1
|
Yao Y, Chen F, Wu C, Chang X, Cheng W, Wang Q, Deng Z, Liu T, Lu L. Structure-based virtual screening aids the identification of glycosyltransferases in the biosynthesis of salidroside. PLANT BIOTECHNOLOGY JOURNAL 2025; 23:1725-1735. [PMID: 39932927 PMCID: PMC12018814 DOI: 10.1111/pbi.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/14/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Glycosylation plays an important role in the structural diversification of plant natural products. The identification of efficient glycosyltransferases is also a crucial step for the biosynthesis of valuable glycoside products. However, functional characterization of glycosyltransferases (GTs) from an extensive plant gene list is labour-intensive and challenging. Salidroside is a bioactive component derived from plants, widely utilized in the fields of food and medicine. Here, through transcriptome analysis and structure-based virtual screening, we identified two GTs that participated in the biosynthesis of salidroside from a rarely studied herbaceous plant, Astilbe chinensis. Ach15909 was found to possess high catalytic activity as evidenced by the determination of its catalytic parameters. The key residues that determine its catalytic activity were further determined. Additionally, Ach15909 shows a preference for substrates with a volume of <150 Å3, and replacing the interdomain linker region located between the N- and C-terminal domains of Ach15909 allows it to accept substrates that were previously not catalyzable. Overall, the structure-based virtual screening approach showed high efficiency and cost-effectiveness; the successful identification of GTs in salidroside glycosylation sheds light on uncovering additional plant biosynthesis enzymes in the forthcoming research.
Collapse
Affiliation(s)
- Yan Yao
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
- Hubei Hongshan LaboratoryWuhanChina
| | - Fangfang Chen
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
- Hubei Hongshan LaboratoryWuhanChina
| | - Chaoyan Wu
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
| | - Xiaosa Chang
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
| | - Weijia Cheng
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
| | - Qiuxia Wang
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
| | - Zixin Deng
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
| | - Tiangang Liu
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
| | - Li Lu
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan UniversityWuhanChina
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical SciencesWuhan UniversityWuhanChina
- Hubei Hongshan LaboratoryWuhanChina
| |
Collapse
|
2
|
Gazzillo E, Colarusso E, Giordano A, Chini MG, Potenza M, Hofstetter RK, Iorizzi M, Werz O, Lauro G, Bifulco G. Repositioning of Small Molecules through the Inverse Virtual Screening in silico Tool: Case of Benzothiazole-Based Inhibitors of Soluble Epoxide Hydrolase (sEH). Chempluschem 2024; 89:e202400234. [PMID: 38753468 DOI: 10.1002/cplu.202400234] [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: 03/28/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Computational techniques accelerate drug discovery by identifying bioactive compounds for specific targets, optimizing molecules with moderate activity, or facilitating the repositioning of inactive items onto new targets. Among them, the Inverse Virtual Screening (IVS) approach is aimed at the evaluation of one or a small set of molecules against a panel of targets for addressing target identification. In this work, a focused library of benzothiazole-based compounds was re-investigated by IVS. Four items, originally synthesized and tested on bromodomain-containing protein 9 (BRD9) but yielding poor binding, were critically re-analyzed, disclosing only a partial fit with 3D structure-based pharmacophore models, which, in the meanwhile, were developed for this target. Afterwards, these compounds were re-evaluated through IVS on a panel of proteins involved in inflammation and cancer, identifying soluble epoxide hydrolase (sEH) as a putative interacting target. Three items were subsequently confirmed as able to interfere with sEH activity, leading to inhibition percentages spanning from 70 % up to 30 % when tested at 10 μM. Finally, one benzothiazole-based compound emerged as the most promising inhibitor featuring an IC50 in the low micromolar range (IC50=6.62±0.13 μM). Our data confirm IVS as a predictive tool for accelerating the target identification and repositioning processes.
Collapse
Affiliation(s)
- Erica Gazzillo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Ester Colarusso
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Assunta Giordano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
- Institute of Biomolecular Chemistry (ICB), Consiglio Nazionale delle Ricerche (CNR), Via Campi Flegrei 34, Pozzuoli, I-80078, Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, Pesche, 86090, Italy
| | - Marianna Potenza
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Robert Klaus Hofstetter
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, Philosophenweg 14, Jena, 07743, Germany
| | - Maria Iorizzi
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, Pesche, 86090, Italy
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, Philosophenweg 14, Jena, 07743, Germany
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| |
Collapse
|
3
|
De Vita S, Colarusso E, Chini MG, Bifulco G, Lauro G. PharmaCore: The Automatic Generation of 3D Structure-Based Pharmacophore Models from Protein/Ligand Complexes. J Chem Inf Model 2024; 64:4263-4276. [PMID: 38728062 DOI: 10.1021/acs.jcim.3c01920] [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: 05/28/2024]
Abstract
In this work, we present PharmaCore: a new, completely automatic workflow aimed at generating three-dimensional (3D) structure-based pharmacophore models toward any target of interest. The proposed approach relies on using cocrystallized ligands to create the input files for generating the pharmacophore hypotheses, integrating not only the three-dimensional structural information on the ligand but also data concerning the binding mode of these molecules put in the protein cavity. We developed a Python library that, starting from the specific UniProt ID of the protein under investigation as the only element that requires user intervention, subsequently collects and aligns the corresponding structures bearing a known ligand in a fully automated fashion, bringing them all into the same coordinate system. The protocol includes a final phase in which the aligned small molecules are used to produce the pharmacophore hypotheses directly onto the protein structure using a specific software, e.g., Phase (Schrödinger LLC). To validate the entire procedure and highlight the possible applications in the field of drug discovery and repositioning, we first generated pharmacophores for soluble epoxide hydrolase (sEH) and compared with already-published ones. Then, we reproduced the binding profile of a reported selective binder of ATAD2 bromodomain (AM879), testing it against a panel of 1741 pharmacophores related to 16 epigenetic proteins and automatically generated with PharmaCore, finally disclosing putative unprecedented off-targets. The computational predictions were successfully validated with AlphaScreen assays, highlighting the applicability of the proposed workflow in drug discovery and repositioning. Finally, the process was also validated on tankyrase 2 and SARS-CoV-2 MPro, confirming the robustness of PharmaCore.
Collapse
Affiliation(s)
- Simona De Vita
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Ester Colarusso
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, Isernia 86090, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
4
|
Panwar U, Murali A, Khan MA, Selvaraj C, Singh SK. Virtual Screening Process: A Guide in Modern Drug Designing. Methods Mol Biol 2024; 2714:21-31. [PMID: 37676591 DOI: 10.1007/978-1-0716-3441-7_2] [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: 09/08/2023]
Abstract
Due to its capacity to drastically cut the cost and time necessary for experimental screening of compounds, virtual screening (VS) has grown to be a crucial component of drug discovery and development. VS is a computational method used in drug design to identify potential drugs from enormous libraries of chemicals. This approach makes use of molecular modeling and docking simulations to assess the small molecule's ability to bind to the desired protein. Virtual screening has a bright future, as high computational power and modern techniques are likely to further enhance the accuracy and speed of the process.
Collapse
Affiliation(s)
- Umesh Panwar
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Aarthy Murali
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Mohammad Aqueel Khan
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Chandrabose Selvaraj
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
- Department of Data Sciences, Centre of Biomedical Research, SGPGIMS Campus, Lucknow, Uttar Pradesh, India
| |
Collapse
|
5
|
De Vita S, Chini MG, Bifulco G, Lauro G. Target identification by structure-based computational approaches: Recent advances and perspectives. Bioorg Med Chem Lett 2023; 83:129171. [PMID: 36739998 DOI: 10.1016/j.bmcl.2023.129171] [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: 08/05/2022] [Revised: 12/15/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
The use of computational techniques in the early stages of drug discovery has recently experienced a boost, especially in the target identification step. Finding the biological partner(s) for new or existing synthetic and/or natural compounds by "wet" approaches may be challenging; therefore, preliminary in silico screening is even more recommended. After a brief overview of some of the most known target identification techniques, recent advances in structure-based computational approaches for target identification are reported in this digest, focusing on Inverse Virtual Screening and its recent applications. Moreover, future perspectives concerning the use of such methodologies, coupled or not with other approaches, are analyzed.
Collapse
Affiliation(s)
- Simona De Vita
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche (IS), Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy.
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy.
| |
Collapse
|
6
|
Wang R, Zhang W, Ma H, Zou D, Zhang Z, Wang S. Structural insights into the binding of zoledronic acid with RANKL via computational simulations. Front Mol Biosci 2022; 9:992473. [PMID: 36200071 PMCID: PMC9527314 DOI: 10.3389/fmolb.2022.992473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
Zoledronic acid (ZOL) inhibits receptor activator of nuclear factor-κB ligand (RANKL) and reduces bone turnover. This plays an important role in the development of bisphosphonate-related osteonecrosis of the jaw (BRONJ). Previous reports have shown that ZOL binds to the enzyme farnesyl pyrophosphate synthase (FPPS) to block its activity. However, the mechanism of action of ZOL and its interaction with RANKL is still unclear. In this study, we confirmed that ZOL significantly suppressed the bone remodeling in ZOL-treated rats, investigated whether ZOL could bind to RANKL and examined the interactions between these molecules at the atomic level. Surface plasmon resonance (SPR) assay was performed to validate that ZOL could directly bind to RANKL in a dose dependent manner, and the equilibrium constant was calculated (KD = 2.28 × 10−4 M). Then, we used molecular docking simulation to predict the binding site and analyze the binding characteristics of ZOL and RANKL. Through molecular dynamics simulation, we confirmed the stable binding between ZOL and RANKL and observed their dynamic interactions over time. Binding free energy calculations and its decomposition were conducted to obtain the binding free energy −70.67 ± 2.62 kJ/mol for the RANKL–ZOL complex. We identified the key residues of RANKL in the binding region, and these included Tyr217(A), Val277(A), Gly278(A), Val277(B), Gly278(B), and Tyr215(C). Taken together, our results demonstrated the direct interaction between ZOL and RANKL, indicating that the pharmacological action of ZOL might be closely related to RANKL. The design of novel small molecules targeting RANKL might reduce the occurrence of BRONJ.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjie Zhang
- Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University, Shanghai, China
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hailong Ma
- Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Duohong Zou
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyuan Zhang
- Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Zhiyuan Zhang, ; Shaoyi Wang,
| | - Shaoyi Wang
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Zhiyuan Zhang, ; Shaoyi Wang,
| |
Collapse
|
7
|
De Vita S, Finamore C, Chini MG, Saviano G, De Felice V, De Marino S, Lauro G, Casapullo A, Fantasma F, Trombetta F, Bifulco G, Iorizzi M. Phytochemical Analysis of the Methanolic Extract and Essential Oil from Leaves of Industrial Hemp Futura 75 Cultivar: Isolation of a New Cannabinoid Derivative and Biological Profile Using Computational Approaches. PLANTS 2022; 11:plants11131671. [PMID: 35807623 PMCID: PMC9269227 DOI: 10.3390/plants11131671] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022]
Abstract
Cannabis sativa L. is a plant belonging to the Cannabaceae family, cultivated for its psychoactive cannabinoid (Δ9-THC) concentration or for its fiber and nutrient content in industrial use. Industrial hemp shows a low Δ9-THC level and is a valuable source of phytochemicals, mainly represented by cannabinoids, flavones, terpenes, and alkaloids, with health-promoting effects. In the present study, we investigated the phytochemical composition of leaves of the industrial hemp cultivar Futura 75, a monoecious cultivar commercially used for food preparations or cosmetic purposes. Leaves are generally discarded, and represent waste products. We analyzed the methanol extract of Futura 75 leaves by HPLC and NMR spectroscopy and the essential oil by GC-MS. In addition, in order to compare the chemical constituents, we prepared the water infusion. One new cannabinoid derivative (1) and seven known components, namely, cannabidiol (2), cannabidiolic acid (3), β-cannabispirol (4), β-cannabispirol (5), canniprene (6), cannabiripsol (7), and cannflavin B (8) were identified. The content of CBD was highest in all preparations. In addition, we present the outcomes of a computational study focused on elucidating the role of 2α-hydroxy-Δ3,7-cannabitriol (1), CBD (2), and CBDA (3) in inflammation and thrombogenesis.
Collapse
Affiliation(s)
- Simona De Vita
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy; (S.D.V.); (G.L.); (A.C.)
| | - Claudia Finamore
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (S.D.M.)
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (M.G.C.); (G.S.); (V.D.F.); (F.F.)
| | - Gabriella Saviano
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (M.G.C.); (G.S.); (V.D.F.); (F.F.)
| | - Vincenzo De Felice
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (M.G.C.); (G.S.); (V.D.F.); (F.F.)
| | - Simona De Marino
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (S.D.M.)
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy; (S.D.V.); (G.L.); (A.C.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy; (S.D.V.); (G.L.); (A.C.)
| | - Francesca Fantasma
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (M.G.C.); (G.S.); (V.D.F.); (F.F.)
| | - Federico Trombetta
- Societa Cooperativa Agricola MarcheSana, Localita San Biagio 40, 61032 Fano, Italy;
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy; (S.D.V.); (G.L.); (A.C.)
- Correspondence: (G.B.); (M.I.); Tel.: +39-089969741 (G.B.); +39-087-4404100 (M.I.)
| | - Maria Iorizzi
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Isernia, Italy; (M.G.C.); (G.S.); (V.D.F.); (F.F.)
- Correspondence: (G.B.); (M.I.); Tel.: +39-089969741 (G.B.); +39-087-4404100 (M.I.)
| |
Collapse
|
8
|
De Vita S, Chini MG, Saviano G, Finamore C, Festa C, Lauro G, De Marino S, Russo R, Avagliano C, Casapullo A, Calignano A, Bifulco G, Iorizzi M. Biological Profile of Two Gentiana lutea L. Metabolites Using Computational Approaches and In Vitro Tests. Biomolecules 2021; 11:1490. [PMID: 34680124 PMCID: PMC8533323 DOI: 10.3390/biom11101490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 12/05/2022] Open
Abstract
Natural products have been the main source of bioactive molecules for centuries. We tested the biological profile of two metabolites extracted from Gentiana lutea L. by means of computational techniques and in vitro assays. The two molecules (loganic acid and gentiopicroside) were tested in silico using an innovative technique, named Inverse Virtual Screening (IVS), to highlight putative partners among a panel of proteins involved in inflammation and cancer events. A positive binding with cyclooxygenase-2 (COX-2), alpha-1-antichymotrypsin, and alpha-1-acid glycoprotein emerged from the computational experiments and the outcomes from the promising interaction with COX-2 were confirmed by Western blot, highlighting the reliability of IVS in the field of the natural products.
Collapse
Affiliation(s)
- Simona De Vita
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (S.D.V.); (G.L.); (A.C.)
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy; (M.G.C.); (G.S.)
| | - Gabriella Saviano
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy; (M.G.C.); (G.S.)
| | - Claudia Finamore
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (C.F.); (S.D.M.); (R.R.); (C.A.); (A.C.)
| | - Carmen Festa
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (C.F.); (S.D.M.); (R.R.); (C.A.); (A.C.)
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (S.D.V.); (G.L.); (A.C.)
| | - Simona De Marino
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (C.F.); (S.D.M.); (R.R.); (C.A.); (A.C.)
| | - Roberto Russo
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (C.F.); (S.D.M.); (R.R.); (C.A.); (A.C.)
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (C.F.); (S.D.M.); (R.R.); (C.A.); (A.C.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (S.D.V.); (G.L.); (A.C.)
| | - Antonio Calignano
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, 80131 Naples, Italy; (C.F.); (C.F.); (S.D.M.); (R.R.); (C.A.); (A.C.)
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (S.D.V.); (G.L.); (A.C.)
| | - Maria Iorizzi
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy; (M.G.C.); (G.S.)
| |
Collapse
|
9
|
Rajasekhar S, Karuppasamy R, Chanda K. Exploration of potential inhibitors for tuberculosis via structure-based drug design, molecular docking, and molecular dynamics simulation studies. J Comput Chem 2021; 42:1736-1749. [PMID: 34216033 DOI: 10.1002/jcc.26712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022]
Abstract
Drug resistance in tuberculosis is major threat to human population. In the present investigation, we aimed to identify novel and potent benzimidazole molecules to overcome the resistance management. A series of 20 benzimidazole derivatives were examined for its activity as selective antitubercular agents. Initially, AutodockVina algorithm was performed to assess the efficacy of the molecules. The results are further enriched by redocking by means of Glide algorithm. The binding free energies of the compounds were then calculated by MM-generalized-born surface area method. Molecular docking studies elucidated that benzimidazole derivatives has revealed formation of hydrogen bond and strong binding affinity in the active site of Mycobacterium tuberculosis protein. Note that ARG308, GLY189, VAL312, LEU403, and LEU190 amino acid residues of Mycobacterium tuberculosis protein PrpR are involved in binding with ligands of benzimidazoles. Interestingly, the ligands exhibited same binding potential to the active site of protein complex PrpR in both the docking programs. In essence, the result portrays that benzimidazole derivatives such as 1p, 1q, and 1 t could be potent and selective antitubercular agents than the standard drug isoniazid. These compounds were then subjected to molecular dynamics simulation to validate the dynamics activity of the compounds against PrpR. Finally, the inhibitory behavior of compounds was predicted using a machine learning algorithm trained on a data collection of 15,000 compounds utilizing graph-based signatures. Overall, the study concludes that designed benzimidazoles can be employed as antitubercular agents. Indeed, the results are helpful for the experimental biologists to develop safe and non-toxic drugs against tuberculosis.
Collapse
Affiliation(s)
- Sreerama Rajasekhar
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, India
| | - Ramanathan Karuppasamy
- Department of Biotechnology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Kaushik Chanda
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
10
|
Chini MG, Lauro G, Bifulco G. Addressing the Target Identification and Accelerating the Repositioning of Anti‐Inflammatory/Anti‐Cancer Organic Compounds by Computational Approaches. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Maria Giovanna Chini
- Department of Biosciences and Territory University of Molise C.da Fonte Lappone 86090 Pesche (IS) Italy
| | - Gianluigi Lauro
- Department of Pharmacy University of Salerno Via Giovanni Paolo II 132 84084 Fisciano (SA) Italy
| | - Giuseppe Bifulco
- Department of Pharmacy University of Salerno Via Giovanni Paolo II 132 84084 Fisciano (SA) Italy
| |
Collapse
|
11
|
Potenza M, Cavalluzzi MM, Milani G, Lauro G, Carino A, Roselli R, Fiorucci S, Zampella A, Pierri CL, Lentini G, Bifulco G. Inverse Virtual Screening for the rapid re-evaluation of the presumed biological safe profile of natural products. The case of steviol from Stevia rebaudiana glycosides on farnesoid X receptor (FXR). Bioorg Chem 2021; 111:104897. [PMID: 33901797 DOI: 10.1016/j.bioorg.2021.104897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/20/2021] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
Nonnutritive sweeteners (NNSs) are widely employed as dietary substitutes for classical sugars thanks to their safety profile and low toxicity. In this study, a re-evaluation of the biological effects of steviol (1), the main metabolite from Stevia rebaudiana glycosides, was performed using the Inverse Virtual Screening (IVS) target fishing computational approach. Starting from well-known pharmacological properties of Stevia rebaudiana glycosides, this computational tool was employed for predicting the putative interacting targets of 1 and, afterwards, of its five synthetic ester derivatives 2-6, accounting a large panel of proteins involved in cancer and inflammation events. Applying this methodology, the farnesoid X receptor (FXR) was identified as the putative target partner of 1-6. The predicted ligand-protein interactions were corroborated by transactivation assays, specifically disclosing the agonistic activity of 1 and the antagonistic activities of 2-6 on FXR. The reported results highlight the feasibility of IVS as a fast and potent tool for predicting the interacting targets of query compounds, addressing the re-evaluation of their bioactivity. In light of the obtained results, the presumably safe profile of known compounds, such as the case of steviol (1), is critically discussed.
Collapse
Affiliation(s)
- Marianna Potenza
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy
| | - Maria Maddalena Cavalluzzi
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Via Edoardo Orabona, 4, Bari 70126, Italy
| | - Gualtiero Milani
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Via Edoardo Orabona, 4, Bari 70126, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy
| | - Adriana Carino
- Department of Surgery and Biomedical Sciences, Nuova facoltà di Medicina, Perugia, Italy
| | - Rosalinda Roselli
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, Naples 80131, Italy
| | - Stefano Fiorucci
- Department of Surgery and Biomedical Sciences, Nuova facoltà di Medicina, Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Naples, Via Domenico Montesano, 49, Naples 80131, Italy
| | - Ciro Leonardo Pierri
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari Aldo Moro, Via Edoardo Orabona, 4, Bari 70126, Italy
| | - Giovanni Lentini
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Via Edoardo Orabona, 4, Bari 70126, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy.
| |
Collapse
|
12
|
Discovery of noscapine derivatives as potential β-tubulin inhibitors. Bioorg Med Chem Lett 2020; 30:127489. [PMID: 32784088 DOI: 10.1016/j.bmcl.2020.127489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/05/2020] [Accepted: 08/08/2020] [Indexed: 10/23/2022]
Abstract
Twenty novel 1,2,3-triazole noscapine derivatives were synthesized starting from noscapine by consecutive N-demethylation, reduction of lactone ring, N-propargylation and Huisgen 1,3-dipolar cycloaddition reaction. In order to select the most promising molecules to subject to further biophysical and biological evaluation, a molecular docking analysis round was performed using noscapine as reference compound. The molecules featuring docking predicted binding affinity better than that of noscapine were then subjected to MTT assay against MCF7 cell line. The obtained results disclosed that all the selected triazole derivatives exhibited a remarkably lower cell viability compared to noscapine in the range of 20 μM in 48 h. In an attempt to correlate the biological activity with the ability to bind tubulin, the surface plasmon resonance (SPR) assay was employed. Compounds 8a, 8h, 9c, 9f and 9j were able to bind tubulin with affinity constant values in the nanomolar range and higher if compared to noscapine. Integrating computational predictions and experimental evaluation, two promising compounds (8h and 9c) were identified, whose relevant cytotoxicity was supposed to be correlated with tubulin binding affinity. These findings shed lights onto structural modifications of noscapine toward the identification of more potent cytotoxic agents targeting tubulin.
Collapse
|
13
|
De Vita S, Terracciano S, Bruno I, Chini MG. From Natural Compounds to Bioactive Molecules through NMR and
In Silico
Methodologies. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Simona De Vita
- Department of Pharmacy University of Salerno Via Giovanni Paolo II, n°132 84084 Fisciano (SA) Italy
| | - Stefania Terracciano
- Department of Pharmacy University of Salerno Via Giovanni Paolo II, n°132 84084 Fisciano (SA) Italy
| | - Ines Bruno
- Department of Pharmacy University of Salerno Via Giovanni Paolo II, n°132 84084 Fisciano (SA) Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory University of Molise C.da Fonte Lappone‐ 86090 Pesche (IS) Italy
| |
Collapse
|
14
|
De Vita S, Chini MG, Lauro G, Bifulco G. Accelerating the repurposing of FDA-approved drugs against coronavirus disease-19 (COVID-19). RSC Adv 2020; 10:40867-40875. [PMID: 35519188 PMCID: PMC9057693 DOI: 10.1039/d0ra09010g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
The recent release of the main protein structures belonging to SARS CoV-2, responsible for the coronavirus disease-19 (COVID-19), strongly pushed for identifying valuable drug treatments. With this aim, we show a repurposing study on FDA-approved drugs applying a new computational protocol and introducing a novel parameter called IVSratio. Starting with a virtual screening against three SARS CoV-2 targets (main protease, papain-like protease, spike protein), the top-ranked molecules were reassessed combining the Inverse Virtual Screening novel approach and MM-GBSA calculations. Applying this protocol, a list of drugs was identified against the three investigated targets. Also, the top-ranked selected compounds on each target (rutin vs. main protease, velpatasvir vs. papain-like protease, lomitapide vs. spike protein) were further tested with molecular dynamics simulations to confirm the promising binding modes, obtaining encouraging results such as high stability of the complex during the simulation and a good protein–ligand interaction network involving some important residues of each target. Moreover, the recent outcomes highlighting the inhibitory activity of quercetin, a natural compound strictly related to rutin, on the SARS-CoV-2 main protease, strengthened the applicability of the proposed workflow. New computational protocol applied to a repurposing campaign against SARS-CoV-2.![]()
Collapse
Affiliation(s)
- Simona De Vita
- Department of Pharmacy
- University of Salerno
- Fisciano 84084
- Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory
- University of Molise
- 86090 Pesche (IS)
- Italy
| | - Gianluigi Lauro
- Department of Pharmacy
- University of Salerno
- Fisciano 84084
- Italy
| | | |
Collapse
|