1
|
Kos J, Strharsky T, Tosso R, Gutierrez L, Kos D, Jurica J, Zendulka O, Pes O, Gregorova J, Degotte G, Gonec T, Oravec M, Vojackova V, Krystof V, Cizek A, Francotte P, Frederich M, Jampilek J, Enriz D. Trifluoromethylcinnamanilides - Effective dual inhibitors of Mycobacterium smegmatis and Plasmodium falciparum. Bioorg Chem 2025; 154:107957. [PMID: 39615279 DOI: 10.1016/j.bioorg.2024.107957] [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: 09/24/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 01/15/2025]
Abstract
A series of eighteen new 2-trifluoromethylcinnamanilides (1a-r) were synthesized by microwave synthesis and investigated for their antimycobacterial and antimalarial activities, along with the complementary (2E)-3-[3-(trifluoromethyl)phenyl]-N-arylprop-2-enanilides (2a-r) and (2E)-3-[4-(trifluoromethyl)phenyl]-N-arylprop-2-enanilides (3a-r) prepared earlier. All the compounds were evaluated in vitro against Mycobacterium smegmatis ATCC 700084 and a chloroquine-sensitive strain of Plasmodium falciparum 3D7/MRA-102. The most active compounds against M. smegmatis (MIC values in the range of 1.17-11.1 µM, more effective than rifampicin) were anilides substituted by 3,5-CF3 (1q, 2q, 3q), 4-OCF3 (1k), and 4-CF3 (1j, 2j). The most effective agents against P. falciparum (IC50 values in the range of 0.32-4.5 µM, comparable to chloroquine) were anilides substituted by 3,5-CF3 (1q, 2q, 3q), 2-Br-4-OCF3 (1r), 4-CF3 (1j, 3j), 4-F (2d), 4-Cl (2g), 2-Cl (1e, 2e). A preliminary in vitro cytotoxicity screening was assessed using human leukemic cell lines and human dermal fibroblasts, revealing the toxic effect of 3,5-CF3 substituted anilides. On the other hand, the other investigated agents showed insignificant cytotoxic effects. Stability assays using rat liver microsomes demonstrated that compounds 1r (R = 2-Br-4-OCF3) and 1q (R = 3,5-CF3) are neither metabolized nor affect cytochrome P450 metabolizing capacity in vitro. Furthermore, complex in silico studies were performed - a combined approach (docking/MD simulations/QTAIM calculations) helped to define the molecular interactions that were applied during the binding of active agents and the subsequent inhibition of their molecular targets - InhA (activity against M. smegmatis) and arginase (activity against P. falciparum). In conclusion, promising active agents with dual antimycobacterial and antimalarial effects were identified.
Collapse
Affiliation(s)
- Jiri Kos
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Tomas Strharsky
- Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho 1946/1, 612 00 Brno, Czech Republic.
| | - Rodrigo Tosso
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, IMIBIO-CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina.
| | - Lucas Gutierrez
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, IMIBIO-CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina.
| | - Dominika Kos
- Department of Molecular Pharmacy, Faculty of Pharmacy, Masaryk University, Palackeho 1946/1, 612 00 Brno, Czech Republic.
| | - Jan Jurica
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Ondrej Zendulka
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Ondrej Pes
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Jana Gregorova
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Gilles Degotte
- Renslo's Lab, Department of Pharmaceutical Chemistry, University California San Francisco, 600 16(th) Street, 94143 San Francisco, CA, USA.
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho 1946/1, 612 00 Brno, Czech Republic.
| | - Michal Oravec
- Global Change Research Institute CAS, Belidla 986/4a, 603 00 Brno, Czech Republic.
| | - Veronika Vojackova
- Department of Experimental Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 779 00 Olomouc, Czech Republic.
| | - Vladimir Krystof
- Department of Experimental Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 779 00 Olomouc, Czech Republic.
| | - Alois Cizek
- Department of Infectious Diseases and Microbiology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Palackeho tr. 1946/1, 612 42 Brno, Czech Republic.
| | - Pierre Francotte
- Laboratory of Medicinal Chemistry, CIRM - Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, 4000 Liege, Belgium.
| | - Michel Frederich
- Laboratory of Pharmacognosy, CIRM - Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, 4000 Liege, Belgium.
| | - Josef Jampilek
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 779 00 Olomouc, Czech Republic; Institute of Chemistry, University of Silesia, Szkolna 9, 40-007 Katowice, Poland.
| | - Daniel Enriz
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, IMIBIO-CONICET, Ejército de los Andes 950, 5700 San Luis, Argentina.
| |
Collapse
|
2
|
Zhang G, Liu M, Han Y, Wang Z, Liu W, Zhang Y, Xu J. The role of aldehydes on sulfur based-new particle formation: a theoretical study. RSC Adv 2024; 14:13321-13335. [PMID: 38694968 PMCID: PMC11061877 DOI: 10.1039/d4ra00952e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/17/2024] [Indexed: 05/04/2024] Open
Abstract
Aldehydes play a crucial role in the formation of atmospheric particles, attracting significant attention due to their environmental impact. However, the microscopic mechanisms underlying the formation of aldehyde-involved particles remain uncertain. In this study, through quantum chemical calculations and molecular dynamics (MD) simulations, we investigate the microscopic formation mechanisms of binary and ternary systems composed of three representative aldehydes, two sulfur-based acids, water, and two bases. Our research findings reveal that the most stable structures of acid-aldehyde clusters involve the connection of acids and aldehyde compounds through hydrogen bonds without involving proton transfer reactions, indicating relatively poor cluster stability. However, with the introduction of a third component, the stability of 18 clusters significantly increase. Among these, in ten systems, acids act as catalysts, facilitating reactions between aldehyde compounds and water or alkaline substances to generate glycols and amino alcohols. However, according to MD simulations conducted at 300 K, these acids readily dissociate from the resulting products. In the remaining eight systems, the most stable structural feature involves ion pairs formed by proton transfer reactions between acids and aldehyde compounds. These clusters exhibit remarkable thermodynamic stability. Furthermore, the acidity of the acid, the nature of nucleophilic agents, and the type of aldehyde all play significant roles in cluster stability and reactivity, and they have synergistic effects on the nucleation process. This study offers microscopic insights into the processes of new particle formation involving aldehydes, contributing to a deeper understanding of atmospheric chemistry at the molecular level.
Collapse
Affiliation(s)
- Guohua Zhang
- Jinhua Advanced Research Institute Jinhua Zhejiang 321013 P. R. China
| | - Min Liu
- Department of Optical Engineering, College of Optical, Mechanical and Electrical Engineering, Zhejiang A&F University Hangzhou Zhejiang 311300 P. R. China
| | - Yaning Han
- Department of Optical Engineering, College of Optical, Mechanical and Electrical Engineering, Zhejiang A&F University Hangzhou Zhejiang 311300 P. R. China
| | - Zhongteng Wang
- Department of Optical Engineering, College of Optical, Mechanical and Electrical Engineering, Zhejiang A&F University Hangzhou Zhejiang 311300 P. R. China
| | - Wei Liu
- Department of Optical Engineering, College of Optical, Mechanical and Electrical Engineering, Zhejiang A&F University Hangzhou Zhejiang 311300 P. R. China
| | - Ying Zhang
- Jinhua Advanced Research Institute Jinhua Zhejiang 321013 P. R. China
| | - Jing Xu
- Department of Optical Engineering, College of Optical, Mechanical and Electrical Engineering, Zhejiang A&F University Hangzhou Zhejiang 311300 P. R. China
| |
Collapse
|
3
|
Isert C, Atz K, Riniker S, Schneider G. Exploring protein-ligand binding affinity prediction with electron density-based geometric deep learning. RSC Adv 2024; 14:4492-4502. [PMID: 38312732 PMCID: PMC10835705 DOI: 10.1039/d3ra08650j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Rational structure-based drug design relies on accurate predictions of protein-ligand binding affinity from structural molecular information. Although deep learning-based methods for predicting binding affinity have shown promise in computational drug design, certain approaches have faced criticism for their potential to inadequately capture the fundamental physical interactions between ligands and their macromolecular targets or for being susceptible to dataset biases. Herein, we propose to include bond-critical points based on the electron density of a protein-ligand complex as a fundamental physical representation of protein-ligand interactions. Employing a geometric deep learning model, we explore the usefulness of these bond-critical points to predict absolute binding affinities of protein-ligand complexes, benchmark model performance against existing methods, and provide a critical analysis of this new approach. The models achieved root-mean-squared errors of 1.4-1.8 log units on the PDBbind dataset, and 1.0-1.7 log units on the PDE10A dataset, not indicating significant advantages over benchmark methods, and thus rendering the utility of electron density for deep learning models context-dependent. The relationship between intermolecular electron density and corresponding binding affinity was analyzed, and Pearson correlation coefficients r > 0.7 were obtained for several macromolecular targets.
Collapse
Affiliation(s)
- Clemens Isert
- ETH Zurich, Department of Chemistry and Applied Biosciences Vladimir-Prelog-Weg 4 8093 Zurich Switzerland +41 44 633 73 27
| | - Kenneth Atz
- ETH Zurich, Department of Chemistry and Applied Biosciences Vladimir-Prelog-Weg 4 8093 Zurich Switzerland +41 44 633 73 27
| | - Sereina Riniker
- ETH Zurich, Department of Chemistry and Applied Biosciences Vladimir-Prelog-Weg 4 8093 Zurich Switzerland +41 44 633 73 27
| | - Gisbert Schneider
- ETH Zurich, Department of Chemistry and Applied Biosciences Vladimir-Prelog-Weg 4 8093 Zurich Switzerland +41 44 633 73 27
| |
Collapse
|
4
|
Lefrancois-Gagnon KM, Mawhinney RC. Toward Universal Substituent Constants: Relating QTAIM Functional Group Descriptors to Substituent Effect Proxies. J Chem Inf Model 2023; 63:6068-6080. [PMID: 37729015 DOI: 10.1021/acs.jcim.3c00987] [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: 09/22/2023]
Abstract
Substituents modulate reactions, but their effects are commonly described by using proxies to their functional group properties. Substituent descriptors from the quantum theory of atoms in molecules, which are true functional group properties, are related here to these proxies, which have historically had chemically relevant meaning. Due to the large number of descriptors, multivariate analysis is used to intuit their significance. Multiple linear regression, principal component, and partial least squares regression analyses highlight that these substituent descriptors contain similar information to the proxies while being intrinsic, predictable substituent properties. Sources of error limiting quantitative reproduction of the proxy data include transferability, experimental accuracy, and solvation issues.
Collapse
Affiliation(s)
| | - Robert C Mawhinney
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario P7B 5E1, Canada
| |
Collapse
|
5
|
Soofi A, Rezaei-Tavirani M, Safari-Alighiarloo N. In silico screening of inhibitors against human dihydrofolate reductase to identify potential anticancer compounds. J Biomol Struct Dyn 2023; 41:14497-14509. [PMID: 36883866 DOI: 10.1080/07391102.2023.2183038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 02/14/2023] [Indexed: 03/09/2023]
Abstract
In all species, dihydrofolate reductase (DHFR) is an essential enzyme that regulates the cellular amount of tetrahydrofolate. Human DHFR (hDHFR) activity inhibition results in tetrahydrofolate depletion and cell death. This property has made hDHFR a therapeutic target for cancer. Methotrexate is a well-known hDHFR inhibitor, but its administration has shown some light to severe adverse effects. Therefore, we aimed to find new potential hDHFR inhibitors using structure-based virtual screening, ADMET prediction, molecular docking, and molecular dynamics simulations. Here, we used the PubChem database to find all compounds with at least 90% structural similarity to known natural DHFR inhibitors. To explore their interaction pattern and estimate their binding affinities, the screened compounds (2023) were subjected to structure-based molecular docking against hDHFR. The fifteen compounds that showed higher binding affinity to the hDHFR than the reference compound (methotrexate) displayed important molecular orientation and interactions with key residues in the enzyme's active site. These compounds were subjected to Lipinski and ADMET prediction. PubChem CIDs: 46886812 and 638190 were identified as putative inhibitors. In addition, molecular dynamics simulations revealed that the binding of compounds (CIDs: 46886812 and 63819) stabilized the hDHFR structure and caused minor conformational changes. Our findings suggest that two compounds (CIDs: 46886812 and 63819) could be promising potential inhibitors of hDHFR in cancer therapy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Asma Soofi
- Department of Physical Chemistry, School of Chemistry, College of Sciences, University of Tehran, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nahid Safari-Alighiarloo
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Zhang Y, Luo M, Wu P, Wu S, Lee TY, Bai C. Application of Computational Biology and Artificial Intelligence in Drug Design. Int J Mol Sci 2022; 23:13568. [PMID: 36362355 PMCID: PMC9658956 DOI: 10.3390/ijms232113568] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 08/24/2023] Open
Abstract
Traditional drug design requires a great amount of research time and developmental expense. Booming computational approaches, including computational biology, computer-aided drug design, and artificial intelligence, have the potential to expedite the efficiency of drug discovery by minimizing the time and financial cost. In recent years, computational approaches are being widely used to improve the efficacy and effectiveness of drug discovery and pipeline, leading to the approval of plenty of new drugs for marketing. The present review emphasizes on the applications of these indispensable computational approaches in aiding target identification, lead discovery, and lead optimization. Some challenges of using these approaches for drug design are also discussed. Moreover, we propose a methodology for integrating various computational techniques into new drug discovery and design.
Collapse
Affiliation(s)
- Yue Zhang
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
- Warshel Institute for Computational Biology, Shenzhen 518172, China
| | - Mengqi Luo
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Peng Wu
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Song Wu
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Tzong-Yi Lee
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Warshel Institute for Computational Biology, Shenzhen 518172, China
| | - Chen Bai
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Warshel Institute for Computational Biology, Shenzhen 518172, China
| |
Collapse
|
7
|
Love O, Pacheco Lima MC, Clark C, Cornillie S, Roalstad S, Cheatham TE. Evaluating the accuracy of the AMBER protein force fields in modeling dihydrofolate reductase structures: misbalance in the conformational arrangements of the flexible loop domains. J Biomol Struct Dyn 2022:1-15. [PMID: 35838167 PMCID: PMC9840716 DOI: 10.1080/07391102.2022.2098823] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Protein flexible loop regions were once thought to be simple linkers between other more functional secondary structural elements. However, as it becomes clearer that these loop domains are critical players in a plethora of biological processes, accurate conformational sampling of 3D loop structures is vital to the advancement of drug design techniques and the overall growth of knowledge surrounding molecular systems. While experimental techniques provide a wealth of structural information, the resolution of flexible loop domains is sometimes low or entirely absent due to their complex and dynamic nature. This highlights an opportunity for de novo structure prediction using in silico methods with molecular dynamics (MDs). This study evaluates some of the AMBER protein force field's (ffs) ability to accurately model dihydrofolate reductase (DHFR) conformations, a protein complex characterized by specific arrangements and interactions of multiple flexible loops whose conformations are determined by the presence or absence of bound ligands and cofactors. Although the AMBER ffs, including ff19SB, studied well model most protein structures with rich secondary structure, results obtained here suggest the inability to significantly sample the expected DHFR loop-loop conformations - of the six distinct protein-ligand systems simulated, a majority lacked consistent stabilization of experimentally derived metrics definitive the three enzyme conformations. Although under-sampling and the chosen ff parameter combinations could be the cause, given past successes with these MD approaches for many protein systems, this suggests a potential misbalance in available ff parameters required to accurately predict the structure of multiple flexible loop regions present in proteins.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Olivia Love
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | | | - Casey Clark
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Sean Cornillie
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Shelly Roalstad
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Thomas E. Cheatham
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
8
|
Liu M, Myllys N, Han Y, Wang Z, Chen L, Liu W, Xu J. Microscopic Insights Into the Formation of Methanesulfonic Acid–Methylamine–Ammonia Particles Under Acid-Rich Conditions. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.875585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Understanding the microscopic mechanisms of new particle formation under acid-rich conditions is of significance in atmospheric science. Using quantum chemistry calculations, we investigated the microscopic formation mechanism of methanesulfonic acid (MSA)–methylamine (MA)–ammonia (NH3) clusters. We focused on the binary (MSA)2n-(MA)n and ternary (MSA)3n-(MA)n-(NH3)n, (n = 1–4) systems which contain more acid than base molecules. We found that the lowest-energy isomers in each system possess considerable thermodynamic and dynamic stabilities. In studied cluster structures, all bases are protonated, and they form stable ion pairs with MSA, which contribute to the charge transfer and the stability of clusters. MA and NH3 have a synergistic effect on NPF under acid-rich conditions, and the role of NH3 becomes more remarkable as cluster size increases. The excess of MSA molecules does not only enhance the stability of clusters, but provides potential sites for further growth.
Collapse
|
9
|
Abdolmaleki B, Maddah M. Screening of indole derivatives as the potent anticancer agents on dihydrofolate reductase: pharmaco-informatics and molecular dynamics simulation. J Biomol Struct Dyn 2022; 41:3667-3679. [PMID: 35318890 DOI: 10.1080/07391102.2022.2053745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Dihydrofolate reductase (DHFR) is a ubiquitous cellular enzyme involved in the biosynthesis of nucleotide and protein precursors, thus, the inhibition of human DHFR can be a promising strategy in cancer treatment. The design of effective anticancer drugs is an urgent need today according to the high spread of cancer. The indole molecule with diverse mechanisms of action and anticancer properties is one of the efficient pharmacophores in drug design. Hence, a virtual library of indole derivatives as a scaffold was selected for designing safer and more effective anticancer drugs against DHFR in this work. All indole derivatives utilized in the library design were selected regarding appreciable tumor growth inhibition. Structure-activity relationship (SAR), docking energy, ADMET (absorption, distribution, metabolism, excretion, and toxicity) parameters, and effective non-covalent interactions were used to identify potential anticancer with indole scaffold. Results showed a higher number of indole moieties provide a strong attachment to the DHFR binding pocket and therefore more effective anticancer activity. The indole scaffold in combination with dichlorobenzene improves DHFR inhibition whereas barbituric acid weakens inhibition activity. In the following to validate the docking results, Molecular dynamics (MD) simulation and molecular mechanics generalized-Born surface area (MM-GBSA) indicated the permanent stability of the selected ligands into the DHFR binding pocket and the key amino acids. Therefore, promising pharmacophores based on indole-DHFR interactions were discovered, and the outcome could be useful in guiding future in vitro and in vivo drug discovery in cancer medicine.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Behnaz Abdolmaleki
- Department of Chemistry, K.N.Toosi University of Technology, Tehran, Iran
| | - Mina Maddah
- Department of Chemistry, K.N.Toosi University of Technology, Tehran, Iran.,Super Computing Institute, University of Tehran, Tehran, Iran
| |
Collapse
|
10
|
Emam AM, Dahal A, Singh SS, Tosso RD, Ibrahim SM, El-Sadek M, Jois SD, Enriz RD, Kothayer H. Quinazoline-tethered hydrazone: A versatile scaffold toward dual anti-TB and EGFR inhibition activities in NSCLC. Arch Pharm (Weinheim) 2021; 354:e2100281. [PMID: 34585758 DOI: 10.1002/ardp.202100281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 11/07/2022]
Abstract
Globally, lung cancer and tuberculosis are considered to be very serious and complex diseases. Evidence suggests that chronic infection with tuberculosis (TB) can often lead to lung tumors; therefore, developing drugs that target both diseases is of great clinical significance. In our study, we designed and synthesized a suite of 14 new quinazolinones (5a-n) and performed biological investigations of these compounds in Mycobacterium tuberculosis (MTB) and cancer cell lines. In addition, we conducted a molecular modeling study to determine the mechanism of action of these compounds at the molecular level. Compounds that showed anticancer activity in the preliminary screening were further evaluated in three cancer cell lines (A549, Calu-3, and BT-474 cells) and characterized in an epidermal growth factor receptor (EGFR) binding assay. Cytotoxicity in noncancerous lung fibroblast cells was also evaluated to obtain safety data. Our theoretical and experimental studies indicated that our compounds showed a mechanism of action similar to that of erlotinib by inhibiting the EGFR tyrosine kinase. In turn, the antituberculosis activity of these compounds would be produced by the inhibition of enoyl-ACP-reductase. From our findings, we were able to identify two potential lead compounds (5i and 5l) with dual activity and elevated safety toward noncancerous lung fibroblast cells. In addition, our data identified three compounds with excellent anti-TB activities (compounds 5i, 5l, and 5n).
Collapse
Affiliation(s)
- Aya M Emam
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Rodrigo D Tosso
- Pharmacy Department, Facultad de Química, Bioquímica y Farmacia, IMIBIO-CONICET, Universidad Nacional de San Luis, San Luis, Argentina
| | - Samy M Ibrahim
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed El-Sadek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Ricardo D Enriz
- Pharmacy Department, Facultad de Química, Bioquímica y Farmacia, IMIBIO-CONICET, Universidad Nacional de San Luis, San Luis, Argentina
| | - Hend Kothayer
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
11
|
Magar P, Parravicini O, Štěpánková Š, Svrčková K, Garro AD, Jendrzejewska I, Pauk K, Hošek J, Jampílek J, Enriz RD, Imramovský A. Novel Sulfonamide-Based Carbamates as Selective Inhibitors of BChE. Int J Mol Sci 2021; 22:9447. [PMID: 34502357 PMCID: PMC8430704 DOI: 10.3390/ijms22179447] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/28/2021] [Indexed: 11/16/2022] Open
Abstract
A series of 14 target benzyl [2-(arylsulfamoyl)-1-substituted-ethyl]carbamates was prepared by multi-step synthesis and characterized. All the final compounds were tested for their ability to inhibit acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) in vitro, and the selectivity index (SI) was determined. Except for three compounds, all compounds showed strong preferential inhibition of BChE, and nine compounds were even more active than the clinically used rivastigmine. Benzyl {(2S)-1-[(2-methoxybenzyl)sulfamoyl]-4-methylpentan-2-yl}carbamate (5k), benzyl {(2S)-1-[(4-chlorobenzyl)sulfamoyl]-4-methylpentan-2-yl}carbamate (5j), and benzyl [(2S)-1-(benzylsulfamoyl)-4-methylpentan-2-yl]carbamate (5c) showed the highest BChE inhibition (IC50 = 4.33, 6.57, and 8.52 µM, respectively), indicating that derivatives 5c and 5j had approximately 5-fold higher inhibitory activity against BChE than rivastigmine, and 5k was even 9-fold more effective than rivastigmine. In addition, the selectivity index of 5c and 5j was approx. 10 and that of 5k was even 34. The process of carbamylation and reactivation of BChE was studied for the most active derivatives 5k, 5j. The detailed information about the mode of binding of these compounds to the active site of both BChE and AChE was obtained in a molecular modeling study. In this study, combined techniques (docking, molecular dynamic simulations, and QTAIM (quantum theory of atoms in molecules) calculations) were employed.
Collapse
Affiliation(s)
- Pratibha Magar
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (P.M.); (K.P.)
| | - Oscar Parravicini
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina; (O.P.); (A.D.G.)
| | - Šárka Štěpánková
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (Š.Š.); (K.S.)
| | - Katarina Svrčková
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (Š.Š.); (K.S.)
| | - Adriana D. Garro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina; (O.P.); (A.D.G.)
| | | | - Karel Pauk
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (P.M.); (K.P.)
| | - Jan Hošek
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic;
| | - Josef Jampílek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia;
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| | - Ricardo D. Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina; (O.P.); (A.D.G.)
| | - Aleš Imramovský
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10 Pardubice, Czech Republic; (P.M.); (K.P.)
| |
Collapse
|
12
|
Structure activity relationships and the binding mode of quinolinone-pyrimidine hybrids as reversal agents of multidrug resistance mediated by P-gp. Sci Rep 2021; 11:16856. [PMID: 34413359 PMCID: PMC8376931 DOI: 10.1038/s41598-021-96226-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
P-gp-associated multidrug resistance is a major impediment to the success of chemotherapy. With the aim of finding non-toxic and effective P-gp inhibitors, we investigated a panel of quinolin-2-one-pyrimidine hybrids. Among the active compounds, two of them significantly increased intracellular doxorubicin and rhodamine 123 accumulation by inhibiting the efflux mediated by P-gp and restored doxorubicin toxicity at nanomolar range. Structure-activity relationships showed that the number of methoxy groups, an optimal length of the molecule in its extended conformation, and at least one flexible methylene group bridging the quinolinone to the moiety bearing the pyrimidine favored the inhibitory potency of P-gp. The best compounds showed a similar binding pattern and interactions to those of doxorubicin and tariquidar, as revealed by MD and hybrid QM/MM simulations performed with the recent experimental structure of P-gp co-crystallized with paclitaxel. Analysis of the molecular interactions stabilizing the different molecular complexes determined by MD and QTAIM showed that binding to key residues from TMH 4-7 and 12 is required for inhibition.
Collapse
|
13
|
Wróbel A, Baradyn M, Ratkiewicz A, Drozdowska D. Synthesis, Biological Activity, and Molecular Dynamics Study of Novel Series of a Trimethoprim Analogs as Multi-Targeted Compounds: Dihydrofolate Reductase (DHFR) Inhibitors and DNA-Binding Agents. Int J Mol Sci 2021; 22:3685. [PMID: 33916202 PMCID: PMC8037161 DOI: 10.3390/ijms22073685] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 01/07/2023] Open
Abstract
Eighteen previously undescribed trimethoprim (TMP) analogs containing amide bonds (1-18) were synthesized and compared with TMP, methotrexate (MTX), and netropsin (NT). These compounds were designed as potential minor groove binding agents (MGBAs) and inhibitors of human dihydrofolate reductase (hDHFR). The all-new derivatives were obtained via solid phase synthesis using 4-nitrophenyl Wang resin. Data from the ethidium displacement test confirmed their DNA-binding capacity. Compounds 13-14 (49.89% and 43.85%) and 17-18 (41.68% and 42.99%) showed a higher binding affinity to pBR322 plasmid than NT. The possibility of binding in a minor groove as well as determination of association constants were performed using calf thymus DNA, T4 coliphage DNA, poly (dA-dT)2, and poly (dG-dC)2. With the exception of compounds 9 (IC50 = 56.05 µM) and 11 (IC50 = 55.32 µM), all of the compounds showed better inhibitory properties against hDHFR than standard, which confirms that the addition of the amide bond into the TMP structures increases affinity towards hDHFR. Derivatives 2, 6, 13, 14, and 16 were found to be the most potent hDHFR inhibitors. This molecular modelling study shows that they interact strongly with a catalytically important residue Glu-30.
Collapse
Affiliation(s)
- Agnieszka Wróbel
- Department of Organic Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Maciej Baradyn
- Department of Physical Chemistry, Institute of Chemistry, University of Bialystok, 15-245 Bialystok, Poland; (M.B.); (A.R.)
| | - Artur Ratkiewicz
- Department of Physical Chemistry, Institute of Chemistry, University of Bialystok, 15-245 Bialystok, Poland; (M.B.); (A.R.)
| | - Danuta Drozdowska
- Department of Organic Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland;
| |
Collapse
|
14
|
Suruzhon M, Bodnarchuk MS, Ciancetta A, Viner R, Wall ID, Essex JW. Sensitivity of Binding Free Energy Calculations to Initial Protein Crystal Structure. J Chem Theory Comput 2021; 17:1806-1821. [PMID: 33534995 DOI: 10.1021/acs.jctc.0c00972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Binding free energy calculations using alchemical free energy (AFE) methods are widely considered to be the most rigorous tool in the computational drug discovery arsenal. Despite this, the calculations suffer from accuracy, precision, and reproducibility issues. In this publication, we perform a high-throughput study of more than a thousand AFE calculations, utilizing over 220 μs of total sampling time, on three different protein systems to investigate the impact of the initial crystal structure on the resulting binding free energy values. We also consider the influence of equilibration time and discover that the initial crystal structure can have a significant effect on free energy values obtained at short timescales that can manifest itself as a free energy difference of more than 1 kcal/mol. At longer timescales, these differences are largely overtaken by important rare events, such as torsional ligand motions, typically resulting in a much higher uncertainty in the obtained values. This work emphasizes the importance of rare event sampling and long-timescale dynamics in free energy calculations even for routinely performed alchemical perturbations. We conclude that an optimal protocol should not only concentrate computational resources on achieving convergence in the alchemical coupling parameter (λ) space but also on longer simulations and multiple repeats.
Collapse
Affiliation(s)
- Miroslav Suruzhon
- School of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, U.K
| | | | | | - Russell Viner
- Syngenta, Jealott's Hill International Research Centre, Bracknell RG42 6EY, U.K
| | - Ian D Wall
- GSK Medicines Research Centre, Gunnels Wood Road, Stevenage SG1 2NY, U.K
| | - Jonathan W Essex
- School of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, U.K
| |
Collapse
|
15
|
A combined structure-based pharmacophore modeling and 3D-QSAR study on a series of N-heterocyclic scaffolds to screen novel antagonists as human DHFR inhibitors. Struct Chem 2021. [DOI: 10.1007/s11224-020-01705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
16
|
Rojas S, Parravicini O, Vettorazzi M, Tosso R, Garro A, Gutiérrez L, Andújar S, Enriz R. Combined MD/QTAIM techniques to evaluate ligand-receptor interactions. Scope and limitations. Eur J Med Chem 2020; 208:112792. [PMID: 32949964 DOI: 10.1016/j.ejmech.2020.112792] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/29/2022]
Abstract
In medicinal chemistry, it is extremely important to evaluate, as accurately as possible, the molecular interactions involved in the formation of different ligand-receptor (L-R) complexes. Evaluating the different molecular interactions by quantum mechanics calculations is not a simple task, since formation of an L-R complex is a dynamic process. In this case, the use of combined techniques of molecular dynamics (MD) and quantum calculations is one the best possible approaches. In this work we report a comparative study using combined MD and QTAIM (Quantum Theory of Atoms In Molecules) calculations for five biological systems with different levels of structural complexity. We have studied Acetylcholinesterase (AChE), D2 Dopamine Receptor (D2DR), beta Secretase (BACE1), Dihydrofolate Reductase (DHFR) and Sphingosine Kinase 1 (SphK1). In these molecular targets, we have analyzed different ligands with diverse structural characteristics. The inhibitory activities of most of them have been previously measured in our laboratory. Our results indicate that QTAIM calculations can be extremely useful for in silico studies. It is possible to obtain very accurate information about the strength of the molecular interactions that stabilize the formation of the different L-R complexes. Better correlations can be obtained between theoretical and experimental data by using QTAIM calculations, allowing us to discriminate among ligands with similar affinities. QTAIM analysis gives fairly accurate information for weak interactions which are not well described by MD simulations. QTAIM study also allowed us to evaluate and determine which parts of the ligand need to be modified in order to increase its interactions with the molecular target. In this study we have discussed the importance of combined MD/QTAIM calculations for this type of simulations, showing their scopes and limitations.
Collapse
Affiliation(s)
- Sebastián Rojas
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Oscar Parravicini
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Marcela Vettorazzi
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Rodrigo Tosso
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Adriana Garro
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Lucas Gutiérrez
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Sebastián Andújar
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina
| | - Ricardo Enriz
- IMIBIO-SL CONICET, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes 950, 5700, San Luis, Argentina.
| |
Collapse
|
17
|
Campos LE, Garibotto F, Angelina E, Kos J, Gonec T, Marvanova P, Vettorazzi M, Oravec M, Jendrzejewska I, Jampilek J, Alvarez SE, Enriz RD. Hydroxynaphthalenecarboxamides and substituted piperazinylpropandiols, two new series of BRAF inhibitors. A theoretical and experimental study. Bioorg Chem 2020; 103:104145. [PMID: 32801082 DOI: 10.1016/j.bioorg.2020.104145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/17/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022]
Abstract
The oncogenic mutated kinase BRAFV600E is an attractive molecular target because it is expressed in several human cancers, including melanoma. To present, only three BRAF small inhibitors are approved by the FDA for the treatment of patients with metastatic melanoma: Vemurafenib, Dabrafenib and Encorafenib. Although many protocol treatments have been probed in clinical trials, BRAF inhibition has a limited effectiveness because patients invariably develop resistance and secondary toxic effects associated with the therapy. These limitations highlight the importance of designing new and better inhibitors with different structures that could establish different interactions in the active site of the enzyme and therefore decrease resistance progress. Considering the data from our previous report, here we studied two series of derivatives of structural scaffolds as potential BRAF inhibitors: hydroxynaphthalenecarboxamides and substituted piperazinylpropandiols. Our results indicate that structural analogues of substituted piperazinylpropandiols do not show significantly better activities to that previously reported. In contrast, the hydroxynaphthalenecarboxamides derivatives significantly inhibited cell viability and ERK phosphorylation, a measure of BRAF activity, in Lu1205 BRAFV600E melanoma cells. In order to better understand these experimental results, we carried out a molecular modeling study using different combined techniques: docking, MD simulations and quantum theory of atoms in molecules (QTAIM) calculations. Thus, by using this approach we determined that the molecular interactions that stabilize the different molecular complexes are closely related to Vemurafenib, a well-documented BRAF inhibitor. Furthermore, we found that bi-substituted compounds may interact more strongly respect to the mono-substituted analogues, by establishing additional interactions with the DFG-loop at the BRAF-active site. On the bases of these results we synthesized and tested a new series of hydroxynaphthalenecarboxamides bi-substituted. Remarkably, all these compounds displayed significant inhibitory effects on the bioassays performed. Thus, the structural information reported here is important for the design of new BRAFV600E inhibitors possessing this type of structural scaffold.
Collapse
Affiliation(s)
- Ludmila E Campos
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Francisco Garibotto
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Química, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Avda. Libertad 5460, (3400) Corrientes, Argentina
| | - Jiri Kos
- Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho 1, 612 00 Brno, Czech Republic
| | - Pavlina Marvanova
- Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho 1, 612 00 Brno, Czech Republic
| | - Marcela Vettorazzi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Michal Oravec
- Global Change Research Institute CAS, Belidla 986/4a, 603 00 Brno, Czech Republic
| | | | - Josef Jampilek
- Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Sergio E Alvarez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina.
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina.
| |
Collapse
|
18
|
Hassanzadeh P. Towards the quantum-enabled technologies for development of drugs or delivery systems. J Control Release 2020; 324:260-279. [DOI: 10.1016/j.jconrel.2020.04.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022]
|
19
|
Arylaminopropanone Derivatives as Potential Cholinesterase Inhibitors: Synthesis, Docking Study and Biological Evaluation. Molecules 2020; 25:molecules25071751. [PMID: 32290227 PMCID: PMC7180927 DOI: 10.3390/molecules25071751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 11/18/2022] Open
Abstract
Neurodegenerative diseases in which the decrease of the acetylcholine is observed are growing worldwide. In the present study, a series of new arylaminopropanone derivatives with N-phenylcarbamate moiety (1–16) were prepared as potential acetylcholinesterase and butyrylcholinesterase inhibitors. In vitro enzyme assays were performed; the results are expressed as a percentage of inhibition and the IC50 values. The inhibitory activities were compared with reference drugs galantamine and rivastigmine showing piperidine derivatives (1–3) as the most potent. A possible mechanism of action for these compounds was determined from a molecular modelling study by using combined techniques of docking, molecular dynamics simulations and quantum mechanics calculations.
Collapse
|
20
|
Understanding the Pyrimethamine Drug Resistance Mechanism via Combined Molecular Dynamics and Dynamic Residue Network Analysis. Molecules 2020; 25:molecules25040904. [PMID: 32085470 PMCID: PMC7070769 DOI: 10.3390/molecules25040904] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
In this era of precision medicine, insights into the resistance mechanism of drugs are integral for the development of potent therapeutics. Here, we sought to understand the contribution of four point mutations (N51I, C59R, S108N, and I164L) within the active site of the malaria parasite enzyme dihydrofolate reductase (DHFR) towards the resistance of the antimalarial drug pyrimethamine. Homology modeling was used to obtain full-length models of wild type (WT) and mutant DHFR. Molecular docking was employed to dock pyrimethamine onto the generated structures. Subsequent all-atom molecular dynamics (MD) simulations and binding free-energy computations highlighted that pyrimethamine's stability and affinity inversely relates to the number of mutations within its binding site and, hence, resistance severity. Generally, mutations led to reduced binding affinity to pyrimethamine and increased conformational plasticity of DHFR. Next, dynamic residue network analysis (DRN) was applied to determine the impact of mutations and pyrimethamine binding on communication dispositions of DHFR residues. DRN revealed residues with distinctive communication profiles, distinguishing WT from drug-resistant mutants as well as pyrimethamine-bound from pyrimethamine-free models. Our results provide a new perspective on the understanding of mutation-induced drug resistance.
Collapse
|
21
|
Second-generation 4,5,6,7-tetrahydrobenzo[ d]thiazoles as novel DNA gyrase inhibitors. Future Med Chem 2020; 12:277-297. [PMID: 32043377 DOI: 10.4155/fmc-2019-0127] [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] [Indexed: 12/14/2022] Open
Abstract
Aim: DNA gyrase and topoisomerase IV are essential bacterial enzymes, and in the fight against bacterial resistance, they are important targets for the development of novel antibacterial drugs. Results: Building from our first generation of 4,5,6,7-tetrahydrobenzo[d]thiazole-based DNA gyrase inhibitors, we designed and prepared an optimized series of analogs that show improved inhibition of DNA gyrase and topoisomerase IV from Staphylococcus aureus and Escherichia coli, with IC50 values in the nanomolar range. Importantly, these inhibitors also show improved antibacterial activity against Gram-positive strains. Conclusion: The most promising inhibitor, 29, is active against Enterococcus faecalis, Enterococcus faecium and S. aureus wild-type and resistant strains, with minimum inhibitory concentrations between 4 and 8 μg/ml, which represents good starting point for development of novel antibacterials.
Collapse
|
22
|
Santos de Oliveira FL, Vieira Carletti J, Azevedo FFN, Freitas de Sousa FJ, Caetano EWS, Freire VN, Zanatta G. mTOR–mLST8 interaction: hot spot identification through quantum biochemistry calculations. NEW J CHEM 2020. [DOI: 10.1039/d0nj04099a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Quantum calculation of mTOR–mLST8 interaction.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Geancarlo Zanatta
- Department of Physics at Federal University of Ceará
- 60455-760 Fortaleza
- Brazil
- Postgraduate Research Program in Biochemistry at Federal University of Ceará
- Fortaleza
| |
Collapse
|
23
|
Luchi A, Villafañe RN, Gómez Chávez JL, Bogado ML, Angelina EL, Peruchena NM. Combining Charge Density Analysis with Machine Learning Tools To Investigate the Cruzain Inhibition Mechanism. ACS OMEGA 2019; 4:19582-19594. [PMID: 31788588 PMCID: PMC6881835 DOI: 10.1021/acsomega.9b01934] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/18/2019] [Indexed: 05/28/2023]
Abstract
Trypanosoma cruzi, a flagellate protozoan parasite, is responsible for Chagas disease. The parasite major cysteine protease, cruzain (Cz), plays a vital role at every stage of its life cycle and the active-site region of the enzyme, similar to those of other members of the papain superfamily, is well characterized. Taking advantage of structural information available in public databases about Cz bound to known covalent inhibitors, along with their corresponding activity annotations, in this work, we performed a deep analysis of the molecular interactions at the Cz binding cleft, in order to investigate the enzyme inhibition mechanism. Our toolbox for performing this study consisted of the charge density topological analysis of the complexes to extract the molecular interactions and machine learning classification models to relate the interactions with biological activity. More precisely, such a combination was useful for the classification of molecular interactions as "active-like" or "inactive-like" according to whether they are prevalent in the most active or less active complexes, respectively. Further analysis of interactions with the help of unsupervised learning tools also allowed the understanding of how these interactions come into play together to trigger the enzyme into a particular conformational state. Most active inhibitors induce some conformational changes within the enzyme that lead to an overall better fit of the inhibitor into the binding cleft. Curiously, some of these conformational changes can be considered as a hallmark of the substrate recognition event, which means that most active inhibitors are likely recognized by the enzyme as if they were its own substrate so that the catalytic machinery is arranged as if it is about to break the substrate scissile bond. Overall, these results contribute to a better understanding of the enzyme inhibition mechanism. Moreover, the information about main interactions extracted through this work is already being used in our lab to guide docking solutions in ongoing prospective virtual screening campaigns to search for novel noncovalent cruzain inhibitors.
Collapse
|
24
|
Campos LE, Garibotto FM, Angelina E, Kos J, Tomašič T, Zidar N, Kikelj D, Gonec T, Marvanova P, Mokry P, Jampilek J, Alvarez SE, Enriz RD. Searching new structural scaffolds for BRAF inhibitors. An integrative study using theoretical and experimental techniques. Bioorg Chem 2019; 91:103125. [PMID: 31401373 DOI: 10.1016/j.bioorg.2019.103125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 01/12/2023]
Abstract
The identification of the V600E activating mutation in the protein kinase BRAF in around 50% of melanoma patients has driven the development of highly potent small inhibitors (BRAFi) of the mutated protein. To date, Dabrafenib and Vemurafenib, two specific BRAFi, have been clinically approved for the treatment of metastatic melanoma. Unfortunately, after the initial response, tumors become resistant and patients develop a progressive and lethal disease, making imperative the development of new therapeutic options. The main objective of this work was to find new BRAF inhibitors with different structural scaffolds than those of the known inhibitors. Our study was carried out in different stages; in the first step we performed a virtual screening that allowed us to identify potential new inhibitors. In the second step, we synthesized and tested the inhibitory activity of the novel compounds founded. Finally, we conducted a molecular modelling study that allowed us to understand interactions at the molecular level that stabilize the formation of the different molecular complexes. Our theoretical and experimental study allowed the identification of four new structural scaffolds, which could be used as starting structures for the design and development of new inhibitors of BRAF. Our experimental data indicate that the most active compounds reduced significantly ERK½ phosphorylation, a measure of BRAF inhibition, and cell viability. Thus, from our theoretical and experimental results, we propose new substituted hydroxynaphthalenecarboxamides, N-(hetero)aryl-piperazinylhydroxyalkylphenylcarbamates, substituted piperazinylethanols and substituted piperazinylpropandiols as initial structures for the development of new inhibitors for BRAF. Moreover, by performing QTAIM analysis, we are able to describe in detail the molecular interactions that stabilize the different Ligand-Receptor complexes. Such analysis indicates which portion of the different molecules must be changed in order to obtain an increase in the binding affinity of these new ligands.
Collapse
Affiliation(s)
- Ludmila E Campos
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Francisco M Garibotto
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Química, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Avda. Libertad 5460, 3400 Corrientes, Argentina
| | - Jiri Kos
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Nace Zidar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Danijel Kikelj
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 61242 Brno, Czech Republic
| | - Pavlina Marvanova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 61242 Brno, Czech Republic
| | - Petr Mokry
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 61242 Brno, Czech Republic
| | - Josef Jampilek
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 78371 Olomouc, Czech Republic; Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 84215 Bratislava, Slovakia
| | - Sergio E Alvarez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina.
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina.
| |
Collapse
|
25
|
Rana RM, Rampogu S, Zeb A, Son M, Park C, Lee G, Yoon S, Baek A, Parameswaran S, Park SJ, Lee KW. In Silico Study Probes Potential Inhibitors of Human Dihydrofolate Reductase for Cancer Therapeutics. J Clin Med 2019; 8:jcm8020233. [PMID: 30754680 PMCID: PMC6406960 DOI: 10.3390/jcm8020233] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 01/10/2023] Open
Abstract
Dihydrofolate reductase (DHFR) is an essential cellular enzyme and thereby catalyzes thereduction of dihydrofolate to tetrahydrofolate (THF). In cancer medication, inhibition of humanDHFR (hDHFR) remains a promising strategy, as it depletes THF and slows DNA synthesis and cellproliferation. In the current study, ligand-based pharmacophore modeling identified and evaluatedthe critical chemical features of hDHFR inhibitors. A pharmacophore model (Hypo1) was generatedfrom known inhibitors of DHFR with a correlation coefficient (0.94), root mean square (RMS)deviation (0.99), and total cost value (125.28). Hypo1 was comprised of four chemical features,including two hydrogen bond donors (HDB), one hydrogen bond acceptor (HBA), and onehydrophobic (HYP). Hypo1 was validated using Fischer's randomization, test set, and decoy setvalidations, employed as a 3D query in a virtual screening at Maybridge, Chembridge, Asinex,National Cancer Institute (NCI), and Zinc databases. Hypo1-retrieved compounds were filtered byan absorption, distribution, metabolism, excretion, and toxicity (ADMET) assessment test andLipinski's rule of five, where the drug-like hit compounds were identified. The hit compounds weredocked in the active site of hDHFR and compounds with Goldfitness score was greater than 44.67(docking score for the reference compound), clustering analysis, and hydrogen bond interactionswere identified. Furthermore, molecular dynamics (MD) simulation identified three compounds asthe best inhibitors of hDHFR with the lowest root mean square deviation (1.2 Å to 1.8 Å), hydrogenbond interactions with hDHFR, and low binding free energy (-127 kJ/mol to -178 kJ/mol). Finally,the toxicity prediction by computer (TOPKAT) affirmed the safety of the novel inhibitors of hDHFRin human body. Overall, we recommend novel hit compounds of hDHFR for cancer and rheumatoidarthritis chemotherapeutics.
Collapse
Affiliation(s)
- Rabia Mukhtar Rana
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Shailima Rampogu
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Amir Zeb
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Minky Son
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Chanin Park
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Gihwan Lee
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Sanghwa Yoon
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Ayoung Baek
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Sarvanan Parameswaran
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| | - Seok Ju Park
- Department of Internal Medicine, College of Medicine, Busan Paik Hospital, Inje University,Busan 47392, Korea.
| | - Keun Woo Lee
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Research Institute of NaturalScience (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea.
| |
Collapse
|
26
|
Indole-substituted 2,4-diamino-5,8-dihydropyrido[2,3-d]pyrimidines from one-pot process and evaluation of their ability to bind dopamine receptors. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.10.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
27
|
Luchi A, Angelina E, Bogado L, Forli S, Olson A, Peruchena N. Flap-site Fragment Restores Back Wild-type Behaviour in Resistant Form of HIV Protease. Mol Inform 2018; 37:e1800053. [PMID: 30051611 DOI: 10.1002/minf.201800053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/11/2018] [Indexed: 11/09/2022]
Abstract
HIV-1 protease (HIV-PR) performs a vital step in the virus life cycle which makes it an excellent target for drug therapy. However, due to the error-prone of HIV reverse transcriptase, mutations in HIV-PR often occur, inducing drug-resistance to inhibitors. Some HIV-PR mutations can make the flaps of the enzyme more flexible thus increasing the flaps opening rate and inhibitor releasing. It has been shown that by targeting novel binding sites on HIV-PR with small molecules, it is possible to alter the equilibrium of flap conformational states. A previous fragment-based crystallographic screen have found two novel binding sites for small fragments in the inhibited, closed form of HIV-PR, termed flap and exo sites. While these experiments were performed in wild type HIV-PR, it still remains to be proven whether these small fragments can stabilize the closed conformation of flaps in resistant forms of the enzyme. Here we performed Molecular Dynamics simulations of wild type and mutant form of HIV-PR bound to inhibitor TL-3. Simulations show that on going from wild type to 6X mutant the equilibrium shifts from closed to semi-open conformation of flaps. However, when fragment Br6 is placed at flap site of mutant form, the enzyme is restored back to closed conformation. This finding supports the hypothesis that allosteric inhibitors, together with active site inhibitors could increase the number of point mutations necessary for appreciable clinical resistance to AIDS therapy.
Collapse
Affiliation(s)
- Adriano Luchi
- Lab. Estructura Molecular y Propiedades, IQUIBA-NEA, Universidad Nacional del Nordeste, CONICET, FACENA, Av. Libertad 5470, Corrientes, 3400, Argentina
| | - Emilio Angelina
- Lab. Estructura Molecular y Propiedades, IQUIBA-NEA, Universidad Nacional del Nordeste, CONICET, FACENA, Av. Libertad 5470, Corrientes, 3400, Argentina
| | - Lucrecia Bogado
- Lab. Estructura Molecular y Propiedades, IQUIBA-NEA, Universidad Nacional del Nordeste, CONICET, FACENA, Av. Libertad 5470, Corrientes, 3400, Argentina
| | - Stefano Forli
- Molecular Graphics Lab, Department of Integrative Structural and Computational Biology, MB-112, the Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, 92037-1000
| | - Arthur Olson
- Molecular Graphics Lab, Department of Integrative Structural and Computational Biology, MB-112, the Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, 92037-1000
| | - Nélida Peruchena
- Lab. Estructura Molecular y Propiedades, IQUIBA-NEA, Universidad Nacional del Nordeste, CONICET, FACENA, Av. Libertad 5470, Corrientes, 3400, Argentina
| |
Collapse
|
28
|
Theoretical models to predict the inhibitory effect of ligands of sphingosine kinase 1 using QTAIM calculations and hydrogen bond dynamic propensity analysis. J Comput Aided Mol Des 2018; 32:781-791. [DOI: 10.1007/s10822-018-0129-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/02/2018] [Indexed: 11/27/2022]
|
29
|
Gutiérrez LJ, Parravicini O, Sánchez E, Rodríguez R, Cobo J, Enriz RD. New substituted aminopyrimidine derivatives as BACE1 inhibitors: in silico design, synthesis and biological assays. J Biomol Struct Dyn 2018; 37:229-246. [DOI: 10.1080/07391102.2018.1424036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Lucas J. Gutiérrez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| | - Oscar Parravicini
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| | - Emilse Sánchez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| | - Ricaurte Rodríguez
- Departamento de Química, Universidad Nacional de Colombia, Ciudad Universitaria, Carrera 30, No. 45-03, Bogotá, Colombia
| | - Justo Cobo
- Departamento de Química Inorgánica y Orgánica, Universidad de Jaén, Campus Las Lagunillas s/n, 23071 Jaén, Spain
| | - Ricardo D. Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, 5700 San Luis, Argentina
- IMIBIO-CONICET, UNSL, Chacabuco 915, 5700 San Luis, Argentina
| |
Collapse
|
30
|
Ortiz JE, Garro A, Pigni NB, Agüero MB, Roitman G, Slanis A, Enriz RD, Feresin GE, Bastida J, Tapia A. Cholinesterase-inhibitory effect and in silico analysis of alkaloids from bulbs of Hieronymiella species. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 39:66-74. [PMID: 29433685 DOI: 10.1016/j.phymed.2017.12.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 10/26/2017] [Accepted: 12/19/2017] [Indexed: 05/27/2023]
Abstract
BACKGROUND In Argentina, the Amaryllidaceae family (59 species) comprises a wide variety of genera, only a few species have been investigated as a potential source of cholinesterases inhibitors to treat Alzheimer disease (AD). PURPOSE To study the acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitory activities of the basic dichloromethane extracts (E) from Hieronymiella aurea, H. caletensis, H. clidanthoides, H. marginata, and H. speciosa species, as well as the isolated compounds from these plant extracts. STUDY DESIGN AND METHODS AChE and BChE inhibitory activities were evaluated with the Ellman's spectrophotometric method. The alkaloids composition from the E was obtained by gas chromatography-mass spectrometry (GC-MS). The E were successively chromatographed on a silica gel column and permeated on Sephadex LH-20 column to afford the main alkaloids identified by means of spectroscopic data. Additionally, an in silico study was carried out. RESULTS Nine known alkaloids were isolated from the E of five Hieronymiella species. Galanthamine was identified in all the species by GC-MS standing out H. caletensis with a relative abundance of 9.79% of the total ion current. Strong AChE (IC50 = 1.84 - 15.40 µg/ml) and moderate BChE (IC50 = 23.74 - 136.40 µg/ml) inhibitory activities were displayed by the extracts. Among the isolated alkaloids, only sanguinine and chlidanthine (galanthamine-type alkaloids) demonstrated inhibitory activity toward both enzymes. The QTAIM study suggests that sanguinine has the strongest affinity towards AChE, attributed to an additional interaction with Ser200 as well as stronger molecular interactions Glu199 and His440.These results allowed us to differentiate the molecular behavior in the active site among alkaloids possessing different in vitro inhibitory activities. CONCLUSION Hieronymiella species growing in Argentina represent a rich and widespread source of galanthamine and others AChE and BChE inhibitors alkaloids. Additionally, the new trend towards the use of natural extracts as pharmaceuticals rather than pure drugs opens a pathway for the development of a phytomedicine derived from extracts of Hieronymiella spp.
Collapse
Affiliation(s)
- Javier E Ortiz
- Instituto de Biotecnología-Instituto de Ciencias Básicas, CONICET, Facultad de Ingeniería, Universidad Nacional de San Juan, Av. Libertador General San Martín 1109 (O), San Juan CP 5400, Argentina
| | - Adriana Garro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, San Luis 5700, Argentina
| | - Natalia B Pigni
- ICYTAC-CONICET, Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; Departament de Productes Naturals, Biologia Vegetal i Edafologia, Facultat de Farmàcia, Universitat de Barcelona, Avda. Joan XXIII s/n, Barcelona 08028, Spain
| | - María Belén Agüero
- Instituto de Biotecnología-Instituto de Ciencias Básicas, CONICET, Facultad de Ingeniería, Universidad Nacional de San Juan, Av. Libertador General San Martín 1109 (O), San Juan CP 5400, Argentina
| | - German Roitman
- Cátedra de Jardinería, Facultad de Agronomía, Universidad de Buenos Aires, Av. San Martín 4453, Buenos Aires 1417, Argentina
| | - Alberto Slanis
- Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán, Fundación Miguel Lillo 251, Tucumán, Argentina
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 915, San Luis 5700, Argentina
| | - Gabriela E Feresin
- Instituto de Biotecnología-Instituto de Ciencias Básicas, CONICET, Facultad de Ingeniería, Universidad Nacional de San Juan, Av. Libertador General San Martín 1109 (O), San Juan CP 5400, Argentina
| | - Jaume Bastida
- Departament de Productes Naturals, Biologia Vegetal i Edafologia, Facultat de Farmàcia, Universitat de Barcelona, Avda. Joan XXIII s/n, Barcelona 08028, Spain
| | - Alejandro Tapia
- Instituto de Biotecnología-Instituto de Ciencias Básicas, CONICET, Facultad de Ingeniería, Universidad Nacional de San Juan, Av. Libertador General San Martín 1109 (O), San Juan CP 5400, Argentina.
| |
Collapse
|
31
|
Padrtova T, Marvanova P, Odehnalova K, Kubinova R, Parravicini O, Garro A, Enriz RD, Humpa O, Oravec M, Mokry P. Synthesis, Analysis, Cholinesterase-Inhibiting Activity and Molecular Modelling Studies of 3-(Dialkylamino)-2-hydroxypropyl 4-[(Alkoxy-carbonyl)amino]benzoates and Their Quaternary Ammonium Salts. Molecules 2017; 22:molecules22122048. [PMID: 29168793 PMCID: PMC6149889 DOI: 10.3390/molecules22122048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/08/2017] [Accepted: 11/21/2017] [Indexed: 11/16/2022] Open
Abstract
Tertiary amines 3-(dialkylamino)-2-hydroxypropyl 4-[(alkoxycarbonyl)amino]benzoates and their quaternary ammonium salts were synthesized. The final step of synthesis of quaternary ammonium salts was carried out by microwave-assisted synthesis. Software-calculated data provided the background needed to compare fifteen new resulting compounds by their physicochemical properties. The acid dissociation constant (pKa) and lipophilicity index (log P) of tertiary amines were determined; while quaternary ammonium salts were characterized by software-calculated lipophilicity index and surface tension. Biological evaluation aimed at testing acetylcholinesterase and butyrylcholinesterase-inhibiting activity of synthesized compounds. A possible mechanism of action of these compounds was determined by molecular modelling study using combined techniques of docking; molecular dynamics simulations and quantum mechanics calculations.
Collapse
Affiliation(s)
- Tereza Padrtova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho 1, 61242 Brno, Czech Republic.
| | - Pavlina Marvanova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho 1, 61242 Brno, Czech Republic.
| | - Klara Odehnalova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho 1, 61242 Brno, Czech Republic.
| | - Renata Kubinova
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho 1, 61242 Brno, Czech Republic.
| | - Oscar Parravicini
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis-IMIBIO-SL-CONICET, Chacabuco 915, San Luis 5700, Argentina.
| | - Adriana Garro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis-IMIBIO-SL-CONICET, Chacabuco 915, San Luis 5700, Argentina.
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis-IMIBIO-SL-CONICET, Chacabuco 915, San Luis 5700, Argentina.
| | - Otakar Humpa
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic.
| | - Michal Oravec
- Global Change Research Institute CAS, Belidla 986/4a, 60300 Brno, Czech Republic.
| | - Petr Mokry
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho 1, 61242 Brno, Czech Republic.
| |
Collapse
|
32
|
Tetrahydroisoquinolines functionalized with carbamates as selective ligands of D2 dopamine receptor. J Mol Model 2017; 23:273. [DOI: 10.1007/s00894-017-3441-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 08/15/2017] [Indexed: 01/11/2023]
|
33
|
Arodola OA, Soliman MES. Quantum mechanics implementation in drug-design workflows: does it really help? Drug Des Devel Ther 2017; 11:2551-2564. [PMID: 28919707 PMCID: PMC5587087 DOI: 10.2147/dddt.s126344] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The pharmaceutical industry is progressively operating in an era where development costs are constantly under pressure, higher percentages of drugs are demanded, and the drug-discovery process is a trial-and-error run. The profit that flows in with the discovery of new drugs has always been the motivation for the industry to keep up the pace and keep abreast with the endless demand for medicines. The process of finding a molecule that binds to the target protein using in silico tools has made computational chemistry a valuable tool in drug discovery in both academic research and pharmaceutical industry. However, the complexity of many protein-ligand interactions challenges the accuracy and efficiency of the commonly used empirical methods. The usefulness of quantum mechanics (QM) in drug-protein interaction cannot be overemphasized; however, this approach has little significance in some empirical methods. In this review, we discuss recent developments in, and application of, QM to medically relevant biomolecules. We critically discuss the different types of QM-based methods and their proposed application to incorporating them into drug-design and -discovery workflows while trying to answer a critical question: are QM-based methods of real help in drug-design and -discovery research and industry?
Collapse
Affiliation(s)
- Olayide A Arodola
- Department of Pharmaceutical Chemistry, University of KwaZulu-Natal, Durban, South Africa
| | - Mahmoud ES Soliman
- Department of Pharmaceutical Chemistry, University of KwaZulu-Natal, Durban, South Africa
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Egypt
| |
Collapse
|
34
|
Vettorazzi M, Angelina E, Lima S, Gonec T, Otevrel J, Marvanova P, Padrtova T, Mokry P, Bobal P, Acosta LM, Palma A, Cobo J, Bobalova J, Csollei J, Malik I, Alvarez S, Spiegel S, Jampilek J, Enriz RD. An integrative study to identify novel scaffolds for sphingosine kinase 1 inhibitors. Eur J Med Chem 2017; 139:461-481. [PMID: 28822281 DOI: 10.1016/j.ejmech.2017.08.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/02/2017] [Accepted: 08/05/2017] [Indexed: 10/19/2022]
Abstract
Sphingosine kinase 1 (SphK1), the enzyme that produces the bioactive sphingolipid metabolite, sphingosine-1-phosphate, is a promising new molecular target for therapeutic intervention in cancer and inflammatory diseases. In view of its importance, the main objective of this work was to find new and more potent inhibitors for this enzyme possessing different structural scaffolds than those of the known inhibitors. Our theoretical and experimental study has allowed us to identify two new structural scaffolds (three new compounds), which could be used as starting structures for the design and then the development of new inhibitors of SphK1. Our study was carried out in different steps: virtual screening, synthesis, bioassays and molecular modelling. From our results, we propose a new dihydrobenzo[b]pyrimido[5,4-f]azepine and two alkyl{3-/4-[1-hydroxy-2-(4-arylpiperazin-1-yl)ethyl]phenyl}carbamates as initial structures for the development of new inhibitors. In addition, our molecular modelling study using QTAIM calculations, allowed us to describe in detail the molecular interactions that stabilize the different Ligand-Receptor complexes. Such analyses indicate that the cationic head of the different compounds must be refined in order to obtain an increase in the binding affinity of these ligands.
Collapse
Affiliation(s)
- Marcela Vettorazzi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Química, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Avda. Libertad 5460, 3400 Corrientes, Argentina
| | - Santiago Lima
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Jan Otevrel
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Pavlina Marvanova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Tereza Padrtova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Petr Mokry
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Pavel Bobal
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic
| | - Lina M Acosta
- Laboratorio de Síntesis Orgánica, Escuela de Química, Universidad Industrial de Santander, Carrera 27, Calle 9, A.A 678, Bucaramanga, Colombia
| | - Alirio Palma
- Laboratorio de Síntesis Orgánica, Escuela de Química, Universidad Industrial de Santander, Carrera 27, Calle 9, A.A 678, Bucaramanga, Colombia
| | - Justo Cobo
- Inorganic and Organic Department, University of Jaén, Campus Las Lagunillas s/n, 23071, Jaén, Spain
| | - Janette Bobalova
- Institute of Analytical Chemistry of the Czech Academy of Sciences, v. v. i., Veveri 97, 602 00 Brno, Czech Republic
| | - Jozef Csollei
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 612 42 Brno, Czech Republic; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Ivan Malik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Sergio Alvarez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA
| | - Josef Jampilek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Chacabuco 915, 5700 San Luis, Argentina.
| |
Collapse
|
35
|
Chao B, Li BX, Xiao X. Design, synthesis and evaluation of antitumor acylated monoaminopyrroloquinazolines. Bioorg Med Chem Lett 2017; 27:3107-3110. [PMID: 28552339 DOI: 10.1016/j.bmcl.2017.05.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/09/2017] [Accepted: 05/13/2017] [Indexed: 12/13/2022]
Abstract
Pyrroloquinazoline is a privileged chemical scaffold with diverse biological activities. We recently described a series of N-3 acylated 1,3-diaminopyrroloquinazolines with potent anticancer activities. The N-1 primary amino group in 1,3-diaminopyrroloquinazoline is critical for its inhibitory activity against dihydrofolate reductase (DHFR). In order to design out this unnecessary DHFR inhibition activity and further expand the chemical space associated with pyrroloquinazoline, we removed the N-1 primary amino group. In this report, we describe our design and synthesis of a series of N-3 acylated monoaminopyrroloquinazolines. Biological evaluation of these compounds identified a naphthamide 4a as a potent anticancer agent (GI50=88-200nM), suggesting that removing the N-1 primary amino group in 1,3-diaminopyrroloquinazoline is a useful chemical modification that can be introduced to improve the anticancer activity.
Collapse
Affiliation(s)
- Bo Chao
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Bingbing X Li
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Xiangshu Xiao
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| |
Collapse
|
36
|
The electronic density obtained from a QTAIM analysis used as molecular descriptor. A study performed in a new series of DHFR inhibitors. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2016.12.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
37
|
Dihydrofolate reductase inhibitors: a quantitative structure–activity relationship study using 2D-QSAR and 3D-QSAR methods. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1742-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
38
|
Luchi AM, Angelina EL, Andujar SA, Enriz RD, Peruchena NM. Halogen bonding in biological context: a computational study of D2 dopamine receptor. J PHYS ORG CHEM 2016; 29:645-655. [DOI: 10.1002/poc.3586] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
In this work, Halogen Bond (X‐bond) interactions formed by halogenated ligands (LX) at the Dopamine Receptor D2 (DRD2) binding pocket were studied by Molecular Dynamics (MD) and charge density analysis. The X‐bonds were contrasted with the Hydrogen Bond (H‐bond) interactions established by hydroxylated analogs (LOH, where X was replaced by OH). The ligands for this study were extracted from a dataset of compounds deposited in ZINC database that were active in binding assays to DRD2. This dataset was subjected to the filtering rules by employing cheminformatics tools to find the LX/LOH pairs that were then submitted to MD simulations. A homology model of DRD2 was employed for the simulations because no crystal structure is yet available for the receptor. To mimic the positive cap (σ‐hole) on the halogen atom, a massless, positive charged extra‐point was introduced in the force field. An analysis of the charge density (QTAIM) was performed on reduced models of simulated complexes to explain their binding differences. Results show that the halogen atom tends to form X‐bond with protein backbone oxygen atom. Two out of the four halogenated ligands studied form a specific X‐bond with the carbonyl oxygen of Ser193. This specific X‐bond decreases the inherent propensity of transmembrane 5 to unfolding. These results suggest a possible role of the X‐bond as a protein secondary structure modulator because of the ability of the halogen to interact with the protein backbone. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Adriano M. Luchi
- Lab. Estructura Molecular y Propiedades, IQUIBA‐NEA Universidad Nacional del Nordeste, CONICET, FACENA Av. Libertad 5470 Corrientes 3400 Argentina
| | - Emilio L. Angelina
- Lab. Estructura Molecular y Propiedades, IQUIBA‐NEA Universidad Nacional del Nordeste, CONICET, FACENA Av. Libertad 5470 Corrientes 3400 Argentina
| | - Sebastián A. Andujar
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis Universidad Nacional de San Luis, CONICET, FQBF Chacabuco 917 San Luis 5700 Argentina
| | - Ricardo D. Enriz
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis Universidad Nacional de San Luis, CONICET, FQBF Chacabuco 917 San Luis 5700 Argentina
| | - Nélida M. Peruchena
- Lab. Estructura Molecular y Propiedades, IQUIBA‐NEA Universidad Nacional del Nordeste, CONICET, FACENA Av. Libertad 5470 Corrientes 3400 Argentina
| |
Collapse
|
39
|
The Eighth Central European Conference "Chemistry towards Biology": Snapshot. Molecules 2016; 21:molecules21101381. [PMID: 27763518 PMCID: PMC5283649 DOI: 10.3390/molecules21101381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/12/2016] [Indexed: 01/27/2023] Open
Abstract
The Eighth Central European Conference "Chemistry towards Biology" was held in Brno, Czech Republic, on August 28-September 1, 2016 to bring together experts in biology, chemistry and design of bioactive compounds; promote the exchange of scientific results, methods and ideas; and encourage cooperation between researchers from all over the world. The topics of the conference covered "Chemistry towards Biology", meaning that the event welcomed chemists working on biology-related problems, biologists using chemical methods, and students and other researchers of the respective areas that fall within the common scope of chemistry and biology. The authors of this manuscript are plenary speakers and other participants of the symposium and members of their research teams. The following summary highlights the major points/topics of the meeting.
Collapse
|
40
|
Párraga J, Andujar SA, Rojas S, Gutierrez LJ, El Aouad N, Sanz MJ, Enriz RD, Cabedo N, Cortes D. Dopaminergic isoquinolines with hexahydrocyclopenta[ij]-isoquinolines as D2-like selective ligands. Eur J Med Chem 2016; 122:27-42. [PMID: 27343851 DOI: 10.1016/j.ejmech.2016.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/11/2023]
Abstract
Dopamine receptors (DR) ligands are potential drug candidates for treating neurological disorders including schizophrenia or Parkinson's disease. Three series of isoquinolines: (E)-1-styryl-1,2,3,4-tetrahydroisoquinolines (series 1), 7-phenyl-1,2,3,7,8,8a-hexahydrocyclopenta[ij]-IQs (HCPIQs) (series 2) and (E)-1-(prop-1-en-1-yl)-1,2,3,4- tetrahydroisoquinolines (series 3), were prepared to determine their affinity for both D1 and D2-like DR. The effect of different substituents on the nitrogen atom (methyl or allyl), the dioxygenated function (methoxyl or catechol), the substituent at the β-position of the THIQ skeleton, and the presence or absence of the cyclopentane motif, were studied. We observed that the most active compounds in the three series (2c, 2e, 3a, 3c, 3e, 5c and 5e) possessed a high affinity for D2-like DR and these remarkable features: a catechol group in the IQ-ring and the N-substitution (methyl or allyl). The series showed the following trend to D2-RD affinity: HCPIQs > 1-styryl > 1-propenyl. Therefore, the substituent at the β-position of the THIQ and the cyclopentane ring also modulated this affinity. Among these dopaminergic isoquinolines, HCPIQs stood out for unexpected selectivity to D2-DR since the Ki D1/D2 ratio reached values of 2465, 1010 and 382 for compounds 3a, 3c and 3e, respectively. None of the most active THIQs in D2 DR displayed relevant cytotoxicity in human neutrophils and HUVEC. Finally, and in agreement with the experimental data, molecular modeling studies on DRs of the most characteristic ligands of the three series revealed stronger molecular interactions with D2 DR than with D1 DR, which further supports to the encountered enhanced selectivity to D2 DR.
Collapse
Affiliation(s)
- Javier Párraga
- Departamento de Farmacología, Laboratorio de Farmacoquímica, Facultad de Farmacia, Universidad de Valencia, 46100, Burjassot, Valencia, Spain
| | - Sebastián A Andujar
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis-IMIBIO-SL, Chacabuco 915, 5700, San Luis, Argentina
| | - Sebastián Rojas
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis-IMIBIO-SL, Chacabuco 915, 5700, San Luis, Argentina
| | - Lucas J Gutierrez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis-IMIBIO-SL, Chacabuco 915, 5700, San Luis, Argentina
| | | | - M Jesús Sanz
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, 46010, Valencia, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010, Valencia, Spain
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis-IMIBIO-SL, Chacabuco 915, 5700, San Luis, Argentina
| | - Nuria Cabedo
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, 46010, Valencia, Spain.
| | - Diego Cortes
- Departamento de Farmacología, Laboratorio de Farmacoquímica, Facultad de Farmacia, Universidad de Valencia, 46100, Burjassot, Valencia, Spain.
| |
Collapse
|
41
|
Ortiz JE, Pigni NB, Andujar SA, Roitman G, Suvire FD, Enriz RD, Tapia A, Bastida J, Feresin GE. Alkaloids from Hippeastrum argentinum and Their Cholinesterase-Inhibitory Activities: An in Vitro and in Silico Study. JOURNAL OF NATURAL PRODUCTS 2016; 79:1241-8. [PMID: 27096334 DOI: 10.1021/acs.jnatprod.5b00785] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Two new alkaloids, 4-O-methylnangustine (1) and 7-hydroxyclivonine (2) (montanine and homolycorine types, respectively), and four known alkaloids were isolated from the bulbs of Hippeastrum argentinum, and their cholinesterase-inhibitory activities were evaluated. These compounds were identified using GC-MS, and their structures were defined by physical data analysis. Compound 2 showed weak butyrylcholinesterase (BuChE)-inhibitory activity, with a half-maximal inhibitory concentration (IC50) value of 67.3 ± 0.09 μM. To better understand the experimental results, a molecular modeling study was also performed. The combination of a docking study, molecular dynamics simulations, and quantum theory of atoms in molecules calculations provides new insight into the molecular interactions of compound 2 with BuChE, which were compared to those of galantamine.
Collapse
Affiliation(s)
- Javier E Ortiz
- Instituto de Biotecnología, Facultad de Ingeniería, Universidad Nacional de San Juan , Avenida Libertador General San Martín 1109 (O), 5400 San Juan, Argentina
| | - Natalia B Pigni
- Departament de Productes Naturals, Biologia Vegetal i Edafologia, Facultat de Farmàcia, Universitat de Barcelona , Avenida Joan XXIII s/n, 08028 Barcelona, Spain
- ICYTAC-CONICET, Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , 5000 Córdoba, Argentina
| | - Sebastián A Andujar
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis , Chacabuco 915, 5700 San Luis, Argentina
| | - German Roitman
- Cátedra de Jardinería, Facultad de Agronomía, Universidad de Buenos Aires , Avenida San Martín 4453, 1417 Buenos Aires, Argentina
| | - Fernando D Suvire
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis , Chacabuco 915, 5700 San Luis, Argentina
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis , Chacabuco 915, 5700 San Luis, Argentina
| | - Alejandro Tapia
- Instituto de Biotecnología, Facultad de Ingeniería, Universidad Nacional de San Juan , Avenida Libertador General San Martín 1109 (O), 5400 San Juan, Argentina
| | - Jaume Bastida
- Departament de Productes Naturals, Biologia Vegetal i Edafologia, Facultat de Farmàcia, Universitat de Barcelona , Avenida Joan XXIII s/n, 08028 Barcelona, Spain
| | - Gabriela E Feresin
- Instituto de Biotecnología, Facultad de Ingeniería, Universidad Nacional de San Juan , Avenida Libertador General San Martín 1109 (O), 5400 San Juan, Argentina
| |
Collapse
|
42
|
Gutierrez LJ, Angelina E, Gyebrovszki A, Fülöp L, Peruchena N, Baldoni HA, Penke B, Enriz RD. New small-size peptides modulators of the exosite of BACE1 obtained from a structure-based design. J Biomol Struct Dyn 2016; 35:413-426. [DOI: 10.1080/07391102.2016.1145143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Lucas J. Gutierrez
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Químicam, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Av. Libertad 5470, Corrientes 3400, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Químicam, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Av. Libertad 5470, Corrientes 3400, Argentina
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL, CONICET), Ejercito de Los Andes 950, 5700 San Luis, Argentina
| | - Andrea Gyebrovszki
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Dóm tér 8., Hungary
| | - Lívia Fülöp
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Dóm tér 8., Hungary
| | - Nelida Peruchena
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Químicam, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Av. Libertad 5470, Corrientes 3400, Argentina
| | - Héctor A. Baldoni
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 917, 5700 San Luis, Argentina
- Instituto de Matemática Aplicada San Luis (IMASL,CONICET), Italia 1556, 5700 San Luis, Argentina
| | - Botond Penke
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Dóm tér 8., Hungary
| | - Ricardo D. Enriz
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL, CONICET), Ejercito de Los Andes 950, 5700 San Luis, Argentina
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco 917, 5700 San Luis, Argentina
| |
Collapse
|
43
|
Conformational transition of Aβ 42 inhibited by a mimetic peptide. A molecular modeling study using QM/MM calculations and QTAIM analysis. COMPUT THEOR CHEM 2016. [DOI: 10.1016/j.comptc.2016.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Two-generation reproduction and teratology studies of feeding aditoprim in Wistar rats. J Appl Toxicol 2015; 35:1531-8. [DOI: 10.1002/jat.3121] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/12/2014] [Accepted: 12/19/2014] [Indexed: 02/02/2023]
|
45
|
Wang X, Su S, Awais I, Liu Q, Chen D, Liu Z, Wang Y, Yuan Z. Two generation reproduction and teratogenicity studies of feeding diaveridine in Wistar rats. Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00001g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Diaveridine (DVD), belonging to the class of compounds called diaminopyrimidines, has been widely used as a bacteriostatic agent.
Collapse
Affiliation(s)
- Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues
- China
| | - Shijia Su
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products
- Huazhong Agricultural University
- Wuhan
- China
| | - Ihsan Awais
- Department of Biosciences
- COMSATS Institute of Information Technology
- Sahiwal
- Pakistan
| | - Qianying Liu
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products
- Huazhong Agricultural University
- Wuhan
- China
| | - Dongmei Chen
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products
- Huazhong Agricultural University
- Wuhan
- China
| | - Zhenli Liu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety
- Wuhan
- China
| | - Yulian Wang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products
- Huazhong Agricultural University
- Wuhan
- China
| | - Zonghui Yuan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues
- China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products
- Huazhong Agricultural University
- Wuhan
| |
Collapse
|
46
|
Angelina E, Andujar S, Moreno L, Garibotto F, Párraga J, Peruchena N, Cabedo N, Villecco M, Cortes D, Enriz RD. 3-Chlorotyramine Acting as Ligand of the D2
Dopamine Receptor. Molecular Modeling, Synthesis and D2
Receptor Affinity. Mol Inform 2014; 34:28-43. [PMID: 27490860 DOI: 10.1002/minf.201400093] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/04/2014] [Indexed: 12/12/2022]
|
47
|
Mokmak W, Chunsrivirot S, Hannongbua S, Yuthavong Y, Tongsima S, Kamchonwongpaisan S. Molecular dynamics of interactions between rigid and flexible antifolates and dihydrofolate reductase from pyrimethamine-sensitive and pyrimethamine-resistant Plasmodium falciparum. Chem Biol Drug Des 2014; 84:450-61. [PMID: 24716467 DOI: 10.1111/cbdd.12334] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/04/2014] [Accepted: 04/03/2014] [Indexed: 01/16/2023]
Abstract
Currently, the usefulness of antimalarials such as pyrimethamine (PYR) is drastically reduced due to the emergence of resistant Plasmodium falciparum (Pf) caused by its dihydrofolate reductase (PfDHFR) mutations, especially the quadruple N51I/C59R/S108N/I164L mutations. The resistance was due to the steric conflict of PYR with S108N. WR99210 (WR), a dihydrotriazine antifolate with a flexible side chain that can avoid such conflict, can overcome this resistance through tight binding with the mutant. To understand factors contributing to different binding affinities of PYR/WR to the wild type (WT) and quadruple mutant (QM), we performed simulations on WR-WT, WR-QM, PYR-WT, and PYR-QM complexes and found that Ile14 and Asp54 were crucial for PYR/WR binding to PfDHFR due to strong hydrogen bonds. The quadruple mutations cause PYR to form, on average, fewer hydrogen bonds with Ile14 and Leu164, and to be displaced from its optimal orientation for Asp54 interaction. The predicted binding affinity ranking (WR-QM ≈ WR-WT ≈ PYR-WT >> PYR-QM) reasonably agrees with the inhibition constant (K(i)) ranking. Our results reveal important residues for tight binding of PYR/WR to WT/QM, which may be used to evaluate the inhibition effectiveness of antimalarials and to provide fundamental information for designing new drugs effective against drug-resistant P. falciparum.
Collapse
Affiliation(s)
- Wanwimon Mokmak
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Klong Luang, Pathum Thani, 12120, Thailand
| | | | | | | | | | | |
Collapse
|
48
|
Angelina EL, Andujar SA, Tosso RD, Enriz RD, Peruchena NM. Non-covalent interactions in receptor-ligand complexes. A study based on the electron charge density. J PHYS ORG CHEM 2013. [DOI: 10.1002/poc.3250] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Emilio L. Angelina
- Lab. Estructura Molecular y Propiedades, Área de Química Física, Departamento de Química, Facultad de Ciencias Exactas y Naturales y Agrimensura; Universidad Nacional del Nordeste; Av. Libertad 5470 Corrientes 3400 Argentina
- Instituto Multidiciplinario de Investigaciones Biológicas (IMIBIO-SL, CONICET); Chacabuco y Pedrenera (5700) San Luis Argentina
| | - Sebastián A. Andujar
- Departamento de Química, Facultad de Química, Bioquímica y Farmacia; Universidad Nacional de San Luis; Chacabuco 917, San Luis (5700) San Luis Argentina
- Instituto Multidiciplinario de Investigaciones Biológicas (IMIBIO-SL, CONICET); Chacabuco y Pedrenera (5700) San Luis Argentina
| | - Rodrigo D. Tosso
- Departamento de Química, Facultad de Química, Bioquímica y Farmacia; Universidad Nacional de San Luis; Chacabuco 917, San Luis (5700) San Luis Argentina
- Instituto Multidiciplinario de Investigaciones Biológicas (IMIBIO-SL, CONICET); Chacabuco y Pedrenera (5700) San Luis Argentina
| | - Ricardo D. Enriz
- Departamento de Química, Facultad de Química, Bioquímica y Farmacia; Universidad Nacional de San Luis; Chacabuco 917, San Luis (5700) San Luis Argentina
- Instituto Multidiciplinario de Investigaciones Biológicas (IMIBIO-SL, CONICET); Chacabuco y Pedrenera (5700) San Luis Argentina
| | - Nélida M. Peruchena
- Lab. Estructura Molecular y Propiedades, Área de Química Física, Departamento de Química, Facultad de Ciencias Exactas y Naturales y Agrimensura; Universidad Nacional del Nordeste; Av. Libertad 5470 Corrientes 3400 Argentina
| |
Collapse
|
49
|
Insight into the molecular mechanism about lowered dihydrofolate binding affinity to dihydrofolate reductase-like 1 (DHFRL1). J Mol Model 2013; 19:5187-98. [PMID: 24122410 DOI: 10.1007/s00894-013-2018-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/15/2013] [Indexed: 10/26/2022]
Abstract
Human dihydrofolate reductase-like 1 (DHFRL1) has been identified as a second human dihydrofolate reductase (DHFR) enzyme. Although DHFRL1 have high sequence homology with human DHFR, dihydrofolate (DHF) exhibits a lowered binding affinity to DHFRL1 and the corresponding molecular mechanism is still unknown. To address this question, we studied the binding of DHF to DHFRL1 and DHFR by using molecular dynamics simulation. Moreover, to investigate the role the 24th residue of DHFR/DHFRL1 plays in DHF binding, R24W DHFRL1 mutant was also studied. The van der Waals interaction are more crucial for the total DHF binding energies, while the difference between the DHF binding energies of human DHFR and DHFRL1 can be attributed to the electrostatic interaction and the polar desolvation free energy.More specifically, lower DHF affinity to DHFRL1 can be mainly attributed to the reduction of net electrostatic interactions of residues Arg32 and Gln35 of DHFRL1 with DHF as being affected by Arg24. The side chain of Arg24 in DHFRL1 can extend deeply into the binding sites of DHF and NADPH, and disturb the DHF binding by steric effect, which rarely happens in human DHFR and R24W DHFRL1 mutant. Additionally, the conformation of loop I in DHFRL1 was also studied in this work. Interestingly, the loop conformation resemble to normal closed state of Escherichia coli DHFR other than the closed state of human DHFR. We hope this work will be useful to understand the general characteristics of DHFRL1.
Collapse
|