201
|
de Mattos Oliveira L, Araújo JSC, Bacelar Costa Junior D, Santana IB, Duarte AA, Leite FHA, Benevides RG, Coelho Dos Santos Junior M. Pharmacophore modeling, docking and molecular dynamics to identify Leishmania major farnesyl pyrophosphate synthase inhibitors. J Mol Model 2018; 24:314. [PMID: 30327889 DOI: 10.1007/s00894-018-3838-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 09/18/2018] [Indexed: 12/01/2022]
Abstract
Leishmaniasis is caused by protozoa of the genus Leishmania spp. and is considered the second most important protozoa in the world due to the number of cases and mortality. Despite its importance in terms of public health, the treatment of patients is limited and has mostly low levels of efficacy and safety. Farnesyl pyrophosphate synthase (FPPS) acts in the early stages of isoprenoid synthesis, and is important for maintaining the integrity of the lipid bilayer of the parasite that causes the disease. The aim of this work was to identify one potential inhibitor of the FPPS of Leishmania major through virtual screening by pharmacophore modeling and docking. A total of 85,000 compounds from a natural products database (ZINC15) was submitted for virtual hierarchical screening, and the top ranked molecule in both methods was analyzed by intermolecular interaction profile and 20 ns molecular dynamics simulations. These results showed a promising compound from natural products that mimic the major interactions present in the substrate/inhibitor.
Collapse
Affiliation(s)
- Larissa de Mattos Oliveira
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil.,Laboratório de Modelagem Molecular, Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - Janay Stefany Carneiro Araújo
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil.,Laboratório de Modelagem Molecular, Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - David Bacelar Costa Junior
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil.,Laboratório de Modelagem Molecular, Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - Isis Bugia Santana
- Laboratório de Modelagem Molecular, Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil.,Programa de Pós-graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - Angelo Amâncio Duarte
- Laboratório de Computação de Alto Desempenho, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - Franco Henrique Andrade Leite
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil.,Laboratório de Modelagem Molecular, Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil.,Programa de Pós-graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - Raquel Guimarães Benevides
- Programa de Pós-graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - Manoelito Coelho Dos Santos Junior
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil. .,Laboratório de Modelagem Molecular, Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil. .,Programa de Pós-graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, Brazil.
| |
Collapse
|
202
|
Synthesis of Bis-indolylmethane sulfonohydrazides derivatives as potent α-Glucosidase inhibitors. Bioorg Chem 2018; 80:112-120. [DOI: 10.1016/j.bioorg.2018.06.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 12/30/2022]
|
203
|
Gimeno A, Ardid-Ruiz A, Ojeda-Montes MJ, Tomás-Hernández S, Cereto-Massagué A, Beltrán-Debón R, Mulero M, Valls C, Aragonès G, Suárez M, Pujadas G, Garcia-Vallvé S. Combined Ligand- and Receptor-Based Virtual Screening Methodology to Identify Structurally Diverse Protein Tyrosine Phosphatase 1B Inhibitors. ChemMedChem 2018; 13:1939-1948. [DOI: 10.1002/cmdc.201800267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/05/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Aleix Gimeno
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Andrea Ardid-Ruiz
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - María José Ojeda-Montes
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Sarah Tomás-Hernández
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Adrià Cereto-Massagué
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Raúl Beltrán-Debón
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Miquel Mulero
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Cristina Valls
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Gerard Aragonès
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Manuel Suárez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
| | - Gerard Pujadas
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
- EURECAT, TECNIO, CEICS; Avinguda Universitat 1 43204 Reus Catalonia Spain
| | - Santiago Garcia-Vallvé
- Research Group in Cheminformatics & Nutrition, Departament de Bioquímica i Biotecnologia; Universitat Rovira i Virgili; Campus de Sescelades 43007 Tarragona Catalonia Spain
- EURECAT, TECNIO, CEICS; Avinguda Universitat 1 43204 Reus Catalonia Spain
| |
Collapse
|
204
|
Szulczyk D, Dobrowolski MA, Roszkowski P, Bielenica A, Stefańska J, Koliński M, Kmiecik S, Jóźwiak M, Wrzosek M, Olejarz W, Struga M. Design and synthesis of novel 1H-tetrazol-5-amine based potent antimicrobial agents: DNA topoisomerase IV and gyrase affinity evaluation supported by molecular docking studies. Eur J Med Chem 2018; 156:631-640. [PMID: 30031974 DOI: 10.1016/j.ejmech.2018.07.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 07/11/2018] [Accepted: 07/15/2018] [Indexed: 12/27/2022]
Abstract
A total of 14 of 1,5-disubstituted tetrazole derivatives were prepared by reacting appropriate thiourea and sodium azide in the presence of mercury (II) chloride and triethylamine. All compounds were evaluated in vitro for their antimicrobial activity. Derivatives 10 and 11 showed the highest inhibition against Gram-positive and Gram-negative strains (standard and hospital strains). The observed minimal inhibitory concentrations values were in the range of 1-208 μM (0.25-64 μg/ml). Inhibitory activity of 1,5-tetrazole derivatives 10 and 11 against gyrase and topoisomerase IV isolated from S. aureus was studied. Evaluation was supported by molecular docking studies for all synthesized derivatives and reference ciprofloxacin. Moreover, selected tetrazoles (2, 3, 5, 6, 8, 9, 10 and 11) were evaluated for their cytotoxicity. All tested compounds are non-cytotoxic against HaCaT and A549 cells (CC50 ≤ 60 μM).
Collapse
Affiliation(s)
- Daniel Szulczyk
- Chair and Department of Biochemistry, Medical University, 02-097 Warszawa, Poland.
| | | | - Piotr Roszkowski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Anna Bielenica
- Chair and Department of Biochemistry, Medical University, 02-097 Warszawa, Poland
| | - Joanna Stefańska
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; Department of Pharmaceutical Microbiology, Medical University, 02-007 Warszawa, Poland
| | - Michał Koliński
- Bioinformatics Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Sebastian Kmiecik
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, 02-089 Warsaw, Poland
| | - Michał Jóźwiak
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warszawa, Poland; Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland; Department of Biochemistry, Second Faculty of Medicine, Medical University of Warsaw, 02-097 Warszawa, Poland
| | - Małgorzata Wrzosek
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warszawa, Poland; Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warszawa, Poland; Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Marta Struga
- Chair and Department of Biochemistry, Medical University, 02-097 Warszawa, Poland; Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| |
Collapse
|
205
|
Vadhadiya PM, Jean MA, Bouzriba C, Tremblay T, Lagüe P, Fortin S, Boukouvalas J, Giguère D. Diversity-Oriented Synthesis of Diol-Based Peptidomimetics as Potential HIV Protease Inhibitors and Antitumor Agents. Chembiochem 2018; 19:1779-1791. [PMID: 29858881 DOI: 10.1002/cbic.201800247] [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: 05/08/2018] [Indexed: 12/15/2022]
Abstract
Peptidomimetic HIV protease inhibitors are an important class of drugs used in the treatment of AIDS. The synthesis of a new type of diol-based peptidomimetics is described. Our route is flexible, uses d-glucal as an inexpensive starting material, and makes minimal use of protection/deprotection cycles. Binding affinities from molecular docking simulations suggest that these compounds are potential inhibitors of HIV protease. Moreover, the antiproliferative activities of compounds 33 a, 35 a, and 35 b on HT-29, M21, and MCF7 cancer cell lines are in the low micromolar range. The results provide a platform that could facilitate the development of medically relevant asymmetrical diol-based peptidomimetics.
Collapse
Affiliation(s)
- Paresh M Vadhadiya
- Département de Chimie, Université Laval-RQRM, 1045 Avenue de la Médecine, Quebec City, QC, G1V 0A6, Canada
| | - Marc-Alexandre Jean
- Département de Chimie, Université Laval-RQRM, 1045 Avenue de la Médecine, Quebec City, QC, G1V 0A6, Canada
| | - Chahrazed Bouzriba
- CHU de Québec-Université Laval Research Center, Oncology Division, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Quebec City, QC, G1L 3L5, Canada
- Faculté de Pharmacie, Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Thomas Tremblay
- Département de Chimie, Université Laval-RQRM, 1045 Avenue de la Médecine, Quebec City, QC, G1V 0A6, Canada
| | - Patrick Lagüe
- Départment de Biochimie, de Microbiologie et de Bio-Informatique, Université Laval, 1045, Avenue de la Médecine, Quebec City, QC, G1V 0A6, Canada
| | - Sébastien Fortin
- CHU de Québec-Université Laval Research Center, Oncology Division, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Quebec City, QC, G1L 3L5, Canada
- Faculté de Pharmacie, Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - John Boukouvalas
- Département de Chimie, Université Laval-RQRM, 1045 Avenue de la Médecine, Quebec City, QC, G1V 0A6, Canada
| | - Denis Giguère
- Département de Chimie, Université Laval-RQRM, 1045 Avenue de la Médecine, Quebec City, QC, G1V 0A6, Canada
| |
Collapse
|
206
|
Mishra V, Pathak C. Structural insights into pharmacophore-assisted in silico identification of protein-protein interaction inhibitors for inhibition of human toll-like receptor 4 - myeloid differentiation factor-2 (hTLR4-MD-2) complex. J Biomol Struct Dyn 2018; 37:1968-1991. [PMID: 29842849 DOI: 10.1080/07391102.2018.1474804] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Toll-like receptor 4 (TLR4) is a member of Toll-Like Receptors (TLRs) family that serves as a receptor for bacterial lipopolysaccharide (LPS). TLR4 alone cannot recognize LPS without aid of co-receptor myeloid differentiation factor-2 (MD-2). Binding of LPS with TLR4 forms a LPS-TLR4-MD-2 complex and directs downstream signaling for activation of immune response, inflammation and NF-κB activation. Activation of TLR4 signaling is associated with various pathophysiological consequences. Therefore, targeting protein-protein interaction (PPI) in TLR4-MD-2 complex formation could be an attractive therapeutic approach for targeting inflammatory disorders. The aim of present study was directed to identify small molecule PPI inhibitors (SMPPIIs) using pharmacophore mapping-based approach of computational drug discovery. Here, we had retrieved the information about the hot spot residues and their pharmacophoric features at both primary (TLR4-MD-2) and dimerization (MD-2-TLR4*) protein-protein interaction interfaces in TLR4-MD-2 homo-dimer complex using in silico methods. Promising candidates were identified after virtual screening, which may restrict TLR4-MD-2 protein-protein interaction. In silico off-target profiling over the virtually screened compounds revealed other possible molecular targets. Two of the virtually screened compounds (C11 and C15) were predicted to have an inhibitory concentration in μM range after HYDE assessment. Molecular dynamics simulation study performed for these two compounds in complex with target protein confirms the stability of the complex. After virtual high throughput screening we found selective hTLR4-MD-2 inhibitors, which may have therapeutic potential to target chronic inflammatory diseases.
Collapse
Affiliation(s)
- Vinita Mishra
- a Department of Cell Biology, School of Biological Sciences & Biotechnology , Indian Institute of Advanced Research, Koba Institutional Area , Gandhinagar , India
| | - Chandramani Pathak
- a Department of Cell Biology, School of Biological Sciences & Biotechnology , Indian Institute of Advanced Research, Koba Institutional Area , Gandhinagar , India
| |
Collapse
|
207
|
de Sarom A, Kumar Jaiswal A, Tiwari S, de Castro Oliveira L, Barh D, Azevedo V, Jose Oliveira C, de Castro Soares S. Putative vaccine candidates and drug targets identified by reverse vaccinology and subtractive genomics approaches to control Haemophilus ducreyi, the causative agent of chancroid. J R Soc Interface 2018; 15:20180032. [PMID: 29792307 PMCID: PMC6000166 DOI: 10.1098/rsif.2018.0032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Chancroid is a sexually transmitted infection (STI) caused by the Gram-negative bacterium Haemophilus ducreyi The control of chancroid is difficult and the only current available treatment is antibiotic therapy; however, antibiotic resistance has been reported in endemic areas. Owing to recent outbreaks of STIs worldwide, it is important to keep searching for new treatment strategies and preventive measures. Here, we applied reverse vaccinology and subtractive genomic approaches for the in silico prediction of potential vaccine and drug targets against 28 strains of H. ducreyi We identified 847 non-host homologous proteins, being 332 exposed/secreted/membrane and 515 cytoplasmic proteins. We also checked their essentiality, functionality and virulence. Altogether, we predicted 13 candidate vaccine targets and three drug targets, where two vaccines (A01_1275, ABC transporter substrate-binding protein; and A01_0690, Probable transmembrane protein) and three drug targets (A01_0698, Purine nucleoside phosphorylase; A01_0702, Transcription termination factor; and A01_0677, Fructose-bisphosphate aldolase class II) are harboured by pathogenicity islands. Finally, we applied a molecular docking approach to analyse each drug target and selected ZINC77257029, ZINC43552589 and ZINC67912117 as promising molecules with favourable interactions with the target active site residues. Altogether, the targets identified here may be used in future strategies to control chancroid worldwide.
Collapse
Affiliation(s)
- Alissa de Sarom
- Institute of Biological Sciences and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Arun Kumar Jaiswal
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sandeep Tiwari
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Letícia de Castro Oliveira
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal, India
| | - Vasco Azevedo
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlo Jose Oliveira
- Institute of Biological Sciences and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Siomar de Castro Soares
- Institute of Biological Sciences and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
208
|
Sukumaran SD, Faraj FL, Lee VS, Othman R, Buckle MJC. 2-Aryl-3-(arylideneamino)-1,2-dihydroquinazoline-4(3 H)-ones as inhibitors of cholinesterases and self-induced β-amyloid (Aβ) aggregation: biological evaluations and mechanistic insights from molecular dynamics simulations. RSC Adv 2018; 8:7818-7831. [PMID: 35539141 PMCID: PMC9078462 DOI: 10.1039/c7ra11872d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/19/2018] [Indexed: 11/21/2022] Open
Abstract
A series of 2-aryl-3-(arylideneamino)-1,2-dihydroquinazoline-4(3H)-ones were evaluated as inhibitors of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and self-induced β-amyloid (Aβ) aggregation. All the compounds were found to inhibit both forms of cholinesterase (IC50 in the range 4-32 μM) with some selectivity for BuChE. Most of the compounds also showed self-induced Aβ aggregation inhibitory activities, which were comparable or higher than those obtained for reference compounds, curcumin and myricetin. Docking and molecular dynamics (MD) simulation experiments suggested that the compounds are able to disrupt the dimer form of Aβ.
Collapse
Affiliation(s)
- Sri Devi Sukumaran
- Department of Pharmacy, Faculty of Medicine, University of Malaya 50603 Kuala Lumpur Malaysia +60-3-7967-4959
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
| | - Fadhil Lafta Faraj
- Department of Chemistry, Faculty of Science, University of Diyala Diyala Governorate Iraq
| | - Vannajan Sanghiran Lee
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
- Department of Chemistry, Faculty of Science, University of Malaya 50603 Kuala Lumpur Malaysia +60 163208906
| | - Rozana Othman
- Department of Pharmacy, Faculty of Medicine, University of Malaya 50603 Kuala Lumpur Malaysia +60-3-7967-4959
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
| | - Michael J C Buckle
- Department of Pharmacy, Faculty of Medicine, University of Malaya 50603 Kuala Lumpur Malaysia +60-3-7967-4959
- Drug Design and Development Research Group (DDDRG), University of Malaya 50603 Kuala Lumpur Malaysia
| |
Collapse
|
209
|
Barrows RD, Blacklock KM, Rablen PR, Khare SD, Knapp S. Computational assessment of thioether isosteres. J Mol Graph Model 2018; 80:282-292. [PMID: 29414047 DOI: 10.1016/j.jmgm.2018.01.018] [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: 11/20/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 11/16/2022]
Abstract
Replacement of the sulfur atom in biologically active diaryl and heteroaryl thioethers (Ar-S-Ar', HAr-S-Ar, and HAr-S-HAr') with any of several one-atom or two-atom linkers can be expected to reduce the susceptibility of the analogue to metabolic oxidation, a well-documented problem for thioethers intended for medicinal chemistry applications. Ab initio calculations indicate how well various proposed thioether isosteric groups, including some new and unusual ones, may perform structurally and electronically in replacing the bridging sulfur atom. Four of these are calculationally evaluated as proposed substructures in Axitinib analogues. The predicted binding behavior of the latter within two different previously crystallographically characterized protein-Axitinib binding sites (VEGFR2 kinase and ABL1 T315I gatekeeper mutant kinase), and an assessment of their suitability and anticipated shortcomings, are presented.
Collapse
Affiliation(s)
- Robert D Barrows
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA
| | - Kristin M Blacklock
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA
| | - Paul R Rablen
- Department of Chemistry & Biochemistry, Swarthmore College, 500 College Ave., Swarthmore, PA 19081 USA
| | - Sagar D Khare
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA
| | - Spencer Knapp
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA.
| |
Collapse
|
210
|
Synthesis, structural and antimicrobial studies of type II topoisomerase-targeted copper(II) complexes of 1,3-disubstituted thiourea ligands. J Inorg Biochem 2018; 182:61-70. [PMID: 29499458 DOI: 10.1016/j.jinorgbio.2018.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/20/2017] [Accepted: 01/07/2018] [Indexed: 12/12/2022]
Abstract
A series of Cu(II) complexes of 3-(trifluoromethyl)phenylthiourea derivatives was synthesized. Their structural properties were investigated by spectroscopic techniques (infrared and electron paramagnetic resonance), as well as molecular modeling. All studied coordination compounds are mononuclear complexes containing two chelating ligands bonded to the metal cation via S and deprotonated N atoms. The new chelates were evaluated for their antimicrobial potency. The complex of 1-(3,4-dichlorophenyl)-3-[3-(trifluoromethyl)phenyl]thiourea (3) presented the highest activity against Gram-positive pathogens, even stronger than the activity of its non-complexed counterpart and the reference drug. The compound also prevented the biofilm formation of methicillin-resistant and standard strains of staphylococcal cocci. The title derivatives were found to be effective inhibitors of DNA gyrase and topoisomerase IV isolated from Staphylococcus aureus. The binding modes of the ligand L3 with DNA gyrase and topoisomerase IV were presented.
Collapse
|
211
|
Molecular modeling and docking of protease from Bacillus sp. for the keratin degradation. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2018. [DOI: 10.1016/j.bcab.2017.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
212
|
Selvam K, Senbagam D, Selvankumar T, Sudhakar C, Kamala-Kannan S, Senthilkumar B, Govarthanan M. Cellulase enzyme: Homology modeling, binding site identification and molecular docking. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.08.067] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
213
|
Shang R, Yi Y, Zhang C, Fu Y, Liang J, Pu W. Antibacterial activity and pharmacokinetic profile of a promising antibacterial agent: 14-O-[(4-Amino-6-hydroxy-pyrimidine-2-yl)thioacetyl] mutilin. Pharmacol Res 2017; 129:424-431. [PMID: 29133214 DOI: 10.1016/j.phrs.2017.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 01/21/2023]
Abstract
A new pleuromutilin derivative, 14-O-[(4-Amino-6-hydroxy-pyrimidine-2-yl)thioacetyl] mutilin (APTM), has been synthesized and proved most potent antibacterial agent in in vitro assays, suggesting that further development of this compound may lead to a promising antibacterial drug. In this study, we further evaluated the cytotoxicity, antibacterial efficacy and the pharmacokinetic profile of APTM. In BRL 3A cells, 50% of viability was obtained when 363μg/mL of APTM was used, while retapamulin and tiamulin fumarate needed 49 and 28μg/mL, respectively, to reach this viability. Compared to tiamulin fumarate, APTM showed higher inhibition efficacy and faster bactericidal activity against S. aureus and lower 50% effective dose (ED50) in mice after a lethal challenge with methicillin-resistant Staphylococcus aureus (MRSA). Docking experiment for APTM showed a similar binding pattern with tiamulin. Furthermore, a simple, accurate and sensitive HPLC method for the determination of APTM in rabbit plasma was developed and successfully applied to pharmacokinetic study, in which the half life (t1/2), clearance rate (Cl) and the area under the plasma concentration-time curve (AUC0→∞) were 3.37h, 0.35L/h/kg and 70.68μg·h/m, respectively.
Collapse
Affiliation(s)
- Ruofeng Shang
- Key Laboratory of New Animal Drug Project of Gansu Province; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, P. R. China; Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, 730050 Lanzhou, China.
| | - Yunpeng Yi
- Key Laboratory of New Animal Drug Project of Gansu Province; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, P. R. China; Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, 730050 Lanzhou, China
| | - Chao Zhang
- Lianyungang Animal Health Inspection Institute, Lianyungang, 222000, Lianyungang, China
| | - Yunxing Fu
- Key Laboratory of New Animal Drug Project of Gansu Province; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, P. R. China; Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, 730050 Lanzhou, China
| | - Jianping Liang
- Key Laboratory of New Animal Drug Project of Gansu Province; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, P. R. China; Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, 730050 Lanzhou, China
| | - Wanxia Pu
- Key Laboratory of New Animal Drug Project of Gansu Province; Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, P. R. China; Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, 730050 Lanzhou, China.
| |
Collapse
|
214
|
From cheminformatics to structure-based design: Web services and desktop applications based on the NAOMI library. J Biotechnol 2017; 261:207-214. [DOI: 10.1016/j.jbiotec.2017.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/31/2017] [Accepted: 06/07/2017] [Indexed: 02/06/2023]
|
215
|
Jha AB, Panchal SS. Neuroprotection and cognitive enhancement by treatment with γ-oryzanol in sporadic Alzheimer's disease. J Appl Biomed 2017. [DOI: 10.1016/j.jab.2017.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
216
|
Taha M, Ullah H, Al Muqarrabun LMR, Khan MN, Rahim F, Ahmat N, Ali M, Perveen S. Synthesis of bis-indolylmethanes as new potential inhibitors of β-glucuronidase and their molecular docking studies. Eur J Med Chem 2017; 143:1757-1767. [PMID: 29133042 DOI: 10.1016/j.ejmech.2017.10.071] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/01/2017] [Accepted: 10/26/2017] [Indexed: 10/18/2022]
Abstract
Thirty-two (32) bis-indolylmethane-hydrazone hybrids 1-32 were synthesized and characterized by 1HNMR, 13CNNMR and HREI-MS. All compounds were evaluated in vitro for β-glucuronidase inhibitory potential. All analogs showed varying degree of β-glucuronidase inhibitory potential ranging from 0.10 ± 0.01 to 48.50 ± 1.10 μM when compared with the standard drug d-saccharic acid-1,4-lactone (IC50 value 48.30 ± 1.20 μM). Derivatives 1-32 showed the highest β-glucuronidase inhibitory potentials which is many folds better than the standard drug d-saccharic acid-1,4-lactone. Further molecular docking study validated the experimental results. It was proposed that bis-indolylmethane may interact with some amino acid residues located within the active site of β-glucuronidase enzyme. This study has culminated in the identification of a new class of potent β-glucuronidase inhibitors.
Collapse
Affiliation(s)
- Muhammad Taha
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia.
| | - Hayat Ullah
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Laode Muhammad Ramadhan Al Muqarrabun
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia; Faculty of Applied Science Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor, Malaysia
| | - Muhammad Naseem Khan
- Department of Chemistry, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Norizan Ahmat
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia; Faculty of Applied Science Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor, Malaysia
| | - Muhammad Ali
- Department of Chemistry, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi 75280, Pakistan
| |
Collapse
|
217
|
Ma Y, Abdelnabi R, Delang L, Froeyen M, Luyten W, Neyts J, Mirabelli C. New class of early-stage enterovirus inhibitors with a novel mechanism of action. Antiviral Res 2017; 147:67-74. [PMID: 28993161 DOI: 10.1016/j.antiviral.2017.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/15/2017] [Accepted: 10/06/2017] [Indexed: 02/03/2023]
Abstract
4-dimethylamino benzoic acid (compound 12, synonym: 4EDMAB) was identified as an in vitro inhibitor of Coxsackie virus B3 (CVB3) replication in CPE-based assays (EC50 of 9.1 ± 1.5 μM). Next, the activity of twenty-three analogues was assessed, their structure-activity relationship was deduced and a more potent analogue was identified (EC50 of 2.6 ± 0.5 μM). The antiviral activity of 4EDMAB was further confirmed by quantifying viral RNA yield. Time-of-drug-addition assay revealed that 4EDMAB exerts its antiviral activity at the early stages of virus replication. Six compound-resistant viruses were selected and genotyped and all the mutations appeared to be in the capsid protein VP1. Reverse engineering showed that single mutants Y75C, A88V, A98V, D133N and R219K were respectively 15-, 2-, 4-, 17- and 76-fold resistant to 4EDMAB. The compound protected both wild type (WT) CVB3 and the five resistant mutants from heat inactivation. The plaque size produced by the A88V, D133N and R219K mutants was smaller than that of WT and these mutants were also more heat-sensitive than WT in the absence of the compound. These findings suggest that these three mutations increase virion capsid flexibility and compensate for the stabilizing effects of 4EDMAB. Molecular modelling suggests that the compound binds to a small cavity in VP1, which is different from the hydrophobic pocket in the canyon where typical capsid binders (such as pleconaril) bind. Modelling studies also suggest a direct ionic interaction between the negatively charged carboxylic group of 4EDMAB and the positively charged guanidino group of arginine 219. Moreover, the in vitro combination of 4EDMAB and pleconaril resulted in synergistic antiviral effect. In conclusion, 4EDMAB is a novel early-stage inhibitor, which targets VP1 with a mechanism that is different from that of known capsid binders.
Collapse
Affiliation(s)
- Yipeng Ma
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Belgium; Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Belgium
| | - Rana Abdelnabi
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Belgium
| | - Leen Delang
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Belgium
| | - Mathy Froeyen
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Belgium
| | - Walter Luyten
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Belgium
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Belgium.
| | - Carmen Mirabelli
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Belgium
| |
Collapse
|
218
|
Structural Basis for the Bidirectional Activity of Bacillus nanoRNase NrnA. Sci Rep 2017; 7:11085. [PMID: 28894100 PMCID: PMC5593865 DOI: 10.1038/s41598-017-09403-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022] Open
Abstract
NanoRNAs are RNA fragments 2 to 5 nucleotides in length that are generated as byproducts of RNA degradation and abortive transcription initiation. Cells have specialized enzymes to degrade nanoRNAs, such as the DHH phosphoesterase family member NanoRNase A (NrnA). This enzyme was originally identified as a 3′ → 5′ exonuclease, but we show here that NrnA is bidirectional, degrading 2–5 nucleotide long RNA oligomers from the 3′ end, and longer RNA substrates from the 5′ end. The crystal structure of Bacillus subtilis NrnA reveals a dynamic bi-lobal architecture, with the catalytic N-terminal DHH domain linked to the substrate binding C-terminal DHHA1 domain via an extended linker. Whereas this arrangement is similar to the structure of RecJ, a 5′ → 3′ DHH family DNase and other DHH family nanoRNases, Bacillus NrnA has gained an extended substrate-binding patch that we posit is responsible for its 3′ → 5′ activity.
Collapse
|
219
|
Barros-Álvarez X, Kerchner KM, Koh CY, Turley S, Pardon E, Steyaert J, Ranade RM, Gillespie JR, Zhang Z, Verlinde CLMJ, Fan E, Buckner FS, Hol WGJ. Leishmania donovani tyrosyl-tRNA synthetase structure in complex with a tyrosyl adenylate analog and comparisons with human and protozoan counterparts. Biochimie 2017; 138:124-136. [PMID: 28427904 PMCID: PMC5484532 DOI: 10.1016/j.biochi.2017.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 04/12/2017] [Indexed: 02/06/2023]
Abstract
The crystal structure of Leishmania donovani tyrosyl-tRNA synthetase (LdTyrRS) in complex with a nanobody and the tyrosyl adenylate analog TyrSA was determined at 2.75 Å resolution. Nanobodies are the variable domains of camelid heavy chain-only antibodies. The nanobody makes numerous crystal contacts and in addition reduces the flexibility of a loop of LdTyrRS. TyrSA is engaged in many interactions with active site residues occupying the tyrosine and adenine binding pockets. The LdTyrRS polypeptide chain consists of two pseudo-monomers, each consisting of two domains. Comparing the two independent chains in the asymmetric unit reveals that the two pseudo-monomers of LdTyrRS can bend with respect to each other essentially as rigid bodies. This flexibility might be useful in the positioning of tRNA for catalysis since both pseudo-monomers in the LdTyrRS chain are needed for charging tRNATyr. An "extra pocket" (EP) appears to be present near the adenine binding region of LdTyrRS. Since this pocket is absent in the two human homologous enzymes, the EP provides interesting opportunities for obtaining selective drugs for treating infections caused by L. donovani, a unicellular parasite causing visceral leishmaniasis, or kala azar, which claims 20,000 to 30,000 deaths per year. Sequence and structural comparisons indicate that the EP is a characteristic which also occurs in the active site of several other important pathogenic protozoa. Therefore, the structure of LdTyrRS could inspire the design of compounds useful for treating several different parasitic diseases.
Collapse
Affiliation(s)
- Ximena Barros-Álvarez
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Laboratorio de Enzimología de Parásitos, Facultad de Ciencias, Universidad de los Andes, Mérida, Venezuela
| | - Keshia M Kerchner
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Cho Yeow Koh
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Stewart Turley
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Els Pardon
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussel, Belgium; VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussel, Belgium; VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Ranae M Ranade
- Division of Allergy and Infectious Diseases, School of Medicine, University of Washington, Seattle, WA, USA
| | - J Robert Gillespie
- Division of Allergy and Infectious Diseases, School of Medicine, University of Washington, Seattle, WA, USA
| | - Zhongsheng Zhang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - Erkang Fan
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Frederick S Buckner
- Division of Allergy and Infectious Diseases, School of Medicine, University of Washington, Seattle, WA, USA
| | - Wim G J Hol
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| |
Collapse
|
220
|
Deng LJ, Wang LH, Peng CK, Li YB, Huang MH, Chen MF, Lei XP, Qi M, Cen Y, Ye WC, Zhang DM, Chen WM. Fibroblast Activation Protein α Activated Tripeptide Bufadienolide Antitumor Prodrug with Reduced Cardiotoxicity. J Med Chem 2017; 60:5320-5333. [DOI: 10.1021/acs.jmedchem.6b01755] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Li-Juan Deng
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Long-Hai Wang
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Cheng-Kang Peng
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Yi-Bin Li
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Mao-Hua Huang
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Min-Feng Chen
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Xue-Ping Lei
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Ming Qi
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Yun Cen
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Wen-Cai Ye
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Dong-Mei Zhang
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| | - Wei-Min Chen
- Guangdong Province Key Laboratory
of Pharmacodynamic Constituents of TCM and New Drugs Research, College
of Pharmacy, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
221
|
Lanosterol Suppresses the Aggregation and Cytotoxicity of Misfolded Proteins Linked with Neurodegenerative Diseases. Mol Neurobiol 2017; 55:1169-1182. [PMID: 28102469 DOI: 10.1007/s12035-016-0377-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 12/28/2016] [Indexed: 10/20/2022]
Abstract
Accumulation of misfolded or aberrant proteins in neuronal cells is linked with neurodegeneration and other pathologies. Which molecular mechanisms fail and cause inappropriate folding of proteins and what is their relationship to cellular toxicity is not well known. How does it happen and what are the probable therapeutic or molecular approaches to counter them are also not clear. Here, we demonstrate that treatment of lanosterol diminishes aberrant proteotoxic aggregation and mitigates their cytotoxicity via induced expression of co-chaperone CHIP and elevated autophagy. The addition of lanosterol not only reduces aggregation of mutant bonafide misfolded proteins but also effectively prevents accumulation of various mutant disease-prone proteotoxic proteins. Finally, we observed that lanosterol mitigates cytotoxicity in cells, mediated by different stress-inducing agents. Taken together, our present results suggest that upregulation of cellular molecular chaperones, primarily using small molecules, can probably offer an efficient therapeutic approach in the future against misfolding of different disease-causing proteins and neurodegenerative disorders. Graphical Abstract ᅟ.
Collapse
|
222
|
Yazawa K, Gimenez-Dejoz J, Masunaga H, Hikima T, Numata K. Chemoenzymatic synthesis of a peptide containing nylon monomer units for thermally processable peptide material application. Polym Chem 2017. [DOI: 10.1039/c7py00770a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chemoenzymatic polymerization by papain produced the copolymer of l-leucine and nylon monomers, which showed melting behavior at around 200 °C.
Collapse
Affiliation(s)
- Kenjiro Yazawa
- Enzyme Research Team
- RIKEN Center for Sustainable Resource Science
- Wako-shi
- Japan
| | - Joan Gimenez-Dejoz
- Enzyme Research Team
- RIKEN Center for Sustainable Resource Science
- Wako-shi
- Japan
| | - Hiroyasu Masunaga
- Japan Synchrotron Radiation Research Institute
- Sayo-gun
- Japan
- RIKEN SPring-8 Center
- Sayo-gun
| | | | - Keiji Numata
- Enzyme Research Team
- RIKEN Center for Sustainable Resource Science
- Wako-shi
- Japan
| |
Collapse
|
223
|
Hermosilla P, Estrada J, Guallar V, Ropinski T, Vinacua A, Vazquez PP. Physics-Based Visual Characterization of Molecular Interaction Forces. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2017; 23:731-740. [PMID: 27875187 DOI: 10.1109/tvcg.2016.2598825] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Molecular simulations are used in many areas of biotechnology, such as drug design and enzyme engineering. Despite the development of automatic computational protocols, analysis of molecular interactions is still a major aspect where human comprehension and intuition are key to accelerate, analyze, and propose modifications to the molecule of interest. Most visualization algorithms help the users by providing an accurate depiction of the spatial arrangement: the atoms involved in inter-molecular contacts. There are few tools that provide visual information on the forces governing molecular docking. However, these tools, commonly restricted to close interaction between atoms, do not consider whole simulation paths, long-range distances and, importantly, do not provide visual cues for a quick and intuitive comprehension of the energy functions (modeling intermolecular interactions) involved. In this paper, we propose visualizations designed to enable the characterization of interaction forces by taking into account several relevant variables such as molecule-ligand distance and the energy function, which is essential to understand binding affinities. We put emphasis on mapping molecular docking paths obtained from Molecular Dynamics or Monte Carlo simulations, and provide time-dependent visualizations for different energy components and particle resolutions: atoms, groups or residues. The presented visualizations have the potential to support domain experts in a more efficient drug or enzyme design process.
Collapse
|
224
|
HajiEbrahimi A, Ghafouri H, Ranjbar M, Sakhteman A. Protein Ligand Interaction Fingerprints. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A most challenging part in docking-based virtual screening is the scoring functions implemented in various docking programs in order to evaluate different poses of the ligands inside the binding cavity of the receptor. Precise and trustable measurement of ligand-protein affinity for Structure-Based Virtual Screening (SB-VS) is therefore, an outstanding problem in docking studies. Empirical post-docking filters can be helpful as a way to provide various types of structure-activity information. Different types of interaction have been presented between the ligands and the receptor so far. Based on the diversity and importance of PLIF methods, this chapter will focus on the comparison of different protocols. The advantages and disadvantages of all methods will be discussed explicitly in this chapter as well as future sights for further progress in this field. Different classifications approaches for the protein-ligand interaction fingerprints were also discussed in this chapter.
Collapse
|
225
|
Frączek T. Simulation-Based Algorithm for Two-Dimensional Chemical Structure Diagram Generation of Complex Molecules and Ligand–Protein Interactions. J Chem Inf Model 2016; 56:2320-2335. [DOI: 10.1021/acs.jcim.6b00391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Tomasz Frączek
- Institute of Applied Radiation
Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| |
Collapse
|
226
|
Affaticati PE, Dai SB, Payongsri P, Hailes HC, Tittmann K, Dalby PA. Structural Analysis of an Evolved Transketolase Reveals Divergent Binding Modes. Sci Rep 2016; 6:35716. [PMID: 27767080 PMCID: PMC5073344 DOI: 10.1038/srep35716] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/22/2016] [Indexed: 11/09/2022] Open
Abstract
The S385Y/D469T/R520Q variant of E. coli transketolase was evolved previously with three successive smart libraries, each guided by different structural, bioinformatical or computational methods. Substrate-walking progressively shifted the target acceptor substrate from phosphorylated aldehydes, towards a non-phosphorylated polar aldehyde, a non-polar aliphatic aldehyde, and finally a non-polar aromatic aldehyde. Kinetic evaluations on three benzaldehyde derivatives, suggested that their active-site binding was differentially sensitive to the S385Y mutation. Docking into mutants generated in silico from the wild-type crystal structure was not wholly satisfactory, as errors accumulated with successive mutations, and hampered further smart-library designs. Here we report the crystal structure of the S385Y/D469T/R520Q variant, and molecular docking of three substrates. This now supports our original hypothesis that directed-evolution had generated an evolutionary intermediate with divergent binding modes for the three aromatic aldehydes tested. The new active site contained two binding pockets supporting π-π stacking interactions, sterically separated by the D469T mutation. While 3-formylbenzoic acid (3-FBA) preferred one pocket, and 4-FBA the other, the less well-accepted substrate 3-hydroxybenzaldehyde (3-HBA) was caught in limbo with equal preference for the two pockets. This work highlights the value of obtaining crystal structures of evolved enzyme variants, for continued and reliable use of smart library strategies.
Collapse
Affiliation(s)
- Pierre E Affaticati
- Department of Biochemical Engineering, Gordon Street, University College London, WC1H 0AH, UK
| | - Shao-Bo Dai
- Albrecht-von-Haller Institute, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, 37077 Göttingen, Germany
| | - Panwajee Payongsri
- Department of Biochemical Engineering, Gordon Street, University College London, WC1H 0AH, UK
| | - Helen C Hailes
- Department of Chemistry, 20 Gordon Street, University College London, WC1H 0AJ, UK
| | - Kai Tittmann
- Albrecht-von-Haller Institute, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, 37077 Göttingen, Germany
| | - Paul A Dalby
- Department of Biochemical Engineering, Gordon Street, University College London, WC1H 0AH, UK
| |
Collapse
|
227
|
von Behren MM, Bietz S, Nittinger E, Rarey M. mRAISE: an alternative algorithmic approach to ligand-based virtual screening. J Comput Aided Mol Des 2016; 30:583-94. [PMID: 27565795 DOI: 10.1007/s10822-016-9940-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/17/2016] [Indexed: 01/31/2023]
Abstract
Ligand-based virtual screening is a well established method to find new lead molecules in todays drug discovery process. In order to be applicable in day to day practice, such methods have to face multiple challenges. The most important part is the reliability of the results, which can be shown and compared in retrospective studies. Furthermore, in the case of 3D methods, they need to provide biologically relevant molecular alignments of the ligands, that can be further investigated by a medicinal chemist. Last but not least, they have to be able to screen large databases in reasonable time. Many algorithms for ligand-based virtual screening have been proposed in the past, most of them based on pairwise comparisons. Here, a new method is introduced called mRAISE. Based on structural alignments, it uses a descriptor-based bitmap search engine (RAISE) to achieve efficiency. Alignments created on the fly by the search engine get evaluated with an independent shape-based scoring function also used for ranking of compounds. The correct ranking as well as the alignment quality of the method are evaluated and compared to other state of the art methods. On the commonly used Directory of Useful Decoys dataset mRAISE achieves an average area under the ROC curve of 0.76, an average enrichment factor at 1 % of 20.2 and an average hit rate at 1 % of 55.5. With these results, mRAISE is always among the top performing methods with available data for comparison. To access the quality of the alignments calculated by ligand-based virtual screening methods, we introduce a new dataset containing 180 prealigned ligands for 11 diverse targets. Within the top ten ranked conformations, the alignment closest to X-ray structure calculated with mRAISE has a root-mean-square deviation of less than 2.0 Å for 80.8 % of alignment pairs and achieves a median of less than 2.0 Å for eight of the 11 cases. The dataset used to rate the quality of the calculated alignments is freely available at http://www.zbh.uni-hamburg.de/mraise-dataset.html . The table of all PDB codes contained in the ensembles can be found in the supplementary material. The software tool mRAISE is freely available for evaluation purposes and academic use (see http://www.zbh.uni-hamburg.de/raise ).
Collapse
Affiliation(s)
- Mathias M von Behren
- ZBH - Center for Bioinformatics, University of Hamburg, Bundesstraße 43, 20146, Hamburg, Germany
| | - Stefan Bietz
- ZBH - Center for Bioinformatics, University of Hamburg, Bundesstraße 43, 20146, Hamburg, Germany
| | - Eva Nittinger
- ZBH - Center for Bioinformatics, University of Hamburg, Bundesstraße 43, 20146, Hamburg, Germany
| | - Matthias Rarey
- ZBH - Center for Bioinformatics, University of Hamburg, Bundesstraße 43, 20146, Hamburg, Germany.
| |
Collapse
|
228
|
Synthesis, Biological Evaluation and Molecular Modelling of 2'-Hydroxychalcones as Acetylcholinesterase Inhibitors. Molecules 2016; 21:molecules21070955. [PMID: 27455222 PMCID: PMC6273369 DOI: 10.3390/molecules21070955] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 11/25/2022] Open
Abstract
A series of 2′-hydroxy- and 2′-hydroxy-4′,6′-dimethoxychalcones was synthesised and evaluated as inhibitors of human acetylcholinesterase (AChE). The majority of the compounds were found to show some activity, with the most active compounds having IC50 values of 40–85 µM. Higher activities were generally observed for compounds with methoxy substituents in the A ring and halogen substituents in the B ring. Kinetic studies on the most active compounds showed that they act as mixed-type inhibitors, in agreement with the results of molecular modelling studies, which suggested that they interact with residues in the peripheral anionic site and the gorge region of AChE.
Collapse
|
229
|
A simple enzyme-substrate localized conjugation method to generate immobilized, functional glutathione S-transferase fusion protein columns for affinity enrichment. Anal Biochem 2016; 505:51-8. [PMID: 27063248 DOI: 10.1016/j.ab.2016.03.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/19/2016] [Accepted: 03/30/2016] [Indexed: 12/28/2022]
Abstract
Immobilized protein receptors and enzymes are tools for isolating or enriching ligands and substrates based on affinity. For example, glutathione S-transferase (GST) is fused to proteins as a tag for binding to its substrate glutathione (GSH) linked to solid supports. One issue with this approach is that high-affinity interactions between receptors and ligands require harsh elution conditions such as low pH, which can result in leached receptor. Another issue is the inherent nonspecific chemical conjugation of reactive groups such as N-hydroxysuccinimide (NHS) that couple lysines to solid supports; the nonspecificity of NHS may result in residue modifications near the binding site(s) of the receptor that can affect ligand specificity. In this study, a simple conjugation procedure is presented that overcomes these limitations and results in immobilized GST fusion proteins that are functional and specific. Here, the affinity of GST for GSH was used to generate an enzyme-substrate site-specific cross-linking reaction; GSH-Sepharose was preactivated with 1-ethyl-3-(dimethylaminopropyl)carbodiimide (EDC) and then incubated Fc gamma receptor IIIa (FcγRIIIa)-GST. The immobilized FcγRIIIa-GST more specifically bound glycosylated immunoglobulin G1s (IgG1s) and was used to enrich nonfucosylated IgG1s from weaker binding species. This technique can be used when modifications of amino acids lead to changes in activity.
Collapse
|
230
|
Fragment oriented molecular shapes. J Mol Graph Model 2016; 66:143-54. [PMID: 27085751 DOI: 10.1016/j.jmgm.2016.03.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 11/23/2022]
Abstract
Molecular shape is an important concept in drug design and virtual screening. Shape similarity typically uses either alignment methods, which dynamically optimize molecular poses with respect to the query molecular shape, or feature vector methods, which are computationally less demanding but less accurate. The computational cost of alignment can be reduced by pre-aligning shapes, as is done with the Volumetric-Aligned Molecular Shapes (VAMS) method. Here, we introduce and evaluate fragment oriented molecular shapes (FOMS), where shapes are aligned based on molecular fragments. FOMS enables the use of shape constraints, a novel method for precisely specifying molecular shape queries that provides the ability to perform partial shape matching and supports search algorithms that function on an interactive time scale. When evaluated using the challenging Maximum Unbiased Validation dataset, shape constraints were able to extract significantly enriched subsets of compounds for the majority of targets, and FOMS matched or exceeded the performance of both VAMS and an optimizing alignment method of shape similarity search.
Collapse
|
231
|
5-HT2 receptor affinity, docking studies and pharmacological evaluation of a series of 1,3-disubstituted thiourea derivatives. Eur J Med Chem 2016; 116:173-186. [PMID: 27061981 DOI: 10.1016/j.ejmech.2016.03.073] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/18/2016] [Accepted: 03/25/2016] [Indexed: 01/06/2023]
Abstract
A series of 10 thiourea derivatives have been synthesized by the reaction of aromatic amine with a substituted aryl (compounds 1-3, 6-8) and alkylphenyl (4, 5, 9, 10) isothiocyanates. Their in vitro and in vivo pharmacological properties were studied. Among the evaluated compounds, two displayed very high affinity for the 5-HT2A receptor (1-0.043 nM and 5-0.6 nM), being selective over the 5-HT2C receptor. Derivatives 3, 5, 9, 10 by 70-89% diminished L-5-HTP-induced head twitch episodes. Compounds 1 and 5 as the 5-HT2A receptor antagonists produced a dose-dependent decrease in the number of DOI-elicited HTR. Compounds 1-5 strongly reduced amphetamine-evoked hyperactivity in rodents. In another test, 1 and 2 caused hyperthermia in mice, whereas 9 and 10 led to hypothermia. Antinociceptive and anticonvulsant properties of selected derivatives were demonstrated. Molecular docking studies using a homology model of 5-HT2A revealed a significant role of hydrogen bonds between both thiourea NH groups and Asp155/Tyr370 residues, as well as π-π interaction with Phe339.
Collapse
|
232
|
Hati S, Bhattacharyya S. Incorporating modeling and simulations in undergraduate biophysical chemistry course to promote understanding of structure-dynamics-function relationships in proteins. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 44:140-159. [PMID: 26801683 DOI: 10.1002/bmb.20942] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/15/2015] [Indexed: 06/05/2023]
Abstract
A project-based biophysical chemistry laboratory course, which is offered to the biochemistry and molecular biology majors in their senior year, is described. In this course, the classroom study of the structure-function of biomolecules is integrated with the discovery-guided laboratory study of these molecules using computer modeling and simulations. In particular, modern computational tools are employed to elucidate the relationship between structure, dynamics, and function in proteins. Computer-based laboratory protocols that we introduced in three modules allow students to visualize the secondary, super-secondary, and tertiary structures of proteins, analyze non-covalent interactions in protein-ligand complexes, develop three-dimensional structural models (homology model) for new protein sequences and evaluate their structural qualities, and study proteins' intrinsic dynamics to understand their functions. In the fourth module, students are assigned to an authentic research problem, where they apply their laboratory skills (acquired in modules 1-3) to answer conceptual biophysical questions. Through this process, students gain in-depth understanding of protein dynamics-the missing link between structure and function. Additionally, the requirement of term papers sharpens students' writing and communication skills. Finally, these projects result in new findings that are communicated in peer-reviewed journals.
Collapse
Affiliation(s)
- Sanchita Hati
- Department of Chemistry, University Wisconsin, Eau Claire, Wisconsin
| | | |
Collapse
|
233
|
Biocca S, Iacovelli F, Matarazzo S, Vindigni G, Oteri F, Desideri A, Falconi M. Molecular mechanism of statin-mediated LOX-1 inhibition. Cell Cycle 2016; 14:1583-95. [PMID: 25950192 DOI: 10.1080/15384101.2015.1026486] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Statins are largely used in clinics in the treatment of patients with cardiovascular diseases for their effect on lowering circulating cholesterol. Lectin-like oxidized low-density lipoprotein (LOX-1), the primary receptor for ox-LDL, plays a central role in the pathogenesis of atherosclerosis and cardiovascular disorders. We have recently shown that chronic exposure of cells to lovastatin disrupts LOX-1 receptor cluster distribution in plasma membranes, leading to a marked loss of LOX-1 function. Here we investigated the molecular mechanism of statin-mediated LOX-1 inhibition and we demonstrate that all tested statins are able to displace the binding of fluorescent ox-LDL to LOX-1 by a direct interaction with LOX-1 receptors in a cell-based binding assay. Molecular docking simulations confirm the interaction and indicate that statins completely fill the hydrophobic tunnel that crosses the C-type lectin-like (CTLD) recognition domain of LOX-1. Classical molecular dynamics simulation technique applied to the LOX-1 CTLD, considered in the entire receptor structure with or without a statin ligand inside the tunnel, indicates that the presence of a ligand largely increases the dimer stability. Electrophoretic separation and western blot confirm that different statins binding stabilize the dimer assembly of LOX-1 receptors in vivo. The simulative and experimental results allow us to propose a CTLD clamp motion, which enables the receptor-substrate coupling. These findings reveal a novel and significant functional effect of statins.
Collapse
Key Words
- Ato, atorvastatin
- CTLD, C-type lectin-like domain
- Cav-1, caveolin-1
- DMEM, Dulbecco's modified Eagle's medium
- DiI, 1,1′-dioctadecyl-3,3,3′,3′-tetramethyllindocarbocyanine perchlorate
- Flu, fluvastatin
- HEK, human embryonic kidney
- HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A
- LDL, low-density lipoprotein
- LDL-C, low-density lipoprotein-cholesterol
- LOX-1 receptor
- LOX-1, lectin-like oxidized low-density lipoprotein receptor-1
- Lov, lovastatin
- Mab, monoclonal antibody
- Pra, pravastatin
- molecular docking
- molecular dynamics simulation
- monomer-dimer ratio
- ox-LDL, oxidized low-density lipoprotein
- statin
- substrate recognition
Collapse
Affiliation(s)
- Silvia Biocca
- a Department of Systems Medicine and Center of Biostatistics and Bioinformatics ; University of Rome Tor Vergata ; Rome , Italy
| | | | | | | | | | | | | |
Collapse
|
234
|
Novel Promising Estrogenic Receptor Modulators: Cytotoxic and Estrogenic Activity of Benzanilides and Dithiobenzanilides. PLoS One 2016; 11:e0145615. [PMID: 26730945 PMCID: PMC4701221 DOI: 10.1371/journal.pone.0145615] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 12/07/2015] [Indexed: 11/24/2022] Open
Abstract
The cytotoxicity of 27 benzanilides and dithiobenzanilides built on a stilbene scaffold and possessing various functional groups in aromatic rings previously described for their spasmolytic properties was assayed on three human cancer cell lines (A549 –lung adenocarcinoma, MCF-7 estrogen dependent breast adenocarcinoma and MDA-MB-231 estrogen independent breast adenocarcinoma) and 2 non-tumorigenic cell lines (CCD39Lu–lung fibroblasts, MCF-12A - breast epithelial). Three compounds (6, 15 and 18) showed selective antiproliferative activity against estrogen dependent MCF-7 cancer cells and their estrogenic activity was further confirmed in MCF-7 transfected with an estrogen receptor reporter plasmid and in HEK239 cells over-expressing the estrogen receptor alpha (ERα). Compound 18 is especially interesting as a potential candidate for therapy since it is highly toxic and selective towards estrogen dependent MCF7 cell lines (IC50 = 5.07 μM versus more than 100 μM for MDA-MB-231) and almost innocuous for normal breast cells (IC50 = 91.46 μM for MCF-12A). Docking studies have shown that compound 18 interacts with the receptor in the same cavity as estradiol although the extra aromatic ring is involved in additional binding interactions with residue W383. The role of W383 and the extended binding mode were confirmed by site-directed mutagenesis.
Collapse
|
235
|
Abstract
Web-based protein structure databases come in a wide variety of types and levels of information content. Those having the most general interest are the various atlases that describe each experimentally determined protein structure and provide useful links, analyses, and schematic diagrams relating to its 3D structure and biological function. Also of great interest are the databases that classify 3D structures by their folds as these can reveal evolutionary relationships which may be hard to detect from sequence comparison alone. Related to these are the numerous servers that compare folds-particularly useful for newly solved structures, and especially those of unknown function. Beyond these are a vast number of databases for the more specialized user, dealing with specific families, diseases, structural features, and so on.
Collapse
Affiliation(s)
- Roman A Laskowski
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
| |
Collapse
|
236
|
Manhas A, Kumar SP, Jha PC. Molecular modeling of Plasmodium falciparum peptide deformylase and structure-based pharmacophore screening for inhibitors. RSC Adv 2016. [DOI: 10.1039/c6ra01071g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The role of metal coordination geometry and actinonin (inhibitor) binding was examined to develop pharmacophore-based inhibitor design strategy forPlasmodium falciparumpeptide deformylase.
Collapse
Affiliation(s)
- Anu Manhas
- School of Chemical Sciences
- Central University of Gujarat
- Gandhinagar-382030
- India
| | | | - Prakash Chandra Jha
- School of Chemical Sciences
- Central University of Gujarat
- Gandhinagar-382030
- India
| |
Collapse
|
237
|
Lemes LFN, de Andrade Ramos G, de Oliveira AS, da Silva FMR, de Castro Couto G, da Silva Boni M, Guimarães MJR, Souza INO, Bartolini M, Andrisano V, do Nascimento Nogueira PC, Silveira ER, Brand GD, Soukup O, Korábečný J, Romeiro NC, Castro NG, Bolognesi ML, Romeiro LAS. Cardanol-derived AChE inhibitors: Towards the development of dual binding derivatives for Alzheimer's disease. Eur J Med Chem 2016; 108:687-700. [DOI: 10.1016/j.ejmech.2015.12.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/12/2015] [Accepted: 12/13/2015] [Indexed: 12/22/2022]
|
238
|
Grulich M, Brezovský J, ŠtĿpánek V, Palyzová A, Kyslíková E, Kyslík P. Resolution of α/β-amino acids by enantioselective penicillin G acylase from Achromobacter sp . ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.molcatb.2015.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
239
|
Durrant JD, Carlson KE, Martin TA, Offutt TL, Mayne CG, Katzenellenbogen JA, Amaro RE. Neural-Network Scoring Functions Identify Structurally Novel Estrogen-Receptor Ligands. J Chem Inf Model 2015; 55:1953-61. [PMID: 26286148 PMCID: PMC4780411 DOI: 10.1021/acs.jcim.5b00241] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The magnitude of the investment required to bring a drug to the market hinders medical progress, requiring hundreds of millions of dollars and years of research and development. Any innovation that improves the efficiency of the drug-discovery process has the potential to accelerate the delivery of new treatments to countless patients in need. "Virtual screening," wherein molecules are first tested in silico in order to prioritize compounds for subsequent experimental testing, is one such innovation. Although the traditional scoring functions used in virtual screens have proven useful, improved accuracy requires novel approaches. In the current work, we use the estrogen receptor to demonstrate that neural networks are adept at identifying structurally novel small molecules that bind to a selected drug target, ultimately allowing experimentalists to test fewer compounds in the earliest stages of lead identification while obtaining higher hit rates. We describe 39 novel estrogen-receptor ligands identified in silico with experimentally determined Ki values ranging from 460 nM to 20 μM, presented here for the first time.
Collapse
Affiliation(s)
- Jacob D. Durrant
- Department of Chemistry & Biochemistry and the National Biomedical Computation Resource, University of California, San Diego, La Jolla, CA, 92093
| | - Kathryn E. Carlson
- Department of Chemistry, University of Illinois at Urbana-Champaign, Champaign, IL, 61801
| | - Teresa A. Martin
- Department of Chemistry, University of Illinois at Urbana-Champaign, Champaign, IL, 61801
| | - Tavina L. Offutt
- Department of Chemistry & Biochemistry and the National Biomedical Computation Resource, University of California, San Diego, La Jolla, CA, 92093
| | - Christopher G. Mayne
- Department of Chemistry, University of Illinois at Urbana-Champaign, Champaign, IL, 61801
| | | | - Rommie E. Amaro
- Department of Chemistry & Biochemistry and the National Biomedical Computation Resource, University of California, San Diego, La Jolla, CA, 92093
| |
Collapse
|
240
|
Song M, Li JL, Li XP, Kan SF. Targeting Human Poly(ADP-Ribose) Polymerase-1 with Natural Medicines and Its Potential Applications in Ovarian Cancer Therapeutics. Arch Pharm (Weinheim) 2015; 348:817-823. [PMID: 26344206 DOI: 10.1002/ardp.201500183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 08/09/2015] [Accepted: 08/18/2015] [Indexed: 01/04/2023]
Abstract
Targeting poly(ADP-ribose) polymerase-1 (PARP-1) has been established as an efficient therapeutics for advanced ovarian cancer. In this study, we describe an integrated procedure that combines virtual computer screening and an experimental enzyme assay to discover novel potent PARP-1 inhibitors from more than 130000 commercially available natural products. The protocol employed a stepwise strategy to fast exclude typical PARP-1 non-binders and then performing rigorous prediction to identify promising candidates with high potency against PARP-1. Consequently, eight natural products were hit and tested to determine their inhibitory activities against the PARP-1 catalytic domain. From these, four compounds, i.e., puerarin, phloretin, chlorogenic acid, and biochanin A, were found to have high or moderate potencies with inhibitory IC50 values of 6, 470, 25, and 86 nM, respectively. The values are comparable to that (IC50 = 1.94 nM) of the FDA-approved agent olaparib. Structural and energetic analyses of the modeled structures of the PARP-1 catalytic domain complexed with the newly identified inhibitors revealed a common binding mode in the complexes: the active site of PARP-1 is composed of a thin polar helix and a flat non-polar pocket; the inhibitors can form a number of hydrogen bonds and electrostatic forces with the helix, while tightly packing against the pocket to define chemical interactions.
Collapse
Affiliation(s)
- Min Song
- Department of Gynaecology and Obstetrics, Jinan Maternity and Child Care Hospital, Jinan, P. R. China
| | - Jun-Lan Li
- Pharmacy Intravenous Admixture Services, Weifang People's Hospital, Weifang, P. R. China
| | - Xiao-Ping Li
- Department of Gynaecology and Obstetrics, Jinan Maternity and Child Care Hospital, Jinan, P. R. China
| | - Shi-Feng Kan
- Department of Oncology, Zaozhuang Municipal Hospital, Zaozhuang, P. R. China
| |
Collapse
|
241
|
Guo Y, Wolter T, Kubař T, Sumser M, Trauner D, Elstner M. Molecular Dynamics Investigation of gluazo, a Photo-Switchable Ligand for the Glutamate Receptor GluK2. PLoS One 2015; 10:e0135399. [PMID: 26308344 PMCID: PMC4550381 DOI: 10.1371/journal.pone.0135399] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/21/2015] [Indexed: 11/25/2022] Open
Abstract
Photochromic ligands (PCLs), defined as photoswitchable molecules that are able to endow native receptors with a sensitivity towards light, have become a promising photopharmacological tool for various applications in biology. In general, PCLs consist of a ligand of the target receptor covalently linked to an azobenzene, which can be reversibly switched between two configurations upon light illumination. Gluazo, as a PCL that targets excitatory amino acid receptors, in its dark-adapted trans iso-form was characterized to be a partial agonist of the kainate glutamate receptor GluK2. Application of UV light leads to the formation of the cis form, with remarkedly reduced affinity towards GluK2. The mechanism of the change of ligand affinity induced by the photoisomerization was unresolved. The presented computational study explains how the isomerization of such a PCL affects the structural changes in the target receptor that lead to its activation.
Collapse
Affiliation(s)
- Yanan Guo
- Department of Theoretical Chemical Biology, Institute of Physical Chemistry, Karlsruhe Institute of Technology, Kaiserstr. 12, 76131, Karlsruhe, Germany
| | - Tino Wolter
- Department of Theoretical Chemical Biology, Institute of Physical Chemistry, Karlsruhe Institute of Technology, Kaiserstr. 12, 76131, Karlsruhe, Germany
| | - Tomáš Kubař
- Department of Theoretical Chemical Biology, Institute of Physical Chemistry, Karlsruhe Institute of Technology, Kaiserstr. 12, 76131, Karlsruhe, Germany
| | - Martin Sumser
- Department of Chemistry, Ludwig-Maximilians-Universität München and Center of Integrated Protein Science, Butenandtstr. 5–13, 81377 Munich, Germany
| | - Dirk Trauner
- Department of Chemistry, Ludwig-Maximilians-Universität München and Center of Integrated Protein Science, Butenandtstr. 5–13, 81377 Munich, Germany
| | - Marcus Elstner
- Department of Theoretical Chemical Biology, Institute of Physical Chemistry, Karlsruhe Institute of Technology, Kaiserstr. 12, 76131, Karlsruhe, Germany
| |
Collapse
|
242
|
Nehybova T, Smarda J, Daniel L, Brezovsky J, Benes P. Wedelolactone induces growth of breast cancer cells by stimulation of estrogen receptor signalling. J Steroid Biochem Mol Biol 2015; 152:76-83. [PMID: 25934092 DOI: 10.1016/j.jsbmb.2015.04.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/09/2015] [Accepted: 04/26/2015] [Indexed: 01/14/2023]
Abstract
Wedelolactone, a plant coumestan, was shown to act as anti-cancer agent for breast and prostate carcinomas in vitro and in vivo targeting multiple cellular proteins including androgen receptors, 5-lipoxygenase and topoisomerase IIα. It is cytotoxic to breast, prostate, pituitary and myeloma cancer cell lines in vitro at μM concentrations. In this study, however, a novel biological activity of nM dose of wedelolactone was demonstrated. Wedelolactone acts as agonist of estrogen receptors (ER) α and β as demonstrated by transactivation of estrogen response element (ERE) in cells transiently expressing either ERα or ERβ and by molecular docking of this coumestan into ligand binding pocket of both ERα and ERβ. In breast cancer cells, wedelolactone stimulates growth of estrogen receptor-positive cells, expression of estrogen-responsive genes and activates rapid non-genomic estrogen signalling. All these effects can be inhibited by pretreatment with pure ER antagonist ICI 182,780 and they are not observed in ER-negative breast cancer cells. We conclude that wedelolactone acts as phytoestrogen in breast cancer cells by stimulating ER genomic and non-genomic signalling pathways.
Collapse
Affiliation(s)
- Tereza Nehybova
- Laboratory of Cellular Differentiation, Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5/A36, 625 00 Brno, Czech Republic
| | - Jan Smarda
- Laboratory of Cellular Differentiation, Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5/A36, 625 00 Brno, Czech Republic; Masaryk Memorial Cancer Institute, RECAMO, Zluty kopec 7, 656 53 Brno, Czech Republic
| | - Lukas Daniel
- Loschmidt Laboratories, Department of Experimental Biology and Research Centre for Toxic Compounds in the Environment RECETOX, Faculty of Science, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Pekarska 53, 656 91 Brno, Czech Republic
| | - Jan Brezovsky
- Loschmidt Laboratories, Department of Experimental Biology and Research Centre for Toxic Compounds in the Environment RECETOX, Faculty of Science, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Pekarska 53, 656 91 Brno, Czech Republic
| | - Petr Benes
- Laboratory of Cellular Differentiation, Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5/A36, 625 00 Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Pekarska 53, 656 91 Brno, Czech Republic.
| |
Collapse
|
243
|
Genome-wide Screening of Human Papillomavirus-Specific CTL Epitopes Presented by HLA-A Alleles in Cervical Cancer. Int J Pept Res Ther 2015. [DOI: 10.1007/s10989-015-9480-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
244
|
Roccatano D. Structure, dynamics, and function of the monooxygenase P450 BM-3: insights from computer simulations studies. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2015; 27:273102. [PMID: 26061496 DOI: 10.1088/0953-8984/27/27/273102] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The monooxygenase P450 BM-3 is a NADPH-dependent fatty acid hydroxylase enzyme isolated from soil bacterium Bacillus megaterium. As a pivotal member of cytochrome P450 superfamily, it has been intensely studied for the comprehension of structure-dynamics-function relationships in this class of enzymes. In addition, due to its peculiar properties, it is also a promising enzyme for biochemical and biomedical applications. However, despite the efforts, the full understanding of the enzyme structure and dynamics is not yet achieved. Computational studies, particularly molecular dynamics (MD) simulations, have importantly contributed to this endeavor by providing new insights at an atomic level regarding the correlations between structure, dynamics, and function of the protein. This topical review summarizes computational studies based on MD simulations of the cytochrome P450 BM-3 and gives an outlook on future directions.
Collapse
Affiliation(s)
- Danilo Roccatano
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany
| |
Collapse
|
245
|
The Calcium Goes Meow: Effects of Ions and Glycosylation on Fel d 1, the Major Cat Allergen. PLoS One 2015. [PMID: 26134118 DOI: 10.1371/journal.pone.0132311.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The major cat allergen, Fel d 1, is a structurally complex protein with two N-glycosylation sites that may be filled by different glycoforms. In addition, the protein contains three putative Ca2+ binding sites. Since the impact of these Fel d 1 structure modifications on the protein dynamics, physiology and pathology are not well established, the present work employed computational biology techniques to tackle these issues. While conformational effects brought upon by glycosylation were identified, potentially involved in cavity volume regulation, our results indicate that only the central Ca2+ ion remains coordinated to Fel d 1 in biological solutions, impairing its proposed role in modulating phospholipase A2 activity. As these results increase our understanding of Fel d 1 structural biology, they may offer new support for understanding its physiological role and impact into cat-promoted allergy.
Collapse
|
246
|
Ligabue-Braun R, Sachett LG, Pol-Fachin L, Verli H. The Calcium Goes Meow: Effects of Ions and Glycosylation on Fel d 1, the Major Cat Allergen. PLoS One 2015; 10:e0132311. [PMID: 26134118 PMCID: PMC4489793 DOI: 10.1371/journal.pone.0132311] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 06/11/2015] [Indexed: 12/20/2022] Open
Abstract
The major cat allergen, Fel d 1, is a structurally complex protein with two N-glycosylation sites that may be filled by different glycoforms. In addition, the protein contains three putative Ca2+ binding sites. Since the impact of these Fel d 1 structure modifications on the protein dynamics, physiology and pathology are not well established, the present work employed computational biology techniques to tackle these issues. While conformational effects brought upon by glycosylation were identified, potentially involved in cavity volume regulation, our results indicate that only the central Ca2+ ion remains coordinated to Fel d 1 in biological solutions, impairing its proposed role in modulating phospholipase A2 activity. As these results increase our understanding of Fel d 1 structural biology, they may offer new support for understanding its physiological role and impact into cat-promoted allergy.
Collapse
Affiliation(s)
- Rodrigo Ligabue-Braun
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Liana Guimarães Sachett
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Laércio Pol-Fachin
- Departamento de Química Fundamental, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Hugo Verli
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- * E-mail:
| |
Collapse
|
247
|
Enzymatic Synthesis of Galactosylated Serine/Threonine Derivatives by β-Galactosidase from Escherichia coli. Int J Mol Sci 2015; 16:13714-28. [PMID: 26084049 PMCID: PMC4490519 DOI: 10.3390/ijms160613714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/11/2015] [Indexed: 01/08/2023] Open
Abstract
The transgalactosylations of serine/threonine derivatives were investigated using β-galactosidase from Escherichia coli as biocatalyst. Using ortho-nitrophenyl-β-d-galactoside as donor, the highest bioconversion yield of transgalactosylated N-carboxy benzyl l-serine benzyl ester (23.2%) was achieved in heptane:buffer medium (70:30), whereas with the lactose, the highest bioconversion yield (3.94%) was obtained in the buffer reaction system. The structures of most abundant galactosylated serine products were characterized by MS/MS. The molecular docking simulation revealed that the binding of serine/threonine derivatives to the enzyme’s active site was stronger (−4.6~−7.9 kcal/mol) than that of the natural acceptor, glucose, and mainly occurred through interactions with aromatic residues. For N-tert-butoxycarbonyl serine methyl ester (6.8%) and N-carboxybenzyl serine benzyl ester (3.4%), their binding affinities and the distances between their hydroxyl side chain and the 1′-OH group of galactose moiety were in good accordance with the quantified bioconversion yields. Despite its lower predicted bioconversion yield, the high experimental bioconversion yield obtained with N-carboxybenzyl serine methyl ester (23.2%) demonstrated the importance of the thermodynamically-driven nature of the transgalactosylation reaction.
Collapse
|
248
|
Zhang Y, Liu B, Wu X, Li R, Ning X, Liu Y, Liu Z, Ge Z, Li R, Yin Y. New pyridin-3-ylmethyl carbamodithioic esters activate pyruvate kinase M2 and potential anticancer lead compounds. Bioorg Med Chem 2015; 23:4815-4823. [PMID: 26081759 DOI: 10.1016/j.bmc.2015.05.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 01/30/2023]
Abstract
Pyruvate kinase M2 (PKM2) is a key protein responsible for cancer's Warburg effect. Activation of PKM2 may alter aberrant metabolism in cancer cells, which suggests PKM2 as a tumor selective therapeutic target. In this paper, the lead compound 8 was first discovered as a new kind of PKM2 activator from a random screening of an in-house compound library. Then, a series of lead compound 8 analogs were designed, synthesized and evaluated for their activation of PKM2 and anticancer activities. 7-Azaindole analog 32 was identified as the most potent PKM2 activator. Compounds with potent enzyme activity also exhibited selective anti-proliferation activity on cancer cell lines HCT116, Hela and H1299 compared with non-tumor cell line BEAS-2B. The structure-activity relationships of these compounds were supported by molecular docking results. Preliminary pharmacological studies also showed that compound 32 arrests the cell cycle at the G2/M phase in HCT116 cell line.
Collapse
Affiliation(s)
- Yu Zhang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Bin Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Xingyu Wu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Ridong Li
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Xianling Ning
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Yu Liu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Zemei Ge
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Runtao Li
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| |
Collapse
|
249
|
Saravanan RR, Seshadri S, Gunasekaran S, Mendoza-Meroño R, Garcia-Granda S. Conformational analysis, X-ray crystallographic, FT-IR, FT-Raman, DFT, MEP and molecular docking studies on 1-(1-(3-methoxyphenyl) ethylidene) thiosemicarbazide. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2015; 139:321-328. [PMID: 25574651 DOI: 10.1016/j.saa.2014.12.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/22/2014] [Accepted: 12/10/2014] [Indexed: 06/04/2023]
Abstract
Conformational analysis, X-ray crystallographic, FT-IR, FT-Raman, DFT, MEP and molecular docking studies on 1-(1-(3-methoxyphenyl) ethylidene) thiosemicarbazide (MPET) are investigated. From conformational analysis the examination of the positions of a molecule taken and the energy changes is observed. The docking studies of the ligand MPET with target protein showed that this is a good molecule which docks well with target related to HMG-CoA. Hence MPET can be considered for developing into a potent anti-cholesterol drug. MEP assists in optimization of electrostatic interactions between the protein and the ligand. The MEP surface displays the molecular shape, size and electrostatic potential values. The optimized geometry of the compound was calculated from the DFT-B3LYP gradient calculations employing 6-31G (d, p) basis set and calculated vibrational frequencies are evaluated via comparison with experimental values.
Collapse
Affiliation(s)
- R R Saravanan
- Department of Physics, Misrimal Navajee Munoth Jain Engineering College, Thoraipakkam, Chennai 600 097, India.
| | - S Seshadri
- Department of Physics, L.N. Govt. Arts College, Ponneri, Thiruvallur 601 001, India
| | - S Gunasekaran
- Research & Development, St. Peter's University, Avadi, Chennai 600 054, India
| | - R Mendoza-Meroño
- Faculty of Chemistry, Department of Physical and Analytical Chemistry, University Oviedo, C/ Julian Claveria, 8, 33006 Oviedo, Asturias, Spain
| | - S Garcia-Granda
- Faculty of Chemistry, Department of Physical and Analytical Chemistry, University Oviedo, C/ Julian Claveria, 8, 33006 Oviedo, Asturias, Spain
| |
Collapse
|
250
|
Ikram NK, Durrant JD, Muchtaridi M, Zalaludin A, Purwitasari N, Mohamed N, Rahim ASA, Lam CK, Normi YM, Rahman NA, Amaro RE, Wahab HA. A virtual screening approach for identifying plants with anti H5N1 neuraminidase activity. J Chem Inf Model 2015; 55:308-16. [PMID: 25555059 PMCID: PMC4340357 DOI: 10.1021/ci500405g] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Indexed: 12/05/2022]
Abstract
Recent outbreaks of highly pathogenic and occasional drug-resistant influenza strains have highlighted the need to develop novel anti-influenza therapeutics. Here, we report computational and experimental efforts to identify influenza neuraminidase inhibitors from among the 3000 natural compounds in the Malaysian-Plants Natural-Product (NADI) database. These 3000 compounds were first docked into the neuraminidase active site. The five plants with the largest number of top predicted ligands were selected for experimental evaluation. Twelve specific compounds isolated from these five plants were shown to inhibit neuraminidase, including two compounds with IC50 values less than 92 μM. Furthermore, four of the 12 isolated compounds had also been identified in the top 100 compounds from the virtual screen. Together, these results suggest an effective new approach for identifying bioactive plant species that will further the identification of new pharmacologically active compounds from diverse natural-product resources.
Collapse
Affiliation(s)
- Nur Kusaira
Khairul Ikram
- Malaysian Institute of Pharmaceuticals
and Nutraceuticals, Ministry of Science, Technology and Innovation, Jalan Bukit Gambir, 11800, Penang, Malaysia
- Pharmaceutical Design and
Simulation Laboratory (PhDs), School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jacob D. Durrant
- Department of Chemistry & Biochemistry
and the National Biomedical Computation Resource, University of California—San Diego, La Jolla, California 92093-0340, United States
| | - Muchtaridi Muchtaridi
- Pharmaceutical Design and
Simulation Laboratory (PhDs), School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
- School of Pharmaceutical
Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Ayunni
Salihah Zalaludin
- Malaysian Institute of Pharmaceuticals
and Nutraceuticals, Ministry of Science, Technology and Innovation, Jalan Bukit Gambir, 11800, Penang, Malaysia
| | - Neny Purwitasari
- School of Pharmaceutical
Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Nornisah Mohamed
- School of Pharmaceutical
Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | | | - Chan Kit Lam
- School of Pharmaceutical
Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Yahaya M. Normi
- Department of Cell and Molecular Biology,
Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | | | - Rommie E. Amaro
- Department of Chemistry & Biochemistry
and the National Biomedical Computation Resource, University of California—San Diego, La Jolla, California 92093-0340, United States
| | - Habibah A Wahab
- Malaysian Institute of Pharmaceuticals
and Nutraceuticals, Ministry of Science, Technology and Innovation, Jalan Bukit Gambir, 11800, Penang, Malaysia
- Pharmaceutical Design and
Simulation Laboratory (PhDs), School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
- School of Pharmaceutical
Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| |
Collapse
|