1
|
La Monica G, Gallo A, Bono A, Alamia F, Lauria A, Alduina R, Martorana A. Novel Antibacterial 4-Piperazinylquinoline Hybrid Derivatives Against Staphylococcus aureus: Design, Synthesis, and In Vitro and In Silico Insights. Molecules 2024; 30:28. [PMID: 39795086 PMCID: PMC11720749 DOI: 10.3390/molecules30010028] [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: 11/18/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Molecular hybridization, which consists of the combination of two or more pharmacophores into a single molecule, is an innovative approach in drug design to afford new chemical entities with enhanced biological activity. In the present study, this strategy was pursued to develop a new series of 6,7-dimethoxy-4-piperazinylquinoline-3-carbonitrile derivatives (5a-k) with potential antibiotic activity by combining the quinoline, the piperazinyl, and the benzoylamino moieties, three recurrent frameworks in antimicrobial research. Initial in silico evaluations were conducted on the designed compounds, highlighting favorable ADMET and drug-likeness properties, which were synthesized through a multistep strategy, isolated, and fully characterized. The whole set was tested in vitro against Staphylococcus aureus ATCC 25923 and Pseudomonas aeruginosa ATCC 10145 representative Gram-positive and Gram-negative strains, respectively. Notably, 5k exhibited potent and selective activity against S. aureus (MIC 10 μM), with a dose- and time-dependent response and capability to affect cell membrane integrity. On the other hand, no significant activity was observed against P. aeruginosa. Further in silico docking and molecular dynamics studies highlighted strong interactions of 5k with bacterial enzymes, such as tyrosyl-tRNA synthetase, pyruvate kinase, and DNA gyrase B, suggesting potential modes of action. These findings underscore the value of the hybridization approach in producing new antimicrobial agents, guiding future optimization for broader-spectrum activity.
Collapse
Affiliation(s)
- Gabriele La Monica
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (G.L.M.); (A.G.); (A.B.); (F.A.); (A.L.); (R.A.)
| | - Annamaria Gallo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (G.L.M.); (A.G.); (A.B.); (F.A.); (A.L.); (R.A.)
| | - Alessia Bono
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (G.L.M.); (A.G.); (A.B.); (F.A.); (A.L.); (R.A.)
| | - Federica Alamia
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (G.L.M.); (A.G.); (A.B.); (F.A.); (A.L.); (R.A.)
| | - Antonino Lauria
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (G.L.M.); (A.G.); (A.B.); (F.A.); (A.L.); (R.A.)
- NBFC, National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy
| | - Rosa Alduina
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (G.L.M.); (A.G.); (A.B.); (F.A.); (A.L.); (R.A.)
- NBFC, National Biodiversity Future Center, Piazza Marina 61, 90133 Palermo, Italy
| | - Annamaria Martorana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (G.L.M.); (A.G.); (A.B.); (F.A.); (A.L.); (R.A.)
| |
Collapse
|
2
|
Han B, Luo J, Xu B. Revealing Molecular Mechanisms of the Bioactive Saponins from Edible Root of Platycodon grandiflorum in Combating Obesity. PLANTS (BASEL, SWITZERLAND) 2024; 13:1123. [PMID: 38674532 PMCID: PMC11053671 DOI: 10.3390/plants13081123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
Obesity has emerged as a significant health concern, as it is a disease linked to metabolic disorders in the body and is characterized by the excessive accumulation of lipids. As a plant-derived food, Platycodon grandiflorum (PG) was reported by many studies, indicating that the saponins from PG can improve obesity effectively. However, the anti-obesity saponins from PG and its anti-obesity mechanisms have not been fully identified. This study identified the active saponins and their molecular targets for treating obesity. The TCMSP database was used to obtain information on 18 saponins in PG. The anti-obesity target of the PG saponins was 115 targets and 44 core targets. GO and KEGG analyses using 44 core anti-obesity genes and targets of PG-active saponins screened from GeneCards, OMIM, Drugbank, and DisGeNet showed that the PI3K-Akt pathway, the JAK-STAT pathway, and the MAPK pathway were the major pathways involved in the anti-obesity effects of PG saponins. BIOVIA Discovery Studio Visualizer and AutoDock Vina were used to perform molecular docking and process the molecular docking results. The molecular docking results showed that the active saponins of PG could bind to the major therapeutic obesity targets to play an obesity-inhibitory role. The results of this study laid the foundation for further research on the anti-obesity saponins in PG and their anti-obesity mechanism and provided a new direction for the development of functional plant-derived food. This research studied the molecular mechanism of PG saponins combating obesity through various signaling pathways, and prosapogenin D can be used to develop as a new potential anti-obesity drug.
Collapse
Affiliation(s)
| | | | - Baojun Xu
- Guangdong Provincial Key Laboratory IRADS and Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China; (B.H.); (J.L.)
| |
Collapse
|
3
|
Ragusa MA, Naselli F, Cruciata I, Volpes S, Schimmenti C, Serio G, Mauro M, Librizzi M, Luparello C, Chiarelli R, La Rosa C, Lauria A, Gentile C, Caradonna F. Indicaxanthin Induces Autophagy in Intestinal Epithelial Cancer Cells by Epigenetic Mechanisms Involving DNA Methylation. Nutrients 2023; 15:3495. [PMID: 37571432 PMCID: PMC10420994 DOI: 10.3390/nu15153495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Autophagy is an evolutionarily conserved process critical in maintaining cellular homeostasis. Recently, the anticancer potential of autophagy inducers, including phytochemicals, was suggested. Indicaxanthin is a betalain pigment found in prickly pear fruit with antiproliferative and pro-apoptotic activities in colorectal cancer cells associated with epigenetic changes in selected methylation-silenced oncosuppressor genes. Here, we demonstrate that indicaxanthin induces the up-regulation of the autophagic markers LC3-II and Beclin1, and increases autophagolysosome production in Caco-2 cells. Methylomic studies showed that the indicaxanthin-induced pro-autophagic activity was associated with epigenetic changes. In addition to acting as a hypermethylating agent at the genomic level, indicaxanthin also induced significant differential methylation in 39 out of 47 autophagy-related genes, particularly those involved in the late stages of autophagy. Furthermore, in silico molecular modelling studies suggested a direct interaction of indicaxanthin with Bcl-2, which, in turn, influenced the function of Beclin1, a key autophagy regulator. External effectors, including food components, may modulate the epigenetic signature of cancer cells. This study demonstrates, for the first time, the pro-autophagic potential of indicaxanthin in human colorectal cancer cells associated with epigenetic changes and contributes to outlining its potential healthy effect in the pathophysiology of the gastrointestinal tract.
Collapse
Affiliation(s)
- Maria Antonietta Ragusa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Flores Naselli
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Ilenia Cruciata
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Sara Volpes
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Chiara Schimmenti
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Graziella Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Maurizio Mauro
- Department of Obstetrics & Gynecology and Women’s Health, Michael F. Price Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Mariangela Librizzi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Claudio Luparello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Roberto Chiarelli
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Chiara La Rosa
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, 10124 Turin, Italy;
| | - Antonino Lauria
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Carla Gentile
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
| | - Fabio Caradonna
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.A.R.); (F.N.); (I.C.); (S.V.); (C.S.); (G.S.); (M.L.); (C.L.); (R.C.); (A.L.); (F.C.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| |
Collapse
|
4
|
Off-Target-Based Design of Selective HIV-1 PROTEASE Inhibitors. Int J Mol Sci 2021; 22:ijms22116070. [PMID: 34199858 PMCID: PMC8200130 DOI: 10.3390/ijms22116070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/17/2022] Open
Abstract
The approval of the first HIV-1 protease inhibitors (HIV-1 PRIs) marked a fundamental step in the control of AIDS, and this class of agents still represents the mainstay therapy for this illness. Despite the undisputed benefits, the necessary lifelong treatment led to numerous severe side-effects (metabolic syndrome, hepatotoxicity, diabetes, etc.). The HIV-1 PRIs are capable of interacting with "secondary" targets (off-targets) characterized by different biological activities from that of HIV-1 protease. In this scenario, the in-silico techniques undoubtedly contributed to the design of new small molecules with well-fitting selectivity against the main target, analyzing possible undesirable interactions that are already in the early stages of the research process. The present work is focused on a new mixed-hierarchical, ligand-structure-based protocol, which is centered on an on/off-target approach, to identify the new selective inhibitors of HIV-1 PR. The use of the well-established, ligand-based tools available in the DRUDIT web platform, in combination with a conventional, structure-based molecular docking process, permitted to fast screen a large database of active molecules and to select a set of structure with optimal on/off-target profiles. Therefore, the method exposed herein, could represent a reliable help in the research of new selective targeted small molecules, permitting to design new agents without undesirable interactions.
Collapse
|
5
|
The dimer-monomer equilibrium of SARS-CoV-2 main protease is affected by small molecule inhibitors. Sci Rep 2021; 11:9283. [PMID: 33927258 PMCID: PMC8085067 DOI: 10.1038/s41598-021-88630-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/14/2021] [Indexed: 12/13/2022] Open
Abstract
The maturation of coronavirus SARS-CoV-2, which is the etiological agent at the origin of the COVID-19 pandemic, requires a main protease Mpro to cleave the virus-encoded polyproteins. Despite a wealth of experimental information already available, there is wide disagreement about the Mpro monomer-dimer equilibrium dissociation constant. Since the functional unit of Mpro is a homodimer, the detailed knowledge of the thermodynamics of this equilibrium is a key piece of information for possible therapeutic intervention, with small molecules interfering with dimerization being potential broad-spectrum antiviral drug leads. In the present study, we exploit Small Angle X-ray Scattering (SAXS) to investigate the structural features of SARS-CoV-2 Mpro in solution as a function of protein concentration and temperature. A detailed thermodynamic picture of the monomer-dimer equilibrium is derived, together with the temperature-dependent value of the dissociation constant. SAXS is also used to study how the Mpro dissociation process is affected by small inhibitors selected by virtual screening. We find that these inhibitors affect dimerization and enzymatic activity to a different extent and sometimes in an opposite way, likely due to the different molecular mechanisms underlying the two processes. The Mpro residues that emerge as key to optimize both dissociation and enzymatic activity inhibition are discussed.
Collapse
|
6
|
Mannino G, Iovino P, Lauria A, Genova T, Asteggiano A, Notarbartolo M, Porcu A, Serio G, Chinigò G, Occhipinti A, Capuzzo A, Medana C, Munaron L, Gentile C. Bioactive Triterpenes of Protium heptaphyllum Gum Resin Extract Display Cholesterol-Lowering Potential. Int J Mol Sci 2021; 22:ijms22052664. [PMID: 33800828 PMCID: PMC7961947 DOI: 10.3390/ijms22052664] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
Hypercholesterolemia is one of the major causes of cardiovascular disease, the risk of which is further increased if other forms of dyslipidemia occur. Current therapeutic strategies include changes in lifestyle coupled with drug administration. Statins represent the most common therapeutic approach, but they may be insufficient due to the onset of resistance mechanisms and side effects. Consequently, patients with mild hypercholesterolemia prefer the use of food supplements since these are perceived to be safer. Here, we investigate the phytochemical profile and cholesterol-lowering potential of Protium heptaphyllum gum resin extract (PHE). Chemical characterization via HPLC-APCI-HRMS2 and GC-FID/MS identified 13 compounds mainly belonging to ursane, oleanane, and tirucallane groups. Studies on human hepatocytes have revealed how PHE is able to reduce cholesterol production and regulate the expression of proteins involved in its metabolism. (HMGCR, PCSK9, LDLR, FXR, IDOL, and PPAR). Moreover, measuring the inhibitory activity of PHE against HMGR, moderate inhibition was recorded. Finally, molecular docking studies identified acidic tetra- and pentacyclic triterpenoids as the main compounds responsible for this action. In conclusion, our study demonstrates how PHE may be a useful alternative to contrast hypercholesterolemia, highlighting its potential as a sustainable multitarget natural extract for the nutraceutical industry that is rapidly gaining acceptance as a source of health-promoting compounds.
Collapse
Affiliation(s)
- Giuseppe Mannino
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Piera Iovino
- Biosfered S.R.L., 10148 Turin, Italy; (P.I.); (A.A.)
| | - Antonino Lauria
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Tullio Genova
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (T.G.); (G.C.); (L.M.)
| | - Alberto Asteggiano
- Biosfered S.R.L., 10148 Turin, Italy; (P.I.); (A.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10125 Torino, Italy (C.M.)
| | - Monica Notarbartolo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Alessandra Porcu
- Abel Nutraceuticals S.R.L., 10148 Turin, Italy; (A.P.); (A.O.); (A.C.)
| | - Graziella Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
| | - Giorgia Chinigò
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (T.G.); (G.C.); (L.M.)
| | - Andrea Occhipinti
- Abel Nutraceuticals S.R.L., 10148 Turin, Italy; (A.P.); (A.O.); (A.C.)
| | - Andrea Capuzzo
- Abel Nutraceuticals S.R.L., 10148 Turin, Italy; (A.P.); (A.O.); (A.C.)
| | - Claudio Medana
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10125 Torino, Italy (C.M.)
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy; (T.G.); (G.C.); (L.M.)
| | - Carla Gentile
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.M.); (A.L.); (M.N.); (G.S.)
- Correspondence: ; Tel.: +39-091-2388-6472
| |
Collapse
|
7
|
Lauria A, Mannino S, Gentile C, Mannino G, Martorana A, Peri D. DRUDIT: web-based DRUgs DIscovery Tools to design small molecules as modulators of biological targets. Bioinformatics 2020; 36:1562-1569. [PMID: 31605102 DOI: 10.1093/bioinformatics/btz783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/19/2019] [Accepted: 10/10/2019] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION New in silico tools to predict biological affinities for input structures are presented. The tools are implemented in the DRUDIT (DRUgs DIscovery Tools) web service. The DRUDIT biological finder module is based on molecular descriptors that are calculated by the MOLDESTO (MOLecular DEScriptors TOol) software module developed by the same authors, which is able to calculate more than one thousand molecular descriptors. At this stage, DRUDIT includes 250 biological targets, but new external targets can be added. This feature extends the application scope of DRUDIT to several fields. Moreover, two more functions are implemented: the multi- and on/off-target tasks. These tools applied to input structures allow for predicting the polypharmacology and evaluating the collateral effects. RESULTS The applications described in the article show that DRUDIT is able to predict a single biological target, to identify similarities among biological targets, and to discriminate different target isoforms. The main advantages of DRUDIT for the scientific community lie in its ease of use by worldwide scientists and the possibility to be used also without specific, and often expensive, hardware and software. In fact, it is fully accessible through the WWW from any device to perform calculations. Just a click or a tap can start tasks to predict biological properties for new compounds or repurpose drugs, lead compounds, or unsuccessful compounds. To date, DRUDIT is supported by four servers each able to execute 8 jobs simultaneously. AVAILABILITY AND IMPLEMENTATION The web service is accessible at the www.drudit.com URL and its use is free of charge. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Antonino Lauria
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF"
| | - Salvatore Mannino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF"
| | - Carla Gentile
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF"
| | - Giuseppe Mannino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF"
| | - Annamaria Martorana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF"
| | - Daniele Peri
- Dipartimento di Ingegneria, University of Palermo, Palermo I-90128, Italy
| |
Collapse
|
8
|
Moreno LM, Quiroga J, Abonia R, Lauria A, Martorana A, Insuasty H, Insuasty B. Synthesis, biological evaluation, and in silico studies of novel chalcone- and pyrazoline-based 1,3,5-triazines as potential anticancer agents. RSC Adv 2020; 10:34114-34129. [PMID: 35519030 PMCID: PMC9056798 DOI: 10.1039/d0ra06799g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/09/2020] [Indexed: 12/17/2022] Open
Abstract
A novel series of triazin-chalcones (7,8)a-g and triazin-N-(3,5-dichlorophenyl)pyrazolines (9,10)a-g were synthesized and evaluated for their anticancer activity against nine different cancer strains. Triazine ketones 5 and 6 were synthesized from the cyanuric chloride 1 by using stepwise nucleophilic substitution of the chlorine atom. These ketones were subsequently subjected to a Claisen-Schmidt condensation reaction with aromatic aldehydes affording chalcones (7,8)a-g. Then, N-(3,5-dichlorophenyl)pyrazolines (9,10)a-g were obtained by cyclocondensation reactions of the respective chalcones (7,8)a-g with 3,5-dichlorophenylhydrazine. Among all the evaluated compounds, chalcones 7d,g and 8g exhibited more potent in vitro anticancer activity, with outstanding GI50 values ranging from 0.422 to 14.9 μM and LC50 values ranging from 5.08 μM to >100 μM. In silico studies, for both ligand- and structure-based, were executed to explore the inhibitory nature of chalcones and triazine derivatives. The results suggested that the evaluated compounds could act as modulators of the human thymidylate synthase enzyme.
Collapse
Affiliation(s)
- Leydi M Moreno
- Heterocyclic Compounds Research Group, Department of Chemistry, Universidad del Valle A.A. 25360 Cali Colombia
| | - Jairo Quiroga
- Heterocyclic Compounds Research Group, Department of Chemistry, Universidad del Valle A.A. 25360 Cali Colombia
- Center for Bioinformatics and Photonics-CIBioFI A.A. 25360 Cali Colombia
| | - Rodrigo Abonia
- Heterocyclic Compounds Research Group, Department of Chemistry, Universidad del Valle A.A. 25360 Cali Colombia
- Center for Bioinformatics and Photonics-CIBioFI A.A. 25360 Cali Colombia
| | - Antonino Lauria
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF", Università di Palermo Viale delle Scienze Ed. 17 I-90128 Palermo Italy
| | - Annamaria Martorana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF", Università di Palermo Viale delle Scienze Ed. 17 I-90128 Palermo Italy
| | - Henry Insuasty
- Heterocyclic Compounds Research Group, Department of Chemistry, Universidad de Nariño A.A. 1175 Pasto Colombia
| | - Braulio Insuasty
- Heterocyclic Compounds Research Group, Department of Chemistry, Universidad del Valle A.A. 25360 Cali Colombia
- Center for Bioinformatics and Photonics-CIBioFI A.A. 25360 Cali Colombia
| |
Collapse
|
9
|
Martorana A, Gentile C, Lauria A. In Silico Insights into the SARS CoV-2 Main Protease Suggest NADH Endogenous Defences in the Control of the Pandemic Coronavirus Infection. Viruses 2020; 12:v12080805. [PMID: 32722574 PMCID: PMC7472248 DOI: 10.3390/v12080805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022] Open
Abstract
COVID-19 is a pandemic health emergency faced by the entire world. The clinical treatment of the severe acute respiratory syndrome (SARS) CoV-2 is currently based on the experimental administration of HIV antiviral drugs, such as lopinavir, ritonavir, and remdesivir (a nucleotide analogue used for Ebola infection). This work proposes a repurposing process using a database containing approximately 8000 known drugs in synergy structure- and ligand-based studies by means of the molecular docking and descriptor-based protocol. The proposed in silico findings identified new potential SARS CoV-2 main protease (MPRO) inhibitors that fit in the catalytic binding site of SARS CoV-2 MPRO. Several selected structures are NAD-like derivatives, suggesting a relevant role of these molecules in the modulation of SARS CoV-2 infection in conditions of cell chronic oxidative stress. Increased catabolism of NAD(H) during protein ribosylation in the DNA damage repair process may explain the greater susceptibility of the elderly population to the acute respiratory symptoms of COVID-19. The molecular modelling studies proposed herein agree with this hypothesis.
Collapse
|
10
|
Martorana A, Lauria A. Design of antitumor drugs targeting c-kit receptor by a new mixed ligand-structure based method. J Mol Graph Model 2020; 100:107666. [PMID: 32659630 DOI: 10.1016/j.jmgm.2020.107666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 10/23/2022]
Abstract
An important challenge, in the medicinal chemistry field, is the research of novel forceful drugs to overcome tumor-acquired resistance. The c-Kit tyrosine kinase receptor (TKR) represents a suitable target for the carcinogenesis control of gastro-intestinal stromal (GIST), leukemia, and mastocytosis tumors; nevertheless, several hotspot mutations of the protein limit the efficacy of a few clinical administered TKRs inhibitors. In this study, a new in silico protocol based on ligand and structure-based combined method is proposed, with the aim to identify a set of new c-Kit inhibitors able to complex c-Kit mutated proteins. A recent and freely available web-server DRUDIT is used for the ligand-based method. The protocol application allows for identifying a new generation of potential TKR inhibitors, which, in silico, complex the V654A and T670I mutated proteins and potentially overcome resistant mutations (D816H). The structure-based analysis is performed by Induced Fit Docking (IFD) studies. The comparison between the explored ligands and well-known drugs highlights the possibility to overcome tumor-acquired resistance. The best-selected structures (630705 and SML1348) provide valuable binding affinities with the mutated c-Kit forms (respectively T670I and V654A).
Collapse
Affiliation(s)
- Annamaria Martorana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Viale delle Scienze - Ed. 17, 90128, Palermo, Italy
| | - Antonino Lauria
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Viale delle Scienze - Ed. 17, 90128, Palermo, Italy.
| |
Collapse
|
11
|
Sakkiah S, Kusko R, Pan B, Guo W, Ge W, Tong W, Hong H. Structural Changes Due to Antagonist Binding in Ligand Binding Pocket of Androgen Receptor Elucidated Through Molecular Dynamics Simulations. Front Pharmacol 2018; 9:492. [PMID: 29867496 PMCID: PMC5962723 DOI: 10.3389/fphar.2018.00492] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/25/2018] [Indexed: 01/28/2023] Open
Abstract
When a small molecule binds to the androgen receptor (AR), a conformational change can occur which impacts subsequent binding of co-regulator proteins and DNA. In order to accurately study this mechanism, the scientific community needs a crystal structure of the Wild type AR (WT-AR) ligand binding domain, bound with antagonist. To address this open need, we leveraged molecular docking and molecular dynamics (MD) simulations to construct a structure of the WT-AR ligand binding domain bound with antagonist bicalutamide. The structure of mutant AR (Mut-AR) bound with this same antagonist informed this study. After molecular docking analysis pinpointed the suitable binding orientation of a ligand in AR, the model was further optimized through 1 μs of MD simulations. Using this approach, three molecular systems were studied: (1) WT-AR bound with agonist R1881, (2) WT-AR bound with antagonist bicalutamide, and (3) Mut-AR bound with bicalutamide. Our structures were very similar to the experimentally determined structures of both WT-AR with R1881 and Mut-AR with bicalutamide, demonstrating the trustworthiness of this approach. In our model, when WT-AR is bound with bicalutamide, Val716/Lys720/Gln733, or Met734/Gln738/Glu897 move and thus disturb the positive and negative charge clumps of the AF2 site. This disruption of the AF2 site is key for understanding the impact of antagonist binding on subsequent co-regulator binding. In conclusion, the antagonist induced structural changes in WT-AR detailed in this study will enable further AR research and will facilitate AR targeting drug discovery.
Collapse
Affiliation(s)
- Sugunadevi Sakkiah
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Rebecca Kusko
- Immuneering Corporation, Cambridge, MA, United States
| | - Bohu Pan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Wenjing Guo
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Weigong Ge
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| |
Collapse
|
12
|
Xiang L, Wang Y, Yi X, He X. Antiproliferative and anti-inflammatory furostanol saponins from the rhizomes of Tupistra chinensis. Steroids 2016; 116:28-37. [PMID: 27770616 DOI: 10.1016/j.steroids.2016.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 10/13/2016] [Accepted: 10/17/2016] [Indexed: 01/10/2023]
Abstract
Phytochemical investigations of the rhizome of Tupistra chinensis led to the isolation of ten new furostanol saponins along with fourteen known spirostanols. Their chemical structures were elucidated on the basis of spectroscopic and chemical methods, including IR, NMR, MS, and GC analyses. The antiproliferative effects against FaDu and Detroit 562 cell lines and inhibitory activities on nitric oxide (NO) production induced by lipopolysaccharide (LPS) in a macrophage cell line RAW 264.7 were assayed for all the isolated compounds. Compound 14 exhibited significant antiproliferative effects against FaDu and Detroit 562 cells with IC50 values of 1.1±0.1 and 1.2±0.1μM, respectively. Compounds 1, 2, 6, 13, 16, 19 and 24 exhibited inhibitory effects on NO production with IC50 values ranging from 15.7 to 46.2μM.
Collapse
Affiliation(s)
- Limin Xiang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yihai Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Xiaomin Yi
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiangjiu He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
13
|
Antiproliferative and anti-inflammatory polyhydroxylated spirostanol saponins from Tupistra chinensis. Sci Rep 2016; 6:31633. [PMID: 27530890 PMCID: PMC4987684 DOI: 10.1038/srep31633] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/22/2016] [Indexed: 02/08/2023] Open
Abstract
Tupistra chinensis is widely distributed in southwestern China and its rhizome is a famous folk medicine for the treatment of carbuncles and pharyngitis. Its chemical identity of potent antiproliferative and anti-inflammatory constituents has been carried out in this study. Twenty-three polyhydroxylated spirostanol saponins, including nine novels, were isolated and identified. The new spirostanol saponins were elucidated as spirost-25(27)-en-1β,2β,3β,4β,5β-pentol-2-O-β-D-xylopyranoside (1), spirost-25(27)- en-1β,2β,3β,4β,5β-pentol-2-O-α-L-arabinopyranoside (2), spirost-25(27)-en- 1β,3α,5β-triol (12), spirost-25(27)-en-1β,3α,4β,5β,6β-pentol (13), spirost-25(27)-en- 1β,2β,3β,5β-tetraol-5-O-β-D-glucopyranoside (16), 5β-spirost-25(27)-en-1β,3β-diol- 3-O-β-D-glucopyranosyl-(1 → 4)-β-D-glucopyranoside (17), (25R)-5β-spirostan- 1β,3β-diol-3-O-β-D-glucopyranosyl-(1 → 6)-β-D-glucopyranoside (18), (25R)-5β- spirostan-1β,3β-diol-3-O-β-D-fructofuranosyl-(2 → 6)-β-D-glucopyranoside (19), 5β-spirost-25(27)-en-3β-ol-3-O-β-D-glucopyranosyl-(1 → 4)-β-D-glucopyranoside (20). The antiproliferative effects against seven human cancer cell lines and inhibitory activities on nitric oxide (NO) production induced by lipopolysaccharide (LPS) in a macrophage cell line RAW 264.7 were assayed for all the isolated compounds. Compounds 17, 19 and 21 exhibited potential antiproliferative activities against all of human cancer cell lines tested. Compounds 21 showed significant inhibition on NO production with IC50 values of 11.5 μM. These results showed that the spirostanol saponins isolated from the dried rhizomes of T. chinensis have potent antiproliferative and anti-inflammatory activities and T. chinensis might be used as anticancer and.anti-inflammatory supplement.
Collapse
|
14
|
Huang NY, Wang WB, Chen L, Luo HJ, Wang JZ, Deng WQ, Zou K. Design, synthesis and biological evaluation of bisabolangelone oxime derivatives as potassium-competitive acid blockers (P-CABs). Bioorg Med Chem Lett 2016; 26:2268-72. [DOI: 10.1016/j.bmcl.2016.03.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/29/2016] [Accepted: 03/14/2016] [Indexed: 10/22/2022]
|
15
|
Xiang L, Wang Y, Yi X, Zheng G, He X. Bioactive spirostanol saponins from the rhizome of Tupistra chinensis. Steroids 2016; 108:39-46. [PMID: 26898537 DOI: 10.1016/j.steroids.2016.02.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/02/2016] [Accepted: 02/14/2016] [Indexed: 11/24/2022]
Abstract
Phytochemical investigations of the rhizome of Tupistra chinensis led to the isolation of six new spirostanol saponins, one new spirostanol, along with eight known spirostanols. Their chemical structures were elucidated on the basis of spectroscopic and chemical methods, including IR, NMR, MS, and GC analyses. The antiproliferative effects against five human cancer cell lines were assayed for all the isolated compounds. Compounds 8, 12 and 15 showed potent cytotoxic activities against K562 cells. The isolated compounds were evaluated for their inhibitory activities on nitric oxide (NO) production induced by lipopolysaccharide in a macrophage cell line RAW 264.7. Compounds 2 and 12 showed significant inhibition on NO production with IC50 values of 16.1±1.8 and 13.5±1.2 μM, respectively.
Collapse
Affiliation(s)
- Limin Xiang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yihai Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaomin Yi
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Guangji Zheng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiangjiu He
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
16
|
Furospirostanol and spirostanol saponins from the rhizome of Tupistra chinensis and their cytotoxic and anti-inflammatory activities. Tetrahedron 2016. [DOI: 10.1016/j.tet.2015.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
17
|
Luo HJ, Wang JZ, Huang NY, Deng WQ, Zou K. Computational insights into the interaction mechanism of triazolyl substituted tetrahydrobenzofuran derivatives with H(+),K(+)-ATPase at different pH. J Comput Aided Mol Des 2015; 30:27-37. [PMID: 26667240 DOI: 10.1007/s10822-015-9886-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/07/2015] [Indexed: 11/29/2022]
Abstract
The interaction mechanism of triazolyl substituted tetrahydrobenzofuran derivatives (compound 1 (N, N-Dipropyl-1-(2-phenyl-4,5,6,7-tetrahydrobenzofuran-4-yl)-1H-1,2,3-triazole-4-methanamine) and 2 (1-(2-Phenyl-4,5,6,7-tetrahydrobenzofuran-4-yl)-4-(morpholin-4-ylmethyl)-1H-1,2,3-triazole)) with H(+),K(+)-ATPase at different pH were studied by induced-fit docking, QM/MM optimization and MM/GBSA binding free energy calculations of two forms (neutral and protonated form) of compounds. The inhibition activity of compound 1 is measured and almost unchanged at different pH, while the activity of compound 2 increases significantly with pH value decreased. This phenomenon could be explained by their protonated form percentages and the calculated binding free energies of protonated and neutral mixture of compounds at different pH. The binding free energy of protonated form is higher than that of neutral form of compound, and the protonated form could be a powerful inhibitor of H(+),K(+)-ATPase. By the decomposed energy comparisons of residues in binding sites, Asp137 should be the key binding site to protonated form of compound because of the hydrogen bond and electrostatic interactions. These calculation results could help for further rational design of novel H(+),K(+)-ATPase inhibitors.
Collapse
Affiliation(s)
- Hua-Jun Luo
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Science, China Three Gorges University, Yichang, China.
| | - Jun-Zhi Wang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Science, China Three Gorges University, Yichang, China
| | - Nian-Yu Huang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Science, China Three Gorges University, Yichang, China
| | - Wei-Qiao Deng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Science, China Three Gorges University, Yichang, China.,State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Kun Zou
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Science, China Three Gorges University, Yichang, China
| |
Collapse
|