeted covalent inhibitors (TCI). The vast chemical space associated with the enormous molecular diversity feasible to explore by means of modern synthetic approaches allows the design of CZP inhibitors capable of exhibiting not only an efficient enzyme inhibition but also an adequate translation to anti-T. cruzi activity. In this work, a computer-aided design strategy was developed to combinatorially construct and screen large libraries of 1,4-disubstituted 1,2,3-triazole analogues, further identifying a selected set of candidates for advancement towards synthetic and biological activity evaluation stages. In this way, a virtual molecular library comprising more than 75 thousand diverse and synthetically feasible analogues was studied by means of molecular docking and molecular dynamic simulations in the search of potential TCI of CZP, guiding the synthetic efforts towards a subset of 48 candidates. These were synthesized by applying a Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) centered synthetic scheme, resulting in moderate to good yields and leading to the identification of 12 hits selectively inhibiting CZP activity with IC50 in the low micromolar range. Furthermore, four triazole derivatives showed good anti-T. cruzi inhibition when studied at 50 μM; and Ald-6 excelled for its high antitrypanocidal activity and low cytotoxicity, exhibiting complete in vitro biological activity translation from CZP to T. cruzi. Overall, not only Ald-6 merits further advancement to preclinical in vivo studies, but these findings also shed light on a valuable chemical space where molecular diversity might be explored in the search for efficient triazole-based antichagasic agents.
Collapse
Affiliation(s)
- Juan Pablo Cerutti
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC), Haya de la Torre y Medina Allende, Córdoba 5000, Argentina
- Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Lucas Abreu Diniz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte 31270-901, Brazil
| | - Viviane Corrêa Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte 31270-901, Brazil
| | - Salomé Catalina Vilchez Larrea
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Vuelta de Obligado 2490, Ciudad de Buenos Aires 1428, Argentina
| | - Guillermo Daniel Alonso
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Vuelta de Obligado 2490, Ciudad de Buenos Aires 1428, Argentina
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte 31270-901, Brazil
| | - Wim Dehaen
- Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Mario Alfredo Quevedo
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC), Haya de la Torre y Medina Allende, Córdoba 5000, Argentina
| |
Collapse
|
48
|
Kabier M, Gambacorta N, Trisciuzzi D, Kumar S, Nicolotti O, Mathew B. MzDOCK: A free ready-to-use GUI-based pipeline for molecular docking simulations. J Comput Chem 2024; 45:1980-1986. [PMID: 38703357 DOI: 10.1002/jcc.27390] [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: 03/01/2024] [Revised: 04/12/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024]
Abstract
Molecular docking is by far the most preferred approach in structure-based drug design for its effectiveness to predict the scoring and posing of a given bioactive small molecule into the binding site of its pharmacological target. Herein, we present MzDOCK, a new GUI-based pipeline for Windows operating system, designed with the intent of making molecular docking easier to use and higher reproducible even for inexperienced people. By harmonic integration of python and batch scripts, which employs various open source packages such as Smina (docking engine), OpenBabel (file conversion) and PLIP (analysis), MzDOCK includes many practical options such as: binding site configuration based on co-crystallized ligands; generation of enantiomers from SMILES input; application of different force fields (MMFF94, MMFF94s, UFF, GAFF, Ghemical) for energy minimization; retention of selectable ions and cofactors; sidechain flexibility of selectable binding site residues; multiple input file format (SMILES, PDB, SDF, Mol2, Mol); generation of reports and of pictures for interactive visualization. Users can download for free MzDOCK at the following link: https://github.com/Muzatheking12/MzDOCK.
Collapse
Affiliation(s)
- Muzammil Kabier
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| | - Nicola Gambacorta
- Division of Medical Genetics, IRCSS Foundation-Casa Sollievo della Sofferenza, San Giovanni Rotondo (Foggia), Foggia, Italy
| | - Daniela Trisciuzzi
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| | - Orazio Nicolotti
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, India
| |
Collapse
|
49
|
Wang Y, Shen Z, Chen R, Chi X, Li W, Xu D, Lu Y, Ding J, Dong X, Zheng X. Discovery and characterization of novel FGFR1 inhibitors in triple-negative breast cancer via hybrid virtual screening and molecular dynamics simulations. Bioorg Chem 2024; 150:107553. [PMID: 38901279 DOI: 10.1016/j.bioorg.2024.107553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 06/22/2024]
Abstract
The overexpression of FGFR1 is thought to significantly contribute to the progression of triple-negative breast cancer (TNBC), impacting aspects such as tumorigenesis, growth, metastasis, and drug resistance. Consequently, the pursuit of effective inhibitors for FGFR1 is a key area of research interest. In response to this need, our study developed a hybrid virtual screening method. Utilizing KarmaDock, an innovative algorithm that blends deep learning with molecular docking, alongside Schrödinger's Residue Scanning. This strategy led us to identify compound 6, which demonstrated promising FGFR1 inhibitory activity, evidenced by an IC50 value of approximately 0.24 nM in the HTRF bioassay. Further evaluation revealed that this compound also inhibits the FGFR1 V561M variant with an IC50 value around 1.24 nM. Our subsequent investigations demonstrate that Compound 6 robustly suppresses the migration and invasion capacities of TNBC cell lines, through the downregulation of p-FGFR1 and modulation of EMT markers, highlighting its promise as a potent anti-metastatic therapeutic agent. Additionally, our use of molecular dynamics simulations provided a deeper understanding of the compound's specific binding interactions with FGFR1.
Collapse
Affiliation(s)
- Yuchen Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zheyuan Shen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Roufen Chen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinglong Chi
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Wenjie Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Donghang Xu
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yan Lu
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianjun Ding
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Xiaoli Zheng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China.
| |
Collapse
|
50
|
Ngo VA. Insight into molecular basis and dynamics of full-length CRaf kinase in cellular signaling mechanisms. Biophys J 2024; 123:2623-2637. [PMID: 38946141 PMCID: PMC11365224 DOI: 10.1016/j.bpj.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/15/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024] Open
Abstract
Raf kinases play key roles in signal transduction in cells for regulating proliferation, differentiation, and survival. Despite decades of research into functions and dynamics of Raf kinases with respect to other cytosolic proteins, understanding Raf kinases is limited by the lack of their full-length structures at the atomic resolution. Here, we present the first model of the full-length CRaf kinase obtained from artificial intelligence/machine learning algorithms with a converging ensemble of structures simulated by large-scale temperature replica exchange simulations. Our model is validated by comparing simulated structures with the latest cryo-EM structure detailing close contacts among three key domains and regions of the CRaf. Our simulations identify potentially new epitopes of intramolecule interactions within the CRaf and reveal a dynamical nature of CRaf kinases, in which the three domains can move back and forth relative to each other for regulatory dynamics. The dynamic conformations are then used in a docking algorithm to shed insight into the paradoxical effect caused by vemurafenib in comparison with a paradox breaker PLX7904. We propose a model of Raf-heterodimer/KRas-dimer as a signalosome based on the dynamics of the full-length CRaf.
Collapse
Affiliation(s)
- Van A Ngo
- Advanced Computing for Life Sciences and Engineering, Science Engagement Section, Computing and Computational Sciences, National Center for Computational Sciences, Oak Ridge National Laboratory, Oak Ridge, Tennessee.
| |
Collapse
|