Ligand-Based Virtual Screening for Discovery of Indole Derivatives as Potent DNA Gyrase ATPase Inhibitors Active against Mycobacterium tuberculosis and Hit Validation by Biological AssaysClick to copy article linkArticle link copied!
- Bongkochawan PakamwongBongkochawan PakamwongDepartment of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Bongkochawan Pakamwong
- Paptawan ThongdeePaptawan ThongdeeDepartment of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Paptawan Thongdee
- Bundit KamsriBundit KamsriDepartment of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Bundit Kamsri
- Naruedon PhusiNaruedon PhusiDepartment of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Naruedon Phusi
- Somjintana TaveepanichSomjintana TaveepanichDepartment of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Somjintana Taveepanich
- Kampanart ChayajarusKampanart ChayajarusDepartment of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Kampanart Chayajarus
- Pharit KamsriPharit KamsriDivision of Chemistry, Faculty of Science, Nakhon Phanom University, Nakhon Phanom 48000, ThailandMore by Pharit Kamsri
- Auradee PunkvangAuradee PunkvangDivision of Chemistry, Faculty of Science, Nakhon Phanom University, Nakhon Phanom 48000, ThailandMore by Auradee Punkvang
- Supa HannongbuaSupa HannongbuaDepartment of Chemistry, Faculty of Science, Kasetsart University, Bangkok 10900, ThailandMore by Supa Hannongbua
- Jidapa SangswanJidapa SangswanDepartment of Biological Science, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Jidapa Sangswan
- Khomson SuttisintongKhomson SuttisintongNational Nanotechnology Center, NSTDA, 111 Thailand Science Park, Klong Luang, Pathum Thani 12120, ThailandMore by Khomson Suttisintong
- Sanya Sureram
- Prasat KittakoopPrasat KittakoopChulabhorn Research Institute, Laksi, Bangkok 10210, ThailandProgram in Chemical Sciences, Chulabhorn Graduate Institute, Bangkok 10210, ThailandCenter of Excellence on Environmental Health and Toxicology (EHT), OPS, Ministry of Higher Education, Science, Research and Innovation, Bangkok 10210, ThailandMore by Prasat Kittakoop
- Poonpilas HongmaneePoonpilas HongmaneeDivision of Clinical Microbiology, Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, ThailandMore by Poonpilas Hongmanee
- Pitak SantanirandPitak SantanirandDivision of Clinical Microbiology, Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, ThailandMore by Pitak Santanirand
- Jiraporn LeanpolchareanchaiJiraporn LeanpolchareanchaiDepartment of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, ThailandMore by Jiraporn Leanpolchareanchai
- James SpencerJames SpencerSchool of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, U.K.More by James Spencer
- Adrian J. MulhollandAdrian J. MulhollandCentre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, U.K.More by Adrian J. Mulholland
- Pornpan Pungpo*Pornpan Pungpo*Email: [email protected]. Tel.: +664 535 3400 ext. 4124. Fax: +664 5288379.Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ubon Ratchathani University, Ubon Ratchathani 34190, ThailandMore by Pornpan Pungpo
Abstract
Mycobacterium tuberculosis is the single most important global infectious disease killer and a World Health Organization critical priority pathogen for development of new antimicrobials. M. tuberculosis DNA gyrase is a validated target for anti-TB agents, but those in current use target DNA breakage-reunion, rather than the ATPase activity of the GyrB subunit. Here, virtual screening, subsequently validated by whole-cell and enzyme inhibition assays, was applied to identify candidate compounds that inhibit M. tuberculosis GyrB ATPase activity from the Specs compound library. This approach yielded six compounds: four carbazole derivatives (1, 2, 3, and 8), the benzoindole derivative 11, and the indole derivative 14. Carbazole derivatives can be considered a new scaffold for M. tuberculosis DNA gyrase ATPase inhibitors. IC50 values of compounds 8, 11, and 14 (0.26, 0.56, and 0.08 μM, respectively) for inhibition of M. tuberculosis DNA gyrase ATPase activity are 5-fold, 2-fold, and 16-fold better than the known DNA gyrase ATPase inhibitor novobiocin. MIC values of these compounds against growth of M. tuberculosis H37Ra are 25.0, 3.1, and 6.2 μg/mL, respectively, superior to novobiocin (MIC > 100.0 μg/mL). Molecular dynamics simulations of models of docked GyrB:inhibitor complexes suggest that hydrogen bond interactions with GyrB Asp79 are crucial for high-affinity binding of compounds 8, 11, and 14 to M. tuberculosis GyrB for inhibition of ATPase activity. These data demonstrate that virtual screening can identify known and new scaffolds that inhibit both M. tuberculosis DNA gyrase ATPase activity in vitro and growth of M. tuberculosis bacteria.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
1. Introduction
2. Materials and Methods
2.1. Virtual Screening Approach
2.2. Antimycobacterial Assay
2.3. Overexpression and Purification of M. tuberculosis GyrA and GyrB
2.4. Inhibition (IC50) Assays of M. tuberculosis DNA Gyrase ATPase Activity
2.5. In Vitro Cytotoxicity Study
2.6. Molecular Docking Calculations
2.7. Molecular Dynamic Simulations
2.8. Binding Free Energy Calculation
2.9. Pairwise Energy Decomposition
3. Results and Discussion
3.1. Ligand-Based Virtual Screening
3.2. Biological Assays
3.3. Cytotoxicity of Lead Compounds
3.4. Interactions of Inhibitors with M. tuberculosis DNA Gyrase B
compound | ΔGGBSA (kcal/mol) | ΔGPBSA (kcal/mol) | ||||||
---|---|---|---|---|---|---|---|---|
replicate | averagea | replicate | averagea | |||||
1 | 2 | 3 | 1 | 2 | 3 | |||
8R | –57.5 | –58.9 | –61.1 | –59.1 ± 1.8 | –46.2 | –48.5 | –50.4 | –48.3 ± 2.1 |
8S | –48.4 | –49.3 | –48.6 | –48.8 ± 0.5 | –48.4 | –48.5 | –46.7 | –47.9 ± 1.0 |
11R | –64.8 | –60.7 | –61.4 | –62.3 ± 2.2 | –57.1 | –55.3 | –56.1 | –56.2 ± 0.9 |
11S | –55.3 | –50.1 | –50.5 | –52.0 ± 2.9 | –56.6 | –52.8 | –53.4 | –54.3 ± 2.0 |
14R | –72.0 | –73.0 | –71.8 | –72.3 ± 0.6 | –62.0 | –62.8 | –62.0 | –62.3 ± 0.5 |
14S | –62.3 | –62.7 | –62.6 | –62.5 ± 0.2 | –58.1 | –58.0 | –57.5 | –57.8 ± 0.3 |
Errors are estimated from the average and the difference between the results from three separate MD simulations for each compound. R and S represent R and S configurations.
3.5. Structure–Activity Relationship (SAR)
4. Conclusions
Data Availability
All chemical structures in SDF format of small molecules were downloaded from Specs database (https://www.specs.net/). PAINS and physicochemical properties of these molecules were calculated by SwissADME (http://www.swissadme.ch/index.php). (47) 2D chemical structures of small molecules in Figure 2 were generated using ChemDraw 20.1.1 (CambridgeSoft, http://www.cambridgesoft.com). IC50 values were calculated using GraphPad Prism8 with free trial license (GraphPad Inc., https://www.graphpad.com/) in Figure 3. Figures 4 and S4 were generated using pyMOL2.2.0 (https://pymol.org/2/).
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.4c00511.
Hit compound cytotoxicity against Caco-2 cells; cells were incubated with different concentrations of compounds (1, 2, 3, 8, 11, 14, G24, and G26) for 24 h and viability measured by MTT assay; all-atom RMSD plots for MD simulations of M. tuberculosis GyrB:inhibitor complexes; per-residue analysis of inhibitor:GyrB interactions; hydrogen bond interactions of known ATPase inhibitors with Asp79 in the M. smegmatis DNA gyrase ATP binding site; hits and leads obtained from three different virtual screening methods; previously characterized compounds identified from virtual similarity screening; details of fit parameters for IC50 determinations; hydrogen bonding of most active compounds (R-stereomers) to GyrB Asp79; and calculated binding free energies for binding of compounds 8, 11, and 14 to the wild type and mutant (D79A) GyrB subunits (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This research was supported by the Thailand Science Research and Innovation (TSRI), National Science, Research and Innovation Fund (NSRF), Center of Excellence for Innovation in Chemistry (PERCH-CIC) and Ubon Ratchathani University, Thailand. This work is partially supported by Thailand Science Research and Innovation (TSRI, Chulabhorn Research Institute (Grant No. 36827/4274406). The financial support from Royal Golden Jubilee Ph.D. Program, Thailand to B.P. (PHD/0155/2560) and B.K. (PHD/0132/2559) is gratefully acknowledged. The Thailand Graduate Institute of Science and Technology, Thailand (SCA-CO-2561-6946-TH and SCA-CO-2563-12135-TH) is acknowledged for financial support to P.T. and N.P., respectively. We thank EPSRC (CCP-BioSim, grant number EP/M022609/1) and BBSRC (International Institutional Award, BB/Y514123/1) for support. A.J.M. and J.S. thank the European Research Council under the European Horizon 2020 research and innovation programme (PREDACTED Advanced Grant Agreement no. 101021207). All MD simulations were carried out using the computational facilities of the Advanced Computing Research Centre, University of Bristol, United Kingdom http://www.bris.ac.uk/acrc/. Ubon Ratchathani University, NECTEC, Thailand, and the University of Bristol, United Kingdom are gratefully acknowledged for supporting this research.
TB | tuberculosis |
MDR-TB | multidrug-resistant TB |
M. tuberculosis | Mycobacterium tuberculosis |
FQ | fluoroquinolones |
GyrA | DNA gyrase A |
GyrB | DNA gyrase B |
MIC | minimal inhibitory concentration |
IC50 | half-maximal inhibitory concentration |
PAINS | pan-assay interference compounds |
MABA | microplate alamar blue assay |
DMSO | dimethyl sulfoxide |
PCR | polymerase chain reaction |
E. coli | Escherichia coli |
LB | Luria Broth |
IPTG | isopropyl-β-D-thiogalactopyranoside |
Ni-NTA | nickel-nitrilotriacetic acid |
SDS-PAGE | sodium dodecyl sulfate polyacrylamide gel electrophoresis |
MTT | 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide |
nm | nanometer |
MD | molecular dynamics simulations |
XP | extra precision |
GPU | graphics processing unit |
GAFF | general amber force field |
RESP | restrained electrostatic potential |
RMSD | root-mean square deviation |
MM/PBSA | molecular mechanics Poisson–Boltzmann surface area |
MM/GBSA | molecular mechanics generalized born surface area |
SASA | Solvent accessible surface area |
S. aureus | Staphylococcus aureus |
S. pneumoniae | Streptococcus pneumoniae |
SAR | structure–activity relationship |
References
This article references 89 other publications.
- 1WHO. Global tuberculosis report 2023. https://www.who.int/publications/i/item/9789240083851 (accessed January 2024).Google ScholarThere is no corresponding record for this reference.
- 2Aubry, A.; Fisher, L. M.; Jarlier, V.; Cambau, E. First functional characterization of a singly expressed bacterial type II topoisomerase: the enzyme from Mycobacterium tuberculosis. Biochem. Biophys. Res. Commun. 2006, 348, 158– 165, DOI: 10.1016/j.bbrc.2006.07.017Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xot1Wqtbg%253D&md5=763bef9f46308f23af208f537e82c1bdFirst functional characterization of a singly expressed bacterial type II topoisomerase: The enzyme from Mycobacterium tuberculosisAubry, Alexandra; Mark Fisher, L.; Jarlier, Vincent; Cambau, EmmanuelleBiochemical and Biophysical Research Communications (2006), 348 (1), 158-165CODEN: BBRCA9; ISSN:0006-291X. (Elsevier)Genome deciphering revealed that Mycobacterium tuberculosis encodes a single type II topoisomerase contrary to common bacteria harboring two type II topoisomerases (DNA gyrase and topoisomerase IV). Functions of the M. tuberculosis type II topoisomerase were explored after cloning and expressing the subunits encoding genes in Escherichia coli. M. tuberculosis type II topoisomerase supercoiled relaxed pBR322 with a specific activity close to that of DNA gyrases of common bacteria whereas it exhibited DNA relaxation and formation of cleavable complexes with activities significantly higher than other DNA gyrases. Intermol. passage activity evaluated by the decatenation of kinetoplast DNA was 25-fold lower than that of the topoisomerase IV from Streptococcus pneumoniae, but was markedly higher than that of the E. coli gyrase. Overall, the type II topoisomerase of M. tuberculosis exhibits classical polyvalent activities of DNA gyrase for supercoiling but enhanced relaxation, cleavage, and decatenation activities.
- 3Cole, S. T.; Brosch, R.; Parkhill, J.; Garnier, T.; Churcher, C.; Harris, D.; Gordon, S. V.; Eiglmeier, K.; Gas, S.; Barry, C. E., 3rd; Tekaia, F.; Badcock, K.; Basham, D.; Brown, D.; Chillingworth, T.; Connor, R.; Davies, R.; Devlin, K.; Feltwell, T.; Gentles, S.; Hamlin, N.; Holroyd, S.; Hornsby, T.; Jagels, K.; Krogh, A.; McLean, J.; Moule, S.; Murphy, L.; Oliver, K.; Osborne, J.; Quail, M. A.; Rajandream, M. A.; Rogers, J.; Rutter, S.; Seeger, K.; Skelton, J.; Squares, R.; Squares, S.; Sulston, J. E.; Taylor, K.; Whitehead, S.; Barrell, B. G. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature. 1998, 393, 537– 544, DOI: 10.1038/31159Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXjvV2ksrY%253D&md5=44d23e7674bfeb1a3e30b55abf5268d2Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequenceCole, S. T.; Brosch, R.; Parkhill, J.; Garnier, T.; Churcher, C.; Harris, D.; Gordon, S. V.; Eiglmeier, K.; Gas, S.; Barry, C. E., III; Tekaia, F.; Badcock, K.; Basham, D.; Brown, D.; Chillingworth, T.; Connor, R.; Davies, R.; Devlin, K.; Feltwell, T.; Gentles, S.; Hamlin, N.; Holroyd, S.; Hornsby, T.; Jagels, K.; Krogh, A.; McLean, J.; Moule, S.; Murphy, L.; Oliver, K.; Osborne, J.; Quail, M. A.; Rajandream, M.-A.; Rogers, J.; Rutter, S.; Seeger, K.; Skelton, J.; Squares, R.; Squares, S.; Sulston, J. E.; Taylor, K.; Whitehead, S.; Barrell, B. G.Nature (London) (1998), 393 (6685), 537-544CODEN: NATUAS; ISSN:0028-0836. (Macmillan Magazines)Countless millions of people have died from tuberculosis, a chronic infectious disease caused by the tubercle bacillus. The complete genome sequence of the best-characterized strain of Mycobacterium tuberculosis, H37Rv, was detd. and analyzed in order to improve our understanding of the biol. of this slow-growing pathogen and to help the conception of new prophylactic and therapeutic interventions. The genome comprises 4,411,529 base pairs, contains around 4000 genes, and has a very high G+C content that is reflected in the biased amino acid content of the proteins. M. tuberculosis differs radically from other bacteria in that a very large portion of its coding capacity is devoted to the prodn. of enzymes involved in lipogenesis and lipolysis, and to 2 new families of glycine-rich proteins with a repetitive structure that may represent a source of antigenic variation.
- 4Bush, N. G.; Evans-Roberts, K.; Maxwell, A. DNA Topoisomerases. EcoSal Plus 2015, 6, 1– 34, DOI: 10.1128/ecosalplus.esp-0010-2014Google ScholarThere is no corresponding record for this reference.
- 5Asif, M.; Siddiqui, A. A.; Husain, A. Quinolone derivatives as antitubercular drugs. Med. Chem. Res. 2013, 22, 1029– 1042, DOI: 10.1007/s00044-012-0101-3Google ScholarThere is no corresponding record for this reference.
- 6Facchinetti, V.; Gomes, C. R. B.; de Souza, M. V. N.; Vasconcelos, T. R. A. Perspectives on the development of novel potentially active quinolones against tuberculosis and cancer. Mini-Rev. Med. Chem. 2012, 12, 866– 874, DOI: 10.2174/138955712800959099Google ScholarThere is no corresponding record for this reference.
- 7Kathrotiya, H. G.; Patel, M. P. Synthesis and identification of β–aryloxyquinoline based diversely fluorine substituted N–aryl quinolone derivatives as a new class of antimicrobial, antituberculosis and antioxidant agents. Eur. J. Med. Chem. 2013, 63, 675– 684, DOI: 10.1016/j.ejmech.2013.03.017Google ScholarThere is no corresponding record for this reference.
- 8Kim, O. K.; Ohemeng, K.; Barrett, J. F. Advances in DNA gyrase inhibitors. Expert Opin. Investig. Drugs. 2001, 10, 199– 212, DOI: 10.1517/13543784.10.2.199Google ScholarThere is no corresponding record for this reference.
- 9Liu, K. L.; Teng, F.; Xiong, L.; Li, X.; Gao, C.; Yu, L. T. Discovery of quinolone derivatives as antimycobacterial agents. RSC Adv. 2021, 11, 24095– 24115, DOI: 10.1039/D0RA09250AGoogle ScholarThere is no corresponding record for this reference.
- 10Sriram, D.; Aubry, A.; Yogeeswari, P.; Fisher, L. M. Gatifloxacin derivatives: synthesis, antimycobacterial activities, and inhibition of Mycobacterium tuberculosis DNA gyrase. Bioorg. Med. Chem. Lett. 2006, 16, 2982– 2985, DOI: 10.1016/j.bmcl.2006.02.065Google ScholarThere is no corresponding record for this reference.
- 11Huang, T. S.; Kunin, C. M.; Lee, S. S. J.; Chen, Y. S.; Tu, H. Z.; Liu, Y. C. Trends in fluoroquinolone resistance of Mycobacterium tuberculosis complex in a Taiwanese medical centre: 1995–2003. J. Antimicrob. Chemother. 2005, 56, 1058– 1062, DOI: 10.1093/jac/dki353Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlagtLfE&md5=84f10b39609dd05e8a35c5e73b1ddec1Trends in fluoroquinolone resistance of Mycobacterium tuberculosis complex in a Taiwanese medical centre: 1995-2003Huang, Tsi-Shu; Kunin, Calvin M.; Lee, Susan Shin-Jung; Chen, Yao-Shen; Tu, Hui-Zin; Liu, Yung-ChingJournal of Antimicrobial Chemotherapy (2005), 56 (6), 1058-1062CODEN: JACHDX; ISSN:0305-7453. (Oxford University Press)Fluoroquinolones are being used more frequently for the treatment of multidrug-resistant (MDR) strains of Mycobacterium tuberculosis complex (MTB). This study was designed to det. the frequency of the emergence of fluoroquinolone-resistant strains in Taiwan and to assess whether this might be due to use of fluoroquinolones for treatment of patients with MDR or because of increased use of fluoroquinolones in the community for treatment of other infections. The authors also sought to det. whether there might be clonal spread of fluoroquinolone resistance. A total of 3497 clin. isolates of M. tuberculosis complex were obtained during 1995-2003, of which 141 were selected. They consisted of 62 isolates fully susceptible to four first-line drugs, 33 isolates resistant to rifampicin and isoniazid (MDR), and 46 isolates with a variety of any drug resistant patterns other than MDR (combination group). The MICs were detd. for ciprofloxacin, ofloxacin and levofloxacin. Results: An increase in the MIC90 and rates of resistance to ciprofloxacin, ofloxacin and levofloxacin were noted only in the MDR group. The rates were higher among strains isolated between 1998-2003 compared with those obtained between 1995-1997 (rate of resistance, 20% vs. 7.7%; MIC ≥ 4 mg/L vs. 1-2 mg/L). Among the 10 fluoroquinolone-resistant isolates, 5 (50%) possessed mutations other than S95T in the gyrA gene. No gyrB mutation was found in any of the clin. isolates. These findings suggest that fluoroquinolone resistance is the result of treatment of patients with MDR strains rather than from use in the general community in Taiwan. The emergence of fluoroquinolone resistance among MDR strains reinforces the need for routine fluoroquinolone susceptibility testing whenever these drugs might be used.
- 12Lee, A. S.; Tang, L. L.; Lim, I. H.; Wong, S. Y. Characterization of pyrazinamide and ofloxacin resistance among drug resistant Mycobacterium tuberculosis isolates from Singapore. Int. J. Infect. Dis. 2002, 6, 48– 51, DOI: 10.1016/s1201-9712(02)90136-0Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD38zntFWltQ%253D%253D&md5=783085804dfb8525f2d9a17d41c27f55Characterization of pyrazinamide and ofloxacin resistance among drug resistant Mycobacterium tuberculosis isolates from SingaporeLee Ann S G; Tang Lynn L H; Lim Irene H K; Wong Sin YewInternational journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases (2002), 6 (1), 48-51 ISSN:1201-9712.OBJECTIVES: To evaluate rapid molecular approaches for the detection of pyrazinamide (PZA) and ofloxacin resistance, by screening 100 known drug-resistant Mycobacterium tuberculosis isolates. METHODS: Mycobacterium tuberculosis isolates were tested for phenotypic resistance to pyrazinamide and ofloxacin using the BACTEC 460 radiometric method and the E-test, respectively. Mutation screening was done by amplifying the pncA, gyrA, and gyrB genes by the polymerase chain reaction (PCR) and direct automated sequencing. RESULTS: Twelve isolates were PZA-resistant and 8 of 12 (66.7%) isolates had missense mutations or deletions at the pncA gene, suggesting that mutation or deletion at the pncA gene is the major molecular mechanism of PZA resistance among the Singaporean isolates. Using the E-test, 48 isolates were resistant to ofloxacin, with minimum inhibitory concentrations of 4 microg/mL or higher. No mutations were observed at the quinolone resistance-determining region (QRDR) of gyrA in all isolates. At the QRDR of gyrB, mutations were present in 1 of 48 ofloxacin-resistant isolates and 0 of 19 ofloxacin-susceptible isolates. CONCLUSIONS: In Singapore, genotypic analysis of resistance to PZA and ofloxacin is inadequate and should be complemented by conventional methods.
- 13Maruri, F.; Sterling, T. R.; Kaiga, A. W.; Blackman, A.; van der Heijden, Y. F.; Mayer, C.; Cambau, E.; Aubry, A. A systematic review of gyrase mutations associated with fluoroquinolone–resistant Mycobacterium tuberculosis and a proposed gyrase numbering system. J. Antimicrob. Chemother. 2012, 67, 819– 831, DOI: 10.1093/jac/dkr566Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xkt1ektbc%253D&md5=dcba60297069b8863c4e390e1098dffdA systematic review of gyrase mutations associated with fluoroquinolone-resistant Mycobacterium tuberculosis and a proposed gyrase numbering systemMaruri, Fernanda; Sterling, Timothy R.; Kaiga, Anne W.; Blackman, Amondrea; van der Heijden, Yuri F.; Mayer, Claudine; Cambau, Emmanuelle; Aubry, AlexandraJournal of Antimicrobial Chemotherapy (2012), 67 (4), 819-831CODEN: JACHDX; ISSN:0305-7453. (Oxford University Press)Fluoroquinolone resistance in Mycobacterium tuberculosis has become increasingly important. A review of mutations in DNA gyrase, the fluoroquinolone target, is needed to improve the mol. detection of resistance. The authors performed a systematic review of studies reporting mutations in DNA gyrase genes in clin. M. tuberculosis isolates. From 42 studies that met inclusion criteria, 1220 fluoroquinolone-resistant M. tuberculosis isolates underwent sequencing of the quinolone resistance-detg. region (QRDR) of gyrA; 780 (64%) had mutations. The QRDR of gyrB was sequenced in 534 resistant isolates; 17 (3%) had mutations. Mutations at gyrA codons 90, 91 or 94 were present in 654/1220 (54%) resistant isolates. Four different GyrB numbering systems were reported, resulting in mutation location discrepancies. The authors propose a consensus numbering system. Most fluoroquinolone-resistant M. tuberculosis isolates had mutations in DNA gyrase, but a substantial proportion did not. The proposed consensus numbering system can improve mol. detection of resistance and identification of novel mutations.
- 14Pitaksajjakul, P.; Wongwit, W.; Punprasit, W.; Eampokalap, B.; Peacock, S.; Ramasoota, P. Mutations in the GyrA and GyrB genes of fluoroquinolone–resistant Mycobacterium tuberculosis from TB patients in Thailand. Southeast Asian J. Trop. Med. Public Health 2005, 36, 228– 237Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFyht78%253D&md5=db80a73a8e5ab2e6f9bb89813c2fac6dMutations in the gyrA and gyrB genes of fluoroquinolone-resistant Mycobacterium tuberculosis from TB patients in ThailandPitaksajjakul, Pannamthip; Wongwit, Waranya; Punprasit, Wantanee; Eampokalap, Boonchuy; Peacock, Sharon; Ramasoota, PongramaSoutheast Asian Journal of Tropical Medicine and Public Health (2005), 36 (Suppl. 4), 228-237CODEN: SJTMAK; ISSN:0125-1562. (SEAMEO-TROPMED Network)Among fluoroquinolone-resistant Mycobacterium tuberculosis (FQr-MTB) isolates, mutation at positions 90, 91, and 94 in gyrA gene and at positions 495, 516, and 533 in gyrB gene have been frequently reported. In this study, 35 isolates of FQr-MTB were collected from Siriraj Hospital and Chest Disease Institute. The quinolone-resistance-detg. regions (QRDR) of gyrA and gyrB genes in all 35 FQr-MTB isolates and from the H37Ra MTB strain were amplified using polymerase chain reaction (PCR). DNA-sequencing and single-strand conformation polymorphism (SSCP) were further utilized for characterization of the mutations in the QRDR of gyrA and gyrB genes and mutation screening, resp. From DNA-sequencing, 21 of 35 (60%) exhibited single-point mutations in different positions, at Ala90Val, Ser91Pro, and Asp94(Gly/Ala/His/Asn); and one novel mutation position at Gly88Cys in the gyrA gene and Asp495Asn in the gyrB gene. These positions were previously frequently reported to be responsible for FQr-MTB. The other 14 FQr-MTB isolates (40%) had no mutation. This study is the first report of mutation occurring only in the QRDR of the gyrB gene, without prior mutation in the gyrA QRDR among FQr-MTB isolates. By SSCP anal. for screening of the mutant FQr-MTB, the SSCP patterns of mutated FQr-MTB isolates were clearly differentiated from the SSCP patterns of FQs-MTB.
- 15Pitaksajjakul, P.; Worakhunpiset, S.; Chaiprasert, A.; Boonyasopun, J.; Ramasoota, P. GyrA and GyrB mutations in ofloxacin–resistant Mycobacterium tuberculosis clinical isolates in Thailand. Southeast Asian J. Trop. Med. Public Health. 2011, 42, 1163– 1167Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlansbfI&md5=c0a8d3f3b0538afe15fcf095a91c32c2gyrA and gyrB mutations in ofloxacin-resistant Mycobacterium tuberculosis clinical isolates in ThailandPitaksajjakul, Pannamthip; Worakhunpiset, Suwalee; Chaiprasert, Angkana; Boonyasopun, Jirakarn; Ramasoota, PongramaSoutheast Asian Journal of Tropical Medicine and Public Health (2011), 42 (5), 1163-1167CODEN: SJTMAK; ISSN:0125-1562. (SEAMEO-TROPMED Network)In order to identify mutations in gyrA and gyrB genes in 92 ofloxacin-resistant Mycobacterium tuberculosis (OFXr-MTB) clin. isolates collected from Siriraj Hospital, Mahidol University and Chest Disease Institute, Thailand. The quinolone resistance-detg. regions (QRDR) of gyrA and gyrB in all 92 OFXr-MTB isolates were amplified using polymerase chain reaction and sequenced. There were 70 isolates with point mutations assocd. with ofloxacin resistance. In gyrA QRDR, 69 isolates had mutations in gyrA Gly88 (Ala/(75), Ala90 (Val), Ser91 (Pro) and Asp94 (Gly/Ala/His/Asn), the latter being the most common (42%). Only one isolate was found with mutation at position Asp495 (Asn). The other 22 isolates had no mutations in both gyrA and gyrB QRDR. Thus, point mutations in gyrA and gyrB QRDR were responsible for OFXr-MTB clin. isolates in Thailand.
- 16Soudani, A.; Hadjfredj, S.; Zribi, M.; Messaoud, T.; Masmoudi, A.; Majed, B.; Fendri, C. First report of molecular characterization of fluoroquinolone–resistant Mycobacterium tuberculosis isolates from a Tunisian hospital. Clin. Microbiol. Infect. 2010, 16, 1454– 1457, DOI: 10.1111/j.1469-0691.2010.03087.xGoogle ScholarThere is no corresponding record for this reference.
- 17Medapi, B.; Renuka, J.; Saxena, S.; Sridevi, J. P.; Medishetti, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Design and synthesis of novel quinoline–aminopiperidine hybrid analogues as Mycobacterium tuberculosis DNA gyrase B inhibitors. Bioorg. Med. Chem. 2015, 23, 2062– 2078, DOI: 10.1016/j.bmc.2015.03.004Google ScholarThere is no corresponding record for this reference.
- 18Medapi, B.; Suryadevara, P.; Renuka, J.; Sridevi, J. P.; Yogeeswari, P.; Sriram, D. 4–Aminoquinoline derivatives as novel Mycobacterium tuberculosis GyrB inhibitors: Structural optimization, synthesis and biological evaluation. Eur. J. Med. Chem. 2015, 103, 1– 16, DOI: 10.1016/j.ejmech.2015.06.032Google ScholarThere is no corresponding record for this reference.
- 19Jeankumar, V. U.; Reshma, R. S.; Vats, R.; Janupally, R.; Saxena, S.; Yogeeswari, P.; Sriram, D. Engineering another class of anti–tubercular lead: Hit to lead optimization of an intriguing class of gyrase ATPase inhibitors. Eur. J. Med. Chem. 2016, 122, 216– 231, DOI: 10.1016/j.ejmech.2016.06.042Google ScholarThere is no corresponding record for this reference.
- 20Jeankumar, V. U.; Renuka, J.; Pulla, V. K.; Soni, V.; Sridevi, J. P.; Suryadevara, P.; Shravan, M.; Medishetti, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Development of novel N–linked aminopiperidine–based mycobacterial DNA gyrase B inhibitors: scaffold hopping from known antibacterial leads. Int. J. Antimicrob Agents. 2014, 43, 269– 278, DOI: 10.1016/j.ijantimicag.2013.12.006Google ScholarThere is no corresponding record for this reference.
- 21Jeankumar, V. U.; Kotagiri, S.; Janupally, R.; Suryadevara, P.; Sridevi, J. P.; Medishetti, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Exploring the gyrase ATPase domain for tailoring newer anti–tubercular drugs: hit to lead optimization of a novel class of thiazole inhibitors. Bioorg. Med. Chem. 2015, 23, 588– 601, DOI: 10.1016/j.bmc.2014.12.001Google ScholarThere is no corresponding record for this reference.
- 22Omar, M. A.; Masaret, G. S.; Abbas, E. M. H.; Abdel Aziz, M. M.; Harras, M. F.; Farghaly, T. A. Novel anti–tubercular and antibacterial based benzosuberone–thiazole moieties: synthesis, molecular docking analysis, DNA gyrase supercoiling and ATPase activity. Bioorg Chem. 2020, 104, 104316 DOI: 10.1016/j.bioorg.2020.104316Google ScholarThere is no corresponding record for this reference.
- 23Jeankumar, V. U.; Renuka, J.; Kotagiri, S.; Saxena, S.; Kakan, S. S.; Sridevi, J. P.; Yellanki, S.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Gyrase ATPase domain as an antitubercular drug discovery platform: structure–based design and lead optimization of nitrothiazolyl carboxamide analogues. Chem. Med. Chem. 2014, 9, 1850– 1859, DOI: 10.1002/cmdc.201402035Google ScholarThere is no corresponding record for this reference.
- 24P, S. H.; Solapure, S.; Mukherjee, K.; Nandi, V.; Waterson, D.; Shandil, R.; Balganesh, M.; Sambandamurthy, V. K.; Raichurkar, A. K.; Deshpande, A.; Ghosh, A.; Awasthy, D.; Shanbhag, G.; Sheikh, G.; McMiken, H.; Puttur, J.; Reddy, J.; Werngren, J.; Read, J.; Kumar, M.; M, R.; Chinnapattu, M.; Madhavapeddi, P.; Manjrekar, P.; Basu, R.; Gaonkar, S.; Sharma, S.; Hoffner, S.; Humnabadkar, V.; Subbulakshmi, V.; Panduga, V. Optimization of pyrrolamides as mycobacterial GyrB ATPase inhibitors: structure–activity relationship and in vivo efficacy in a mouse model of tuberculosis. Antimicrob. Agents Chemother. 2014, 58, 61– 70, DOI: 10.1128/AAC.01751-13Google ScholarThere is no corresponding record for this reference.
- 25Reddy, K. I.; Srihari, K.; Renuka, J.; Sree, K. S.; Chuppala, A.; Jeankumar, V. U.; Sridevi, J. P.; Babu, K. S.; Yogeeswari, P.; Sriram, D. An efficient synthesis and biological screening of benzofuran and benzo[d]isothiazole derivatives for Mycobacterium tuberculosis DNA GyrB inhibition. Bioorg. Med. Chem. 2014, 22, 6552– 6563, DOI: 10.1016/j.bmc.2014.10.016Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVSls7fO&md5=35443a5527173e382e720eec298aa5d6An efficient synthesis and biological screening of benzofuran and benzo[d]isothiazole derivatives for Mycobacterium tuberculosis DNA GyrB inhibitionReddy, Kummetha Indrasena; Srihari, Konduri; Renuka, Janupally; Sree, Komanduri Shruthi; Chuppala, Aruna; Jeankumar, Variam Ullas; Sridevi, Jonnalagadda Padma; Babu, Kondra Sudhakar; Yogeeswari, Perumal; Sriram, DharmarajanBioorganic & Medicinal Chemistry (2014), 22 (23), 6552-6563CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A series of twenty eight mols. of Et 5-(piperazin-1-yl)benzofuran-2-carboxylate and 3-(piperazin-1-yl)benzo[d]isothiazole were designed by mol. hybridization of thiazole aminopiperidine core and carbamide side chain in eight steps and were screened for their in vitro Mycobacterium smegmatis (MS) GyrB ATPase assay, Mycobacterium tuberculosis (MTB) DNA gyrase super coiling assay, antitubercular activity, cytotoxicity and protein-inhibitor interaction assay through differential scanning fluorometry. Also the orientation and the ligand-protein interactions of the top hit mols. with MS DNA gyrase B subunit active site were investigated applying extra precision mode (XP) of Glide. Among the compds. studied, 4-(benzo[d]isothiazol-3-yl)-N-(4-chlorophenyl)piperazine-1-carboxamide (26) was found to be the most promising inhibitor with an MS GyrB IC50 of 1.77 ± 0.23 μM, 0.42 ± 0.23 against MTB DNA gyrase, MTB MIC of 3.64 μM, and was not cytotoxic in eukaryotic cells at 100 μM. Moreover the interaction of protein-ligand complex was stable and showed a pos. shift of 3.5 °C in differential scanning fluorimetric evaluations.
- 26Renuka, J.; Reddy, K. I.; Srihari, K.; Jeankumar, V. U.; Shravan, M.; Sridevi, J. P.; Yogeeswari, P.; Babu, K. S.; Sriram, D. Design, synthesis, biological evaluation of substituted benzofurans as DNA gyrase B inhibitors of Mycobacterium tuberculosis. Bioorg. Med. Chem. 2014, 22, 4924– 4934, DOI: 10.1016/j.bmc.2014.06.041Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlGmu7bM&md5=4e84e0c1d803e5579a10314a199872d4Design, synthesis, biological evaluation of substituted benzofurans as DNA gyrase B inhibitors of Mycobacterium tuberculosisRenuka, Janupally; Reddy, Kummetha Indrasena; Srihari, Konduri; Jeankumar, Variam Ullas; Shravan, Morla; Sridevi, Jonnalagadda Padma; Yogeeswari, Perumal; Babu, Kondra Sudhakar; Sriram, DharmarajanBioorganic & Medicinal Chemistry (2014), 22 (17), 4924-4934CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)DNA gyrase of Mycobacterium tuberculosis (MTB) is a type II topoisomerase and is a well established and validated target for the development of novel therapeutics. The authors present the discovery and optimization of an Et 5-(piperazin-1-yl) benzofuran-2-carboxylate series of mycobacterial DNA gyrase B inhibitors, selected from the Birla Institute of Technol. and Science (BITS) database chem. library of about 3000 mols. When these compds. were tested for their biol. activity, the compd. 22 emerged as the most active potent lead with an IC50 of 3.2 ± 0.15 μM against Mycobacterium smegmatis DNA gyrase B enzyme and 0.81 ± 0.24 μM in MTB supercoiling activity. The binding of this most active compd. to the DNA gyrase B enzyme and its thermal stability was further characterized using a differential scanning fluorimetry method.
- 27Jeankumar, V. U.; Saxena, S.; Vats, R.; Reshma, R. S.; Janupally, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Structure–guided discovery of antitubercular agents that target the gyrase ATPase domain. Chem. Med. Chem. 2016, 11, 539– 548, DOI: 10.1002/cmdc.201500556Google ScholarThere is no corresponding record for this reference.
- 28Saxena, S.; Samala, G.; Renuka, J.; Sridevi, J. P.; Yogeeswari, P.; Sriram, D. Development of 2–amino–5–phenylthiophene–3–carboxamide derivatives as novel inhibitors of Mycobacterium tuberculosis DNA GyrB domain. Bioorg. Med. Chem. 2015, 23, 1402– 1412, DOI: 10.1016/j.bmc.2015.02.032Google ScholarThere is no corresponding record for this reference.
- 29Saxena, S.; Renuka, J.; Yogeeswari, P.; Sriram, D. Discovery of novel mycobacterial DNA gyrase B inhibitors: In silico and in vitro biological evaluation. Mol. Inform. 2014, 33, 597– 609, DOI: 10.1002/minf.201400058Google ScholarThere is no corresponding record for this reference.
- 30McGarry, D. H.; Cooper, I. R.; Walker, R.; Warrilow, C. E.; Pichowicz, M.; Ratcliffe, A. J.; Salisbury, A. M.; Savage, V. J.; Moyo, E.; Maclean, J.; Smith, A.; Charrier, C.; Stokes, N. R.; Lindsay, D. M.; Kerr, W. J. Design, synthesis and antibacterial properties of pyrimido[4,5–b]indol–8–amine inhibitors of DNA gyrase. Bioorg. Med. Chem. Lett. 2018, 28, 2998– 3003, DOI: 10.1016/j.bmcl.2018.05.049Google ScholarThere is no corresponding record for this reference.
- 31Henderson, S. R.; Stevenson, C. E. M.; Malone, B.; Zholnerovych, Y.; Mitchenall, L. A.; Pichowicz, M.; McGarry, D. H.; Cooper, I. R.; Charrier, C.; Salisbury, A. M.; Lawson, D. M.; Maxwell, A. Structural and mechanistic analysis of ATPase inhibitors targeting mycobacterial DNA gyrase. J. Antimicrob. Chemother. 2020, 75, 2835– 2842, DOI: 10.1093/jac/dkaa286Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitl2jtrs%253D&md5=78dddb492907ec5f0215e19f55655990Structural and mechanistic analysis of ATPase inhibitors targeting mycobacterial DNA gyraseHenderson, Sara R.; Stevenson, Clare E. M.; Malone, Brandon; Zholnerovych, Yelyzaveta; Mitchenall, Lesley A.; Pichowicz, Mark; McGarry, David H.; Cooper, Ian R.; Charrier, Cedric; Salisbury, Anne-Marie; Lawson, David M.; Maxwell, AnthonyJournal of Antimicrobial Chemotherapy (2020), 75 (10), 2835-2842CODEN: JACHDX; ISSN:1460-2091. (Oxford University Press)To evaluate the efficacy of two novel compds. against mycobacteria and det. the mol. basis of their action on DNA gyrase using structural and mechanistic approaches. Redx03863 and Redx04739 were tested in antibacterial assays, and also against their target, DNA gyrase, using DNA supercoiling and ATPase assays. X-ray crystallog. was used to det. the structure of the gyrase B protein ATPase sub-domain from Mycobacterium smegmatis complexed with the aminocoumarin drug novobiocin, and structures of the same domain from Mycobacterium thermoresistibile complexed with novobiocin, and also with Redx03863. Both compds., Redx03863 and Redx04739, were active against selected Gram-pos. and Gram-neg. species, with Redx03863 being the more potent, and Redx04739 showing selectivity against M. smegmatis. Both compds. were potent inhibitors of the supercoiling and ATPase reactions of DNA gyrase, but did not appreciably affect the ATP-independent relaxation reaction. The structure of Redx03863 bound to the gyrase B protein ATPase sub-domain from M. thermoresistibile shows that it binds at a site adjacent to the ATP- and novobiocin-binding sites. We found that most of the mutations that we made in the Redx03863-binding pocket, based on the structure, rendered gyrase inactive. Redx03863 and Redx04739 inhibit gyrase by preventing the binding of ATP. The fact that the Redx03863-binding pocket is distinct from that of novobiocin, coupled with the lack of activity of resistant mutants, suggests that such compds. could have potential to be further exploited as antibiotics.
- 32Kamsri, B.; Pakamwong, B.; Thongdee, P.; Phusi, N.; Kamsri, P.; Punkvang, A.; Ketrat, S.; Saparpakorn, P.; Hannongbua, S.; Sangswan, J.; Suttisintong, K.; Sureram, S.; Kittakoop, P.; Hongmanee, P.; Santanirand, P.; Leanpolchareanchai, J.; Goudar, K. E.; Spencer, J.; Mulholland, A. J.; Pungpo, P. Bioisosteric Design Identifies Inhibitors of Mycobacterium tuberculosis DNA Gyrase ATPase Activity. J. Chem. Inf Model. 2023, 63, 2707– 2718, DOI: 10.1021/acs.jcim.2c01376Google ScholarThere is no corresponding record for this reference.
- 33Pakamwong, B.; Thongdee, P.; Kamsri, B.; Phusi, N.; Kamsri, P.; Punkvang, A.; Ketrat, S.; Saparpakorn, P.; Hannongbua, S.; Ariyachaokun, K.; Suttisintong, K.; Sureram, S.; Kittakoop, P.; Hongmanee, P.; Santanirand, P.; Spencer, J.; Mulholland, A. J.; Pungpo, P. Identification of Potent DNA Gyrase Inhibitors Active against Mycobacterium tuberculosis. J. Chem. Inf Model. 2022, 62, 1680– 1690, DOI: 10.1021/acs.jcim.1c01390Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XosFeqsbc%253D&md5=60557f6d45a2d8df0a071a3d37afe03fIdentification of Potent DNA Gyrase Inhibitors Active against Mycobacterium tuberculosisPakamwong, Bongkochawan; Thongdee, Paptawan; Kamsri, Bundit; Phusi, Naruedon; Kamsri, Pharit; Punkvang, Auradee; Ketrat, Sombat; Saparpakorn, Patchreenart; Hannongbua, Supa; Ariyachaokun, Kanchiyaphat; Suttisintong, Khomson; Sureram, Sanya; Kittakoop, Prasat; Hongmanee, Poonpilas; Santanirand, Pitak; Spencer, James; Mulholland, Adrian J.; Pungpo, PornpanJournal of Chemical Information and Modeling (2022), 62 (7), 1680-1690CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Mycobacterium tuberculosis DNA gyrase manipulates the DNA topol. using controlled breakage and religation of DNA driven by ATP hydrolysis. DNA gyrase has been validated as the enzyme target of fluoroquinolones (FQs), second-line antibiotics used for the treatment of multidrug-resistant tuberculosis. Mutations around the DNA gyrase DNA-binding site result in the emergence of FQ resistance in M. tuberculosis; inhibition of DNA gyrase ATPase activity is one strategy to overcome this. Here, virtual screening, subsequently validated by biol. assays, was applied to select candidate inhibitors of the M. tuberculosis DNA gyrase ATPase activity from the Specs compd. library (www.specs.net). Thirty compds. were identified and selected as hits for in vitro biol. assays, of which two compds., G24 and G26, inhibited the growth of M. tuberculosis H37Rv with a minimal inhibitory concn. of 12.5 μg/mL. The two compds. inhibited DNA gyrase ATPase activity with IC50 values of 2.69 and 2.46 μM, resp., suggesting this to be the likely basis of their antitubercular activity. Models of complexes of compds. I and II bound to the M. tuberculosis DNA gyrase ATP-binding site, generated by mol. dynamics simulations followed by pharmacophore mapping anal., showed hydrophobic interactions of inhibitor hydrophobic headgroups and electrostatic and hydrogen bond interactions of the polar tails, which are likely to be important for their inhibition. Decreasing compd. lipophilicity by increasing the polarity of these tails then presents a likely route to improving the soly. and activity. Thus, compds. I and II provide attractive starting templates for the optimization of antitubercular agents that act by targeting DNA gyrase.
- 34Maxwell, A. The interaction between coumarin drugs and DNA gyrase. Mol. Microbiol. 1993, 9, 681– 686, DOI: 10.1111/j.1365-2958.1993.tb01728.xGoogle Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXmsVCjsr8%253D&md5=936de9ec8f1f199de584ec2cedb941b6The interaction between coumarin drugs and DNA gyraseMaxwell, AnthonyMolecular Microbiology (1993), 9 (4), 681-6CODEN: MOMIEE; ISSN:0950-382X.A review with 49 refs. The coumarin group of antibiotics have as their target bacterial DNA gyrase (I). The drugs bind to the B subunit of I and inhibit DNA supercoiling by blocking the ATPase activity. Recent data show that the binding site for the drugs lies within the N-terminal part of the B protein, and individual amino acids involved in coumarin interaction are being identified. The mode of inhibition of the I ATPase reaction by coumarins is unlikely to be simple competitive inhibition, and the drugs may act by stabilizing a conformation of the enzyme with low affinity for ATP.
- 35Grossman, S.; Fishwick, C. W. G.; McPhillie, M. J. Developments in non–intercalating bacterial topoisomerase inhibitors: allosteric and ATPase inhibitors of DNA gyrase and topoisomerase IV. Pharm. 2023, 16, 261, DOI: 10.3390/ph16020261Google ScholarThere is no corresponding record for this reference.
- 36Kar, S.; Roy, K. How far can virtual screening take us in drug discovery?. Expert Opin Drug Discovery 2013, 8, 245– 261, DOI: 10.1517/17460441.2013.761204Google ScholarThere is no corresponding record for this reference.
- 37Lionta, E.; Spyrou, G.; Vassilatis, D. K.; Cournia, Z. Structure–based virtual screening for drug discovery: principles, applications and recent advances. Curr. Top Med. Chem. 2014, 14, 1923– 1938, DOI: 10.2174/1568026614666140929124445Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVGgt7fJ&md5=0550a396dd011adf52699b091548345bStructure-Based Virtual Screening for Drug Discovery: Principles, Applications and Recent AdvancesLionta, Evanthia; Spyrou, George; Vassilatis, Demetrios K.; Cournia, ZoeCurrent Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2014), 14 (16), 1923-1938CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)A review. Structure-based drug discovery (SBDD) is becoming an essential tool in assisting fast and cost-efficient lead discovery and optimization. The application of rational, structure-based drug design is proven to be more efficient than the traditional way of drug discovery since it aims to understand the mol. basis of a disease and utilizes the knowledge of the three-dimensional structure of the biol. target in the process. In this review, we focus on the principles and applications of Virtual Screening (VS) within the context of SBDD and examine different procedures ranging from the initial stages of the process that include receptor and library pre-processing, to docking, scoring and post-processing of top scoring hits. Recent improvements in structure-based virtual screening (SBVS) efficiency through ensemble docking, induced fit and consensus docking are also discussed. The review highlights advances in the field within the framework of several success studies that have led to nM inhibition directly from VS and provides recent trends in library design as well as discusses limitations of the method. Applications of SBVS in the design of substrates for engineered proteins that enable the discovery of new metabolic and signal transduction pathways and the design of inhibitors of multifunctional proteins are also reviewed. Finally, we contribute two promising VS protocols recently developed by us that aim to increase inhibitor selectivity. In the first protocol, we describe the discovery of micromolar inhibitors through SBVS designed to inhibit the mutant H1047R PI3Kα kinase. Second, we discuss a strategy for the identification of selective binders for the RXRα nuclear receptor. In this protocol, a set of target structures is constructed for ensemble docking based on binding site shape characterization and clustering, aiming to enhance the hit rate of selective inhibitors for the desired protein target through the SBVS process.
- 38Oliveira, T. A. D.; Silva, M. P. D.; Maia, E. H. B.; Silva, A. M. D.; Taranto, A. G. Virtual screening algorithms in drug discovery: a review focused on machine and deep learning methods. Drugs Drug Candidates. 2023, 2, 311– 334, DOI: 10.3390/ddc2020017Google ScholarThere is no corresponding record for this reference.
- 39Thongdee, P.; Hanwarinroj, C.; Pakamwong, B.; Kamsri, P.; Punkvang, A.; Leanpolchareanchai, J.; Ketrat, S.; Saparpakorn, P.; Hannongbua, S.; Ariyachaokun, K.; Suttisintong, K.; Sureram, S.; Kittakoop, P.; Hongmanee, P.; Santanirand, P.; Mukamolova, G. V.; Blood, R. A.; Takebayashi, Y.; Spencer, J.; Mulholland, A. J.; Pungpo, P. Virtual screening identifies novel and potent inhibitors of Mycobacterium tuberculosis PknB with antibacterial activity. J. Chem. Inf Model. 2022, 62, 6508– 6518, DOI: 10.1021/acs.jcim.2c00531Google ScholarThere is no corresponding record for this reference.
- 40Arica Sosa, A.; Alcántara, R.; Jiménez Avalos, G.; Zimic, M.; Milón, P.; Quiliano, M. Identifying RO9021 as a potential inhibitor of PknG from Mycobacterium tuberculosis: combinative computational and in vitro studies. ACS Omega. 2022, 7, 20204– 20218, DOI: 10.1021/acsomega.2c02093Google ScholarThere is no corresponding record for this reference.
- 4141. Sheikh, B. A.; Bhat, B. A.; Rizvi, M. A.; Ahmad, Z.; Almilaibary, A.; Alkhanani, M.; Mir, M. A. Computational studies to identify potential inhibitors targeting the DprE1 protein in Mycobacterium tuberculosis. Int. J. Pharm. Investig. 2023, 13, 129– 138, DOI: 10.5530/223097131750Google ScholarThere is no corresponding record for this reference.
- 42Verma, A.; Kumar, V.; Naik, B.; Khan, J. M.; Singh, P.; Saris, P. E. J.; Gupta, S. Screening and molecular dynamics simulation of compounds inhibiting MurB enzyme of drug–resistant Mycobacterium tuberculosis: An in–silico approach. Saudi J. Biol. Sci. 2023, 30, 103730 DOI: 10.1016/j.sjbs.2023.103730Google ScholarThere is no corresponding record for this reference.
- 43Kumar, G. S.; Dubey, A.; Panda, S. P.; Alawi, M. M.; Sindi, A. A.; Azhar, E. I.; Dwivedi, V. D.; Agrawal, S. Repurposing of antibacterial compounds for suppression of Mycobacterium tuberculosis dormancy reactivation by targeting resuscitation–promoting factors B. J. Biomol. Struct. Dyn. 2023, 1– 13, DOI: 10.1080/07391102.2023.2245059Google ScholarThere is no corresponding record for this reference.
- 44Johannsen, S.; Gierse, R. M.; Olshanova, A.; Smerznak, E.; Laggner, C.; Eschweiler, L.; Adeli, Z.; Hamid, R.; Alhayek, A.; Reiling, N.; Haupenthal, J.; Hirsch, A. K. H. Not every hit–identification technique works on 1–deoxy–d–xylulose 5–phosphate synthase (DXPS): making the most of a virtual screening campaign. ChemMedChem 2023, 18, e202200590 DOI: 10.1002/cmdc.202200590Google ScholarThere is no corresponding record for this reference.
- 45Damera, T.; Kondaparthi, V.; Bingi, M.; Mustyala, K. K.; Malkhed, V. Identification of novel scaffolds to inhibit Mycobacterium tuberculosis PimA protein–A computational approach. J. Cell Biochem. 2023, 124, 836– 848, DOI: 10.1002/jcb.30412Google ScholarThere is no corresponding record for this reference.
- 46Varon, H. A.; Santos, P.; Lopez-Vallejo, F.; Soto, C. Y. Novel scaffolds targeting Mycobacterium tuberculosis plasma membrane Ca2+ transporter CtpF by structure–based strategy. Bioorg. Chem. 2023, 138, 106648 DOI: 10.1016/j.bioorg.2023.106648Google ScholarThere is no corresponding record for this reference.
- 47Daina, A.; Michielin, O.; Zoete, V. SwissADME: a free web tool to evaluate pharmacokinetics, drug–likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717, DOI: 10.1038/srep42717Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czisFSrtg%253D%253D&md5=9715b8cb8a34b17c4c73ff69a5a8cc50SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small moleculesDaina Antoine; Michielin Olivier; Zoete Vincent; Michielin Olivier; Michielin OlivierScientific reports (2017), 7 (), 42717 ISSN:.To be effective as a drug, a potent molecule must reach its target in the body in sufficient concentration, and stay there in a bioactive form long enough for the expected biologic events to occur. Drug development involves assessment of absorption, distribution, metabolism and excretion (ADME) increasingly earlier in the discovery process, at a stage when considered compounds are numerous but access to the physical samples is limited. In that context, computer models constitute valid alternatives to experiments. Here, we present the new SwissADME web tool that gives free access to a pool of fast yet robust predictive models for physicochemical properties, pharmacokinetics, drug-likeness and medicinal chemistry friendliness, among which in-house proficient methods such as the BOILED-Egg, iLOGP and Bioavailability Radar. Easy efficient input and interpretation are ensured thanks to a user-friendly interface through the login-free website http://www.swissadme.ch. Specialists, but also nonexpert in cheminformatics or computational chemistry can predict rapidly key parameters for a collection of molecules to support their drug discovery endeavours.
- 48Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B. A.; Thiessen, P. A.; Yu, B.; Zaslavsky, L.; Zhang, J.; Bolton, E. E. PubChem in 2021: new data content and improved web interfaces. Nucleic Acids Res. 2021, 49, D1388– D1395, DOI: 10.1093/nar/gkaa971Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXntFCit7Y%253D&md5=5bbf4c2b84fc02bbb043cbcc75d4b948PubChem in 2021: new data content and improved web interfacesKim, Sunghwan; Chen, Jie; Cheng, Tiejun; Gindulyte, Asta; He, Jia; He, Siqian; Li, Qingliang; Shoemaker, Benjamin A.; Thiessen, Paul A.; Yu, Bo; Zaslavsky, Leonid; Zhang, Jian; Bolton, Evan E.Nucleic Acids Research (2021), 49 (D1), D1388-D1395CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)PubChem is a popular chem. information resource that serves the scientific community as well as the general public, with millions of unique users per mo. In the past 2 yr, PubChem made substantial improvements. Data from >100 new data sources were added to PubChem, including chem.-literature links from Thieme Chem., chem. and phys. property links from SpringerMaterials, and patent links from the World Intellectual Properties Organization (WIPO). PubChem's homepage and individual record pages were updated to help users find desired information faster. This update involved a data model change for the data objects used by these pages as well as by programmatic users. Several new services were introduced, including the PubChem Periodic Table and Element pages, Pathway pages, and Knowledge panels. Addnl., in response to the coronavirus disease 2019 (COVID-19) outbreak, PubChem created a special data collection that contains PubChem data related to COVID-19 and the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- 49Backman, T. W. H.; Cao, Y.; Girke, T. ChemMine tools: an online service for analyzing and clustering small molecules. Nucleic Acids Res. 2011, 39, W486– W491, DOI: 10.1093/nar/gkr320Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXosVOntbk%253D&md5=99a8d31fed2f8e47c0c81ffd192078a7ChemMine tools: an online service for analyzing and clustering small moleculesBackman, Tyler W. H.; Cao, Yiqun; Girke, ThomasNucleic Acids Research (2011), 39 (Web Server), W486-W491CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChemMine Tools is an online service for small mol. data anal. It provides a web interface to a set of chemoinformatics and data mining tools that are useful for various anal. routines performed in chem. genomics and drug discovery. The service also offers programmable access options via the R library ChemmineR. The primary functionalities of ChemMine Tools fall into five major application areas: data visualization, structure comparisons, similarity searching, compd. clustering and prediction of chem. properties. First, users can upload compd. data sets to the online Compd. Workbench. Numerous utilities are provided for compd. viewing, structure drawing and format interconversion. Second, pairwise structural similarities among compds. can be quantified. Third, interfaces to ultra-fast structure similarity search algorithms are available to efficiently mine the chem. space in the public domain. These include fingerprint and embedding/indexing algorithms. Fourth, the service includes a Clustering Toolbox that integrates chemoinformatic algorithms with data mining utilities to enable systematic structure and activity based analyses of custom compd. sets. Fifth, physicochem. property descriptors of custom compd. sets can be calcd. These descriptors are important for assessing the bioactivity profile of compds. in silico and quant. structure-activity relationship (QSAR) analyses. ChemMine Tools is available at: http://chemmine.ucr.edu.
- 50Studier, F. W.; Moffatt, B. A. Use of bacteriophage T7 RNA polymerase to direct selective high–level expression of cloned genes. J. Mol. Biol. 1986, 189, 113– 130, DOI: 10.1016/0022-2836(86)90385-2Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XktlKrsr4%253D&md5=3219300bc2f640fe9830c7518eb99bfcUse of bacteriophage T7 RNA polymerase to direct selective high-level expression of cloned genesStudier, F. William; Moffatt, Barbara A.Journal of Molecular Biology (1986), 189 (1), 113-30CODEN: JMOBAK; ISSN:0022-2836.A gene expression system based on phage T7 RNA polymerase [9014-24-8] was developed. T7 RNA polymerase is highly selective for its own promoters, which do not occur naturally in Escherichia coli. A relatively small amt. of T7 RNA polymerase provided from a cloned copy of T7 gene 1 is sufficient to direct high-level transcription from a T7 promoter in a multicopy plasmid. Such transcription can proceed several times around the plasmid without terminating and can be so active that transcription by E. coli RNA polymerase is greatly decreased. When a cleavage site for RNase III is introduced, discrete RNAs of plasmid length can accumulate. The natural transcription terminator from T7 DNA also works effectively in the plasmid. Both the rate of synthesis and the accumulation of RNA directed by T7 RNA polymerase can reach levels comparable with those for rRNAs in a normal cell. These high levels of accumulation suggest that the RNAs are relatively stable, perhaps in part because their great length and(or) stem-and-loop structures at their 3' ends help to protect them against exonucleolytic degrdn. Apparently, a specific mRNA produced by T7 RNA polymerase can rapidly sat. the translational machinery of E. coli, so that the rate of protein synthesis from such an mRNA will depend primarily on the efficiency of its translation. When the mRNA is efficiently translated, a target protein can accumulate to >50% of the total cell protein in ≤3 h. Two ways were used to deliver active T7 RNA polymerase to the cell: (1) infection by a λ deriv. that carried gene 1; or (2) induction of a chromosomal copy of gene 1 under control of the lacUV5 promoter. When gene 1 is delivered by infection, very toxic target genes can be maintained silently in the cell until T7 RNA polymerase is introduced, when they rapidly become expressed at high levels. When gene 1 is resident in the chromosome, even the very low basal levels of T7 RNA polymerase present in the uninduced cell can prevent the establishment of plasmids carrying toxic target genes, or make the plasmid unstable. But if the target plasmid can be maintained, induction of chromosomal gene 1 can be a convenient way to produce large amts. of target RNA and(or) protein. T7 RNA polymerase seems to be capable of transcribing almost any DNA linked to a T7 promoter, so the T7 expression system should be capable of transcribing almost any gene or its complement in E. coli. Comparable T7 expression systems can be developed in other types of cells.
- 51Laemmli, U. K. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970, 227, 680– 685, DOI: 10.1038/227680a0Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXlsFags7s%253D&md5=fff3e668784b8bb3669f854be60a216bCleavage of structural proteins during the assembly of the head of bacteriophage T4Laemmli, U. K.Nature (London, United Kingdom) (1970), 227 (5259), 680-685CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Using an improved method of polyacrylamide gel electrophoresis based on the capability of SDS to break down proteins into their individual polypeptide chains, many previously unknown proteins have been found in bacteriophage T4 and some of these have been identified with specific gene products. Four major components of the head are cleaved during the process of assembly, apparently after the precursor proteins have assembled into some large intermediate structure.
- 52Wang, X. D.; Meng, M. X.; Gao, L. B.; Liu, T.; Xu, Q.; Zeng, S. Permeation of astilbin and taxifolin in Caco–2 cell and their effects on the P–gp. Int. J. Pharm. 2009, 378, 1– 8, DOI: 10.1016/j.ijpharm.2009.05.022Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXovFKqtLw%253D&md5=59d129ebdfbe70949d731fdfce20abfdPermeation of astilbin and taxifolin in Caco-2 cell and their effects on the P-gpWang, Xiao-dan; Meng, Min-xin; Gao, Ling-bo; Liu, Ting; Xu, Qiang; Zeng, SuInternational Journal of Pharmaceutics (2009), 378 (1-2), 1-8CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)This study was designed to understand the transport profiles of astilbin and taxifolin in Caco-2 cell model and their effects on the function and expression of P-glycoprotein. The transport studies were examd. using Caco-2 cells cultured on Transwell inserts. Their effects on the function and expression of P-glycoprotein were detected using Western Blot and RT-PCR. The transport was concn. and temp. dependent. The apparent permeability (P app) of these two compds. in the secretory direction was larger than that in the absorptive direction in the concn. range of 10-1000 μM. Those compds. had no effects on the P-glycoprotein-mediated transport of Rhodamine 123. Caco-2 cells exposed to astilbin or taxifolin for 36 h exhibited higher P-glycoprotein activity through up-regulating P-glycoprotein expression at protein and mRNA levels. These results indicated that P-glycoprotein and Multidrug Resistance Protein 2 might play important roles in limiting the bioavailability of those compds. Drugs which are the inhibitors of P-glycoprotein or Multidrug Resistance Protein 2 may increase the oral bioavailability of astilbin or taxifolin and the possibility of unwanted drug-food interactions. The increased expression of P-glycoprotein in Caco-2 cells may serve as an adaptation and defense mechanism in limiting the entry of xenobiotics into the body.
- 53Faralli, A.; Shekarforoush, E.; Ajalloueian, F.; Mendes, A. C.; Chronakis, I. S. In vitro permeability enhancement of curcumin across Caco–2 cells monolayers using electrospun xanthan–chitosan nanofibers. Carbohydr. Polym. 2019, 206, 38– 47, DOI: 10.1016/j.carbpol.2018.10.073Google ScholarThere is no corresponding record for this reference.
- 54Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessmen of docking accuracy. J. Med. Chem. 2004, 47, 1739– 1749, DOI: 10.1021/jm0306430Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit74%253D&md5=8cc2f0022318b12dd972e9c493375bf9Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessment of docking accuracyFriesner, Richard A.; Banks, Jay L.; Murphy, Robert B.; Halgren, Thomas A.; Klicic, Jasna J.; Mainz, Daniel T.; Repasky, Matthew P.; Knoll, Eric H.; Shelley, Mee; Perry, Jason K.; Shaw, David E.; Francis, Perry; Shenkin, Peter S.Journal of Medicinal Chemistry (2004), 47 (7), 1739-1749CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Unlike other methods for docking ligands to the rigid 3D structure of a known protein receptor, Glide approximates a complete systematic search of the conformational, orientational, and positional space of the docked ligand. In this search, an initial rough positioning and scoring phase that dramatically narrows the search space is followed by torsionally flexible energy optimization on an OPLS-AA nonbonded potential grid for a few hundred surviving candidate poses. The very best candidates are further refined via a Monte Carlo sampling of pose conformation; in some cases, this is crucial to obtaining an accurate docked pose. Selection of the best docked pose uses a model energy function that combines empirical and force-field-based terms. Docking accuracy is assessed by redocking ligands from 282 cocrystd. PDB complexes starting from conformationally optimized ligand geometries that bear no memory of the correctly docked pose. Errors in geometry for the top-ranked pose are less than 1 Å in nearly half of the cases and are greater than 2 Å in only about one-third of them. Comparisons to published data on rms deviations show that Glide is nearly twice as accurate as GOLD and more than twice as accurate as FlexX for ligands having up to 20 rotatable bonds. Glide is also found to be more accurate than the recently described Surflex method.
- 55Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra Precision Glide: Docking and scoring incorporating a model of hydrophobic enclosure for protein–ligand complexes. J. Med. Chem. 2006, 49, 6177– 6196, DOI: 10.1021/jm051256oGoogle Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpvVGmurg%253D&md5=ea428c82ead0d8c27f8c1a7b694a1edfExtra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 56Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. J. Med. Chem. 2004, 47, 1750– 1759, DOI: 10.1021/jm030644sGoogle Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit78%253D&md5=33d68dd968e65626b449df61e44e37beGlide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screeningHalgren, Thomas A.; Murphy, Robert B.; Friesner, Richard A.; Beard, Hege S.; Frye, Leah L.; Pollard, W. Thomas; Banks, Jay L.Journal of Medicinal Chemistry (2004), 47 (7), 1750-1759CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Glide's ability to identify active compds. in a database screen is characterized by applying Glide to a diverse set of nine protein receptors. In many cases, two, or even three, protein sites are employed to probe the sensitivity of the results to the site geometry. To make the database screens as realistic as possible, the screens use sets of "druglike" decoy ligands that have been selected to be representative of what we believe is likely to be found in the compd. collection of a pharmaceutical or biotechnol. company. Results are presented for releases 1.8, 2.0, and 2.5 of Glide. The comparisons show that av. measures for both "early" and "global" enrichment for Glide 2.5 are 3 times higher than for Glide 1.8 and more than 2 times higher than for Glide 2.0 because of better results for the least well-handled screens. This improvement in enrichment stems largely from the better balance of the more widely parametrized GlideScore 2.5 function and the inclusion of terms that penalize ligand-protein interactions that violate established principles of phys. chem., particularly as it concerns the exposure to solvent of charged protein and ligand groups. Comparisons to results for the thymidine kinase and estrogen receptors published by Rognan and co-workers (J. Med. Chem. 2000, 43, 4759-4767) show that Glide 2.5 performs better than GOLD 1.1, FlexX 1.8, or DOCK 4.01.
- 57Petrella, S.; Capton, E.; Raynal, B.; Giffard, C.; Thureau, A.; Bonneté, F.; Alzari, P. M.; Aubry, A.; Mayer, C. Overall Structures of Mycobacterium tuberculosis DNA Gyrase Reveal the Role of a Corynebacteriales GyrB-Specific Insert in ATPase Activity. Structure. 2019, 27, 579– 589, DOI: 10.1016/j.str.2019.01.004Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisl2lurw%253D&md5=492e76347e4646be2fb760e0655c4d14Overall Structures of Mycobacterium tuberculosis DNA Gyrase Reveal the Role of a Corynebacteriales GyrB-Specific Insert in ATPase ActivityPetrella, Stephanie; Capton, Estelle; Raynal, Bertrand; Giffard, Clement; Thureau, Aurelien; Bonnete, Francoise; Alzari, Pedro M.; Aubry, Alexandra; Mayer, ClaudineStructure (Oxford, United Kingdom) (2019), 27 (4), 579-589.e5CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)Despite sharing common features, previous studies have shown that gyrases from different species have been modified throughout evolution to modulate their properties. Here, we report two crystal structures of Mycobacterium tuberculosis DNA gyrase, an apo and AMPPNP-bound form at 2.6-Å and 3.3-Å resoln., resp. These structures provide high-resoln. structural data on the quaternary organization and interdomain connections of a gyrase (full-length GyrB-GyrA57)2 thus providing crucial inputs on this essential drug target. Together with small-angle X-ray scattering studies, they revealed an "extremely open" N-gate state, which persists even in the DNA-free gyrase-AMPPNP complex and an unexpected connection between the ATPase and cleavage core domains mediated by two Corynebacteriales-specific motifs, resp. the C-loop and DEEE-loop. We show that the C-loop participates in the stabilization of this open conformation, explaining why this gyrase has a lower ATPase activity. Our results image a conformational state which might be targeted for drug discovery.
- 58Agrawal, A.; Roue, M.; Spitzfaden, C.; Petrella, S.; Aubry, A.; Hann, M.; Bax, B.; Mayer, C. Mycobacterium tuberculosis DNA gyrase ATPase domain structures suggest a dissociative mechanism that explains how ATP hydrolysis is coupled to domain motion. Biochem. J. 2013, 456, 263– 273, DOI: 10.1042/BJ20130538Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCmur3F&md5=761b89faa80294350062b3cbf31ab137Mycobacterium tuberculosis DNA gyrase ATPase domain structures suggest a dissociative mechanism that explains how ATP hydrolysis is coupled to domain motionAgrawal, Alka; Roue, Melanie; Spitzfaden, Claus; Petrella, Stephanie; Aubry, Alexandra; Hann, Michael; Bax, Benjamin; Mayer, ClaudineBiochemical Journal (2013), 456 (2), 263-273CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)DNA gyrase, a type II topoisomerase, regulates DNA topol. by creating a double-stranded break in one DNA duplex and transporting another DNA duplex [T-DNA (transported DNA)] through this break. The ATPase domains dimerize, in the presence of ATP, to trap the T-DNA segment. Hydrolysis of only one of the two ATPs, and release of the resulting Pi, is ratelimiting in DNA strand passage. A long unresolved puzzle is how the non-hydrolysable ATP analog AMP-PNP (adenosine 5'-[β,γ-imido]triphosphate) can catalyze one round of DNA strand passage without Pi release. In the present paper we discuss two crystal structures of the Mycobacterium tuberculosis DNA gyrase ATPase domain: one complexed with AMP-PCP (adenosine 5'-[β,γ-methylene]triphosphate) was unexpectedly monomeric, the other, an AMP-PNP complex, crystd. as a dimer. In the AMP-PNP structure, the unprotonated nitrogen (P-N=P imino) accepts hydrogen bonds from a well-ordered 'ATP lid', which is known to be required for dimerization. The equivalent CH2 group, in AMP-PCP, cannot accept hydrogen bonds, leaving the 'ATP lid' region disordered. Further anal. suggested that AMP-PNP can be converted from the imino (P-N=P) form into the imido form (P-NH-P) during the catalytic cycle. A main-chain NH is proposed to move to either protonate AMP-P-N=P to AMP-P-NH-P, or to protonate ATP to initiate ATP hydrolysis. This suggests a novel dissociative mechanism for ATP hydrolysis that could be applicable not only to GHKL phosphotransferases, but also to unrelated ATPases and GTPases such as Ras. On the basis of the domain orientation in our AMP-PCP structure we propose a mechanochem. scheme to explain how ATP hydrolysis is coupled to domain motion.
- 59Shirude, P. S.; Madhavapeddi, P.; Tucker, J. A.; Murugan, K.; Patil, V.; Basavarajappa, H.; Raichurkar, A. V.; Humnabadkar, V.; Hussein, S.; Sharma, S.; Ramya, V. K.; Narayan, C. B.; Balganesh, T. S.; Sambandamurthy, V. K. Aminopyrazinamides: novel and specific GyrB inhibitors that kill replicating and nonreplicating Mycobacterium tuberculosis. ACS Chem. Biol. 2013, 8, 519– 523, DOI: 10.1021/cb300510wGoogle Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvV2gsLrI&md5=4f8053bd1721c40019c70fab7f6147beAminopyrazinamides: novel and specific GyrB inhibitors that kill replicating and nonreplicating Mycobacterium tuberculosisShirude, Pravin S.; Madhavapeddi, Prashanti; Tucker, Julie A.; Murugan, Kannan; Patil, Vikas; Basavarajappa, Halesha; Raichurkar, Anandkumar V.; Humnabadkar, Vaishali; Hussein, Syeed; Sharma, Sreevalli; Ramya, V. K.; Narayan, Chandan B.; Balganesh, Tanjore S.; Sambandamurthy, Vasan K.ACS Chemical Biology (2013), 8 (3), 519-523CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Aminopyrazinamides originated from a high throughput screen targeting the Mycobacterium smegmatis (Msm) GyrB ATPase. This series displays chem. tractability, robust structure-activity relationship, and potent antitubercular activity. The crystal structure of Msm GyrB in complex with one of the aminopyrazinamides revealed promising attributes of specificity against other broad spectrum pathogens and selectivity against eukaryotic kinases due to novel interactions at hydrophobic pocket, unlike other known GyrB inhibitors. The aminopyrazinamides display excellent mycobacterial kill under in vitro, intracellular, and hypoxic conditions.
- 60Greenwood, J. R.; Calkins, D.; Sullivan, A. P.; Shelley, J. C. Towards the comprehensive, rapid, and accurate prediction of the favorable tautomeric states of drug–like molecules in aqueous solution. J. Comput. Aided Mol. Des. 2010, 24, 591– 604, DOI: 10.1007/s10822-010-9349-1Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnsFGqtbo%253D&md5=1d7bc0f966ca793d6be80554868367b8Towards the comprehensive, rapid, and accurate prediction of the favorable tautomeric states of drug-like molecules in aqueous solutionGreenwood, Jeremy R.; Calkins, David; Sullivan, Arron P.; Shelley, John C.Journal of Computer-Aided Molecular Design (2010), 24 (6-7), 591-604CODEN: JCADEQ; ISSN:0920-654X. (Springer)A review. Generating the appropriate protonation states of drug-like mols. in soln. is important for success in both ligand- and structure-based virtual screening. Screening collections of millions of compds. requires a method for detg. tautomers and their energies that is sufficiently rapid, accurate, and comprehensive. To maximize enrichment, the lowest energy tautomers must be detd. from heterogeneous input, without over-enumerating unfavorable states. While computationally expensive, the d. functional theory (DFT) method M06-2X/aug-cc-pVTZ(-f) [PB-SCRF] provides accurate energies for enumerated model tautomeric systems. The empirical Hammett-Taft methodol. can very rapidly extrapolate substituent effects from model systems to drug-like mols. via the relationship between pKT and pKa. Combining the 2 complementary approaches transforms the tautomer problem from a scientific challenge to one of engineering scale-up, and avoids issues that arise due to the very limited no. of measured pKT values, esp. for the complicated heterocycles often favored by medicinal chemists for their novelty and versatility. Several hundreds of pre-calcd. tautomer energies and substituent pKa effects are tabulated in databases for use in structural adjustment by the program Epik, which treats tautomers as a subset of the larger problem of the protonation states in aq. ensembles and their energy penalties. Accuracy and coverage is continually improved and expanded by parameterizing new systems of interest using DFT and exptl. data. Recommendations are made for how to best incorporate tautomers in mol. design and virtual screening workflows.
- 61Shelley, J. C.; Cholleti, A.; Frye, L. L.; Greenwood, J. R.; Timlin, M. R.; Uchimaya, M. Epik: a software program for pKa prediction and protonation state generation for drug–like molecules. J. Comput. Aided Mol. Des. 2007, 21, 681– 691, DOI: 10.1007/s10822-007-9133-zGoogle Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVKrtbzP&md5=f4f429ea3894e1ad2519cdf3333a5645Epik: a software program for pKa prediction and protonation state generation for drug-like moleculesShelley, John C.; Cholleti, Anuradha; Frye, Leah L.; Greenwood, Jeremy R.; Timlin, Mathew R.; Uchimaya, MakotoJournal of Computer-Aided Molecular Design (2007), 21 (12), 681-691CODEN: JCADEQ; ISSN:0920-654X. (Springer)Epik is a computer program for predicting pKa values for drug-like mols. Epik can use this capability in combination with technol. for tautomerization to adjust the protonation state of small drug-like mols. to automatically generate one or more of the most probable forms for use in further mol. modeling studies. Many medicinal chems. can exchange protons with their environment, resulting in various ionization and tautomeric states, collectively known as protonation states. The protonation state of a drug can affect its soly. and membrane permeability. In modeling, the protonation state of a ligand will also affect which conformations are predicted for the mol., as well as predictions for binding modes and ligand affinities based upon protein-ligand interactions. Despite the importance of the protonation state, many databases of candidate mols. used in drug development do not store reliable information on the most probable protonation states. Epik is sufficiently rapid and accurate to process large databases of drug-like mols. to provide this information. Several new technologies are employed. Extensions to the well-established Hammett and Taft approaches are used for pKa prediction, namely, mesomer standardization, charge cancellation, and charge spreading to make the predicted results reflect the nature of the mol. itself rather just for the particular Lewis structure used on input. In addn., a new iterative technol. for generating, ranking and culling the generated protonation states is employed.
- 62Case, D. A.; Aktulga, H. M.; Belfon, K.; Ben Shalom, I. Y.; Brozell, S. R.; Cerutti, D. S.; Cheatham, III, T. E.; Cisneros, G. A.; Cruzeiro, V. W. D.; Darden, T. A.; Duke, R. E.; Giambasu, G.; Gilson, M. K.; Gohlke, H.; Goetz, A. W.; Harris, R.; Izadi, S.; Izmailov, S. A.; Jin, C.; Kasavajhala, K.; Kaymak, M. C.; King, E.; Kovalenko, A.; Kurtzman, T.; Lee, T. S.; LeGrand, S.; Li, P.; Lin, C.; Liu, J.; Luchko, T.; Luo, R.; Machado, M.; Man, V.; Manathunga, M.; Merz, K. M.; Miao, Y.; Mikhailovskii, O.; Monard, G.; Nguyen, H.; O’Hearn, K. A.; Onufriev, A.; Pan, F.; Pantano, S.; Qi, R.; Rahnamoun, A.; Roe, D. R.; Roitberg, A.; Sagui, C.; Schott Verdugo, S.; Shen, J.; Simmerling, C. L.; Skrynnikov, N. R.; Smith, J.; Swails, J.; Walker, R. C.; Wang, J.; Wei, H.; Wolf, R. M.; Wu, X.; Xue, Y.; York, D. M.; Zhao, S.; Kollman, P. A. Amber 2020; University of California: San Francisco, 2021.Google ScholarThere is no corresponding record for this reference.
- 63Wang, J.; Wolf, R. M.; Caldwell, J. W.; Kollman, P. A.; Case, D. A. Development and testing of a general amber force field. J. Comput. Chem. 2004, 25, 1157– 1174, DOI: 10.1002/jcc.20035Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksFakurc%253D&md5=2992017a8cf51f89290ae2562403b115Development and testing of a general Amber force fieldWang, Junmei; Wolf, Romain M.; Caldwell, James W.; Kollman, Peter A.; Case, David A.Journal of Computational Chemistry (2004), 25 (9), 1157-1174CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)We describe here a general Amber force field (GAFF) for org. mols. GAFF is designed to be compatible with existing Amber force fields for proteins and nucleic acids, and has parameters for most org. and pharmaceutical mols. that are composed of H, C, N, O, S, P, and halogens. It uses a simple functional form and a limited no. of atom types, but incorporates both empirical and heuristic models to est. force consts. and partial at. charges. The performance of GAFF in test cases is encouraging. In test I, 74 crystallog. structures were compared to GAFF minimized structures, with a root-mean-square displacement of 0.26 Å, which is comparable to that of the Tripos 5.2 force field (0.25 Å) and better than those of MMFF 94 and CHARMm (0.47 and 0.44 Å, resp.). In test II, gas phase minimizations were performed on 22 nucleic acid base pairs, and the minimized structures and intermol. energies were compared to MP2/6-31G* results. The RMS of displacements and relative energies were 0.25 Å and 1.2 kcal/mol, resp. These data are comparable to results from Parm99/RESP (0.16 Å and 1.18 kcal/mol, resp.), which were parameterized to these base pairs. Test III looked at the relative energies of 71 conformational pairs that were used in development of the Parm99 force field. The RMS error in relative energies (compared to expt.) is about 0.5 kcal/mol. GAFF can be applied to wide range of mols. in an automatic fashion, making it suitable for rational drug design and database searching.
- 64Bayly, C. I.; Cieplak, P.; Cornell, W. D.; Kollman, P. A. A Well behaved electrostatic potential based method using charge restraints for deriving atomic charges: The RESP model. J. Phys. Chem. 1993, 97, 10269– 10280, DOI: 10.1021/j100142a004Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXlvVyqsLs%253D&md5=e65c6a556ffc174df4f327687912a0bdA well-behaved electrostatic potential based method using charge restraints for deriving atomic charges: the RESP modelBayly, Christopher I.; Cieplak, Piotr; Cornell, Wendy; Kollman, Peter A.Journal of Physical Chemistry (1993), 97 (40), 10269-80CODEN: JPCHAX; ISSN:0022-3654.The authors present a new approach to generating electrostatic potential (ESP) derived charges for mols. The major strength of electrostatic potential derived charges is that they optimally reproduce the intermol. interaction properties of mols. with a simple two-body additive potential, provided, of course, that a suitably accurate level of quantum mech. calcn. is used to derive the ESP around the mol. Previously, the major weaknesses of these charges have been that they were not easily transferably between common functional groups in related mols., they have often been conformationally dependent, and the large charges that frequently occur can be problematic for simulating intramol. interactions. Introducing restraints in the form of a penalty function into the fitting process considerably reduces the above problems, with only a minor decrease in the quality of the fit to the quantum mech. ESP. Several other refinements in addn. to the restrained electrostatic potential (RESP) fit yield a general and algorithmic charge fitting procedure for generating atom-centered point charges. This approach can thus be recommended for general use in mol. mechanics, mol. dynamics, and free energy calcns. for any org. or bioorg. system.
- 65Cieplak, P.; Cornell, W. D.; Bayly, C.; Kollman, P. A. Application of the multimolecule and multiconformational RESP methodology to biopolymers: Charge derivation for DNA, RNA, and proteins. J. Comput. Chem. 1995, 16, 1357– 1377, DOI: 10.1002/jcc.540161106Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXovVKqtrY%253D&md5=27e75f5f3f53d777737661e591e957cdApplication of the multimolecule and multiconformational RESP methodology to biopolymers: charge derivation for DNA, RNA, and proteinsCieplak, Piotr; Cornell, Wendy D.; Bayly, Christopher; Kollman, Peter A.Journal of Computational Chemistry (1995), 16 (11), 1357-77CODEN: JCCHDD; ISSN:0192-8651. (Wiley)The authors present the derivation of charges of ribo- and deoxynucleosides, nucleotides, and peptide fragments using electrostatic potentials obtained from ab initio calcns. with the 6-31G* basis set. For the nucleic acid fragments, the authors used electrostatic potentials of the four deoxyribonucleoside (A, G, C, T) and four ribonucleosides (A, G, C, U) and dimethylphosphate. The charges for the deoxyribose nucleosides and nucleotides are derived using multiple-mol. fitting and restrained electrostatic potential (RESP) fits, with Lagrangian multipliers ensuring a net charge of 0 or. The authors suggest that the preferred approach for deriving charges for nucleosides and nucleotides involves allowing only C1' and H1' of the sugar to vary as the nucleic acid base, with the remainder of sugar and backbone atoms forced to be equiv. For peptide fragments, the authors have combined multiple conformation fitting, previously employed by Williams and Reynolds et al., with the RESP approach to derive charges for blocked dipeptides appropriate for each of the 20 naturally occurring amino acids. Based on the results for Pr amine, the authors suggest that two conformations for each peptide suffice to give charges that represent well the conformationally dependent electrostatic properties of mols., provided that these two conformations contain different values of the dihedral angles that terminate in heteroatoms or hydrogens attached to heteroatoms or hydrogens attached to heteroatoms. In these blocked dipeptide models, it is useful to require equiv. N-H and C=O charges for all amino acids with a given net charge (except proline), and this is accomplished in a straightforward fashion with multiple-mol. fitting. Finally, the application of multiple Lagrangian constraints allows for the derivation of monomeric residues with the appropriate net charge from a chem. blocked version of the residue. The multiple Lagrange constraints also enable charges from two or more mols. to be spliced together in a well-defined fashion. Thus, the combined use of multiple mols., multiple conformations, multiple Lagrangian constraints, and RESP fitting is shown to be a powerful approach to deriving electrostatic charges for biopolymers.
- 66Fox, T.; Kollman, P. A. Application of the RESP methodology in the parametrization of organic solvents. J. Phys. Chem. B 1998, 102, 8070– 8079, DOI: 10.1021/jp9717655Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXmtVelu78%253D&md5=848186c21337d0ff82cafd35a3a526a4Application of the RESP Methodology in the Parametrization of Organic SolventsFox, Thomas; Kollman, Peter A.Journal of Physical Chemistry B (1998), 102 (41), 8070-8079CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)We present parameterizations for the nonaq. solvents DMSO, ethanol, CCl4, CHCl3, and CH2Cl2 that are compatible with the recent AMBER force field by Cornell et al. (J. Am. Chem. Soc. 1995, 117, 5179-5197). With the general procedure for generating new parameters and the RESP approach to obtain the at. charges, we achieve flexible all-atom solvent models whose d., heat of vaporization, diffusion const., and rotational correlation times are-esp. for a generic force field-in good agreement with available exptl. data.
- 67Jorgensen, W. L.; Chandrasekhar, J.; Madura, J. D.; Impey, R. W.; Klein, M. L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 1983, 79, 926– 935, DOI: 10.1063/1.445869Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3sXksF2htL4%253D&md5=a1161334e381746be8c9b15a5e56f704Comparison of simple potential functions for simulating liquid waterJorgensen, William L.; Chandrasekhar, Jayaraman; Madura, Jeffry D.; Impey, Roger W.; Klein, Michael L.Journal of Chemical Physics (1983), 79 (2), 926-35CODEN: JCPSA6; ISSN:0021-9606.Classical Monte Carlo simulations were carried out for liq. H2O in the NPT ensemble at 25° and 1 atm using 6 of the simpler intermol. potential functions for the dimer. Comparisons were made with exptl. thermodn. and structural data including the neutron diffraction results of Thiessen and Narten (1982). The computed densities and potential energies agree with expt. except for the original Bernal-Fowler model, which yields an 18% overest. of the d. and poor structural results. The discrepancy may be due to the correction terms needed in processing the neutron data or to an effect uniformly neglected in the computations. Comparisons were made for the self-diffusion coeffs. obtained from mol. dynamics simulations.
- 68Roe, D. R.; Cheatham, T., III. PTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory Data. J. Chem. Theory Comput. 2013, 9, 3084– 3095, DOI: 10.1021/ct400341pGoogle Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFehtr8%253D&md5=6f1bee934f13f180bd7e1feb6b78036dPTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory DataRoe, Daniel R.; Cheatham, Thomas E.Journal of Chemical Theory and Computation (2013), 9 (7), 3084-3095CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We describe PTRAJ and its successor CPPTRAJ, two complementary, portable, and freely available computer programs for the anal. and processing of time series of three-dimensional at. positions (i.e., coordinate trajectories) and the data therein derived. Common tools include the ability to manipulate the data to convert among trajectory formats, process groups of trajectories generated with ensemble methods (e.g., replica exchange mol. dynamics), image with periodic boundary conditions, create av. structures, strip subsets of the system, and perform calcns. such as RMS fitting, measuring distances, B-factors, radii of gyration, radial distribution functions, and time correlations, among other actions and analyses. Both the PTRAJ and CPPTRAJ programs and source code are freely available under the GNU General Public License version 3 and are currently distributed within the AmberTools 12 suite of support programs that make up part of the Amber package of computer programs (see http://ambermd.org). This overview describes the general design, features, and history of these two programs, as well as algorithmic improvements and new features available in CPPTRAJ.
- 69Wang, E.; Sun, H.; Wang, J.; Wang, Z.; Liu, H.; Zhang, J. Z. H.; Hou, T. End–point binding free energy calculation with MM/PBSA and MM/GBSA: Strategies and applications in drug design. Chem. Rev. 2019, 119, 9478– 9508, DOI: 10.1021/acs.chemrev.9b00055Google ScholarThere is no corresponding record for this reference.
- 70Hou, T.; Yu, R. Molecular dynamics and free energy studies on the wild–type and double mutant HIV–1 protease complexed with amprenavir and two amprenavir–related inhibitors: mechanism for binding and drug resistance. J. Med. Chem. 2007, 50, 1177– 1188, DOI: 10.1021/jm0609162Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhs1Ghtbk%253D&md5=e13654603bac0ccfc57e8720e98cf8eeMolecular Dynamics and Free Energy Studies on the Wild-type and Double Mutant HIV-1 Protease Complexed with Amprenavir and Two Amprenavir-Related Inhibitors: Mechanism for Binding and Drug ResistanceHou, Tingjun; Yu, RonJournal of Medicinal Chemistry (2007), 50 (6), 1177-1188CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The V82F/I84V double mutation is considered as the key residue mutation of the HIV-1 protease drug resistance because it can significantly lower the binding affinity of protease inhibitors in clin. uses. In the current work, the binding of amprenavir to both of the wild-type and the drug-resistant V82F/I84V mutant of the HIV-1 protease was investigated by mol. dynamics (MD) simulations and was compared to those of two inhibitors in development, TMC126 and TMC114. Abs. binding free energies were calcd. by mol. mechanics/Poisson-Boltzmann surface area (MM/ PBSA) methodol. The predicted binding affinities give a good explanation of structure-affinity relation (SAR) of three studied inhibitors. Furthermore, in the 18 ns MD simulations on the free wild-type and the mutated proteases, we obsd. that the free mutated protease shows similar dynamic characteristics of the flap opening and a little higher structural stability than the free wild-type protease. This suggests that the effect of the mutations may not significantly affect the equil. between the semi-open and the closed conformations. Finally, decompn. anal. of binding free energies and the further structural anal. indicate that the dominating effect of the V82F/I84V double mutation is to distort the geometry of the binding site and hence weaken the interactions of inhibitors pre-shaped to the wild-type binding site.
- 71Li, W.; Zhang, J.; Guo, L.; Wang, Q. Importance of three–body problems and protein–protein interactions in proteolysis–targeting chimera modeling: Insights from molecular dynamics simulations. J. Chem. Inf. Model. 2022, 62, 523– 532, DOI: 10.1021/acs.jcim.1c01150Google ScholarThere is no corresponding record for this reference.
- 72Miller, B. R.; McGee, T. D.; Swails, J. M.; Homeyer, N.; Gohlke, H.; Roitberg, A. E. MMPBSA.py: An efficient program for end–state free energy calculations. J. Chem. Theory Comput. 2012, 8, 3314– 3321, DOI: 10.1021/ct300418hGoogle Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2gtrzP&md5=cc4148bd8f70c7cad94fd3ec6f580e52MMPBSA.py: An Efficient Program for End-State Free Energy CalculationsMiller, Bill R., III; McGee, T. Dwight, Jr.; Swails, Jason M.; Homeyer, Nadine; Gohlke, Holger; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (9), 3314-3321CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)MM-PBSA is a post-processing end-state method to calc. free energies of mols. in soln. MMPBSA.py is a program written in Python for streamlining end-state free energy calcns. using ensembles derived from mol. dynamics (MD) or Monte Carlo (MC) simulations. Several implicit solvation models are available with MMPBSA.py, including the Poisson-Boltzmann Model, the Generalized Born Model, and the Ref. Interaction Site Model. Vibrational frequencies may be calcd. using normal mode or quasi-harmonic anal. to approx. the solute entropy. Specific interactions can also be dissected using free energy decompn. or alanine scanning. A parallel implementation significantly speeds up the calcn. by dividing frames evenly across available processors. MMPBSA.py is an efficient, user-friendly program with the flexibility to accommodate the needs of users performing end-state free energy calcns. The source code can be downloaded at http://ambermd.org/ with AmberTools, released under the GNU General Public License.
- 73Onufriev, A.; Bashford, D.; Case, D. A. Exploring protein native states and large–scale conformational changes with a modified generalized born model. Proteins. 2004, 55, 383– 394, DOI: 10.1002/prot.20033Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjtFKhs78%253D&md5=06bab80e7df579625cb29a4ae7260d0cExploring protein native states and large-scale conformational changes with a modified Generalized Born modelOnufriev, Alexey; Bashford, Donald; Case, David A.Proteins: Structure, Function, and Bioinformatics (2004), 55 (2), 383-394CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)Implicit solvation models provide, for many applications, a reasonably accurate and computationally effective way to describe the electrostatics of aq. solvation. Here, a popular anal. Generalized Born (GB) solvation model is modified to improve its accuracy in calcg. the solvent polarization part of free energy changes in large-scale conformational transitions, such as protein folding. In contrast to an earlier GB model (implemented in the AMBER-6 program), the improved version does not overstabilize the native structures relative to the finite-difference Poisson-Boltzmann continuum treatment. In addn. to improving the energy balance between folded and unfolded conformers, the algorithm (available in the AMBER-7 and NAB mol. modeling packages) is shown to perform well in more than 50 ns of native-state mol. dynamics (MD) simulations of thioredoxin, protein-A, and ubiquitin, as well as in a simulation of Barnase/Barstar complex formation. For thioredoxin, various combinations of input parameters have been explored, such as the underlying gas-phase force fields and the at. radii. The best performance is achieved with a previously proposed modification to the torsional potential in the Amber ff99 force field, which yields stable native trajectories for all of the tested proteins, with back-bone root-mean-square deviations from the native structures being ∼ 1.5 Å after 6 ns of simulation time. The structure of Barnase/Barstar complex is regenerated, starting from an unbound state, to within 1.9 Å relative to the crystal structure of the complex.
- 74Weiser, J.; Shenkin, P. S.; Still, W. C. Approximate atomic surfaces from linear combinations of pairwise overlaps (LCPO). J. Comput. Chem. 1999, 20, 217– 230, DOI: 10.1002/(SICI)1096-987X(19990130)20:2<217::AID-JCC4>3.0.CO;2-AGoogle Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXltVCltQ%253D%253D&md5=1c6cf923fe089cf61a1952fdcf55f788Approximate atomic surfaces from linear combinations of pairwise overlaps (LCPO)Weiser, Jorg; Shenkin, Peter S.; Still, W. ClarkJournal of Computational Chemistry (1999), 20 (2), 217-230CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)A fast anal. formula was derived for the calcn. of approx. at. and mol. van der Waals (vdWSA), and solvent-accessible surface areas (SASAs), as well as the first and second derivs. of these quantities with respect to at. coordinates. This method makes use of linear combinations of terms composed from pairwise overlaps of hard spheres; therefore, we term this the LCPO method for linear combination of pairwise overlaps. For higher performance, neighbor-list redn. (NLR) was applied as a preprocessing step. Eighteen compds. of different sizes (8-2366 atoms) and classes (org., proteins, DNA, and various complexes) were chosen as representative test cases. LCPO/NLR computed the SASA and first derivs. of penicillopepsin, a protein with 2366 atoms, in 0.87 s (0.22 s for the creation of the neighbor list, 0.35 s for NLR, and 0.30 s for SASA and first derivs.) on an SGI R10000/194 Mhz processor. This appears comparable to or better than timings reported previously for other algorithms. The vdWSAs were in good agreement with the numerical results: relative errors for total mol. surface areas ranged from 0.1 to 2.0% and av. abs. at. surface area deviations from 0.3 to 0.7 Å2. For SASAs without NLR, the LCPO method exhibited relative errors in the range of 0.4-9.2% for total mol. surface areas and av. abs. at. surface area deviations of 2.0-2.7 Å2; with NLR the relative mol. errors ranged from 0.1 to 7.8% and the av. abs. at. surface area deviation from 1.6 to 3.0 Å2.
- 75Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 2001, 46, 3– 26, DOI: 10.1016/S0169-409X(00)00129-0Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXitVOhs7o%253D&md5=c60bb89da68f051c0ee7ac4c0468a0e4Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settingsLipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J.Advanced Drug Delivery Reviews (2001), 46 (1-3), 3-26CODEN: ADDREP; ISSN:0169-409X. (Elsevier Science Ireland Ltd.)A review with 50 refs. Exptl. and computational approaches to est. soly. and permeability in discovery and development settings are described. In the discovery setting 'the rule of 5' predicts that poor absorption or permeation is more likely when there are more than 5 H-bond donors, 10 H-bond acceptors, the mol. wt. (MWT) is greater than 500 and the calcd. Log P (CLogP) is greater than 5 (or MlogP >4.15). Computational methodol. for the rule-based Moriguchi Log P (MLogP) calcn. is described. Turbidimetric soly. measurement is described and applied to known drugs. High throughput screening (HTS) leads tend to have higher MWT and Log P and lower turbidimetric soly. than leads in the pre-HTS era. In the development setting, soly. calcns. focus on exact value prediction and are difficult because of polymorphism. Recent work on linear free energy relationships and Log P approaches are critically reviewed. Useful predictions are possible in closely related analog series when coupled with exptl. thermodn. soly. measurements.
- 76Baell, J. B.; Holloway, G. A. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J. Med. Chem. 2010, 53, 2719– 2740, DOI: 10.1021/jm901137jGoogle Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsF2qsLw%253D&md5=fbf397aa4910753c550425708c866fd2New Substructure Filters for Removal of Pan Assay Interference Compounds (PAINS) from Screening Libraries and for Their Exclusion in BioassaysBaell, Jonathan B.; Holloway, Georgina A.Journal of Medicinal Chemistry (2010), 53 (7), 2719-2740CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)This report describes a no. of substructural features which can help to identify compds. that appear as frequent hitters (promiscuous compds.) in many biochem. high throughput screens. The compds. identified by such substructural features are not recognized by filters commonly used to identify reactive compds. Even though these substructural features were identified using only one assay detection technol., such compds. have been reported to be active from many different assays. In fact, these compds. are increasingly prevalent in the literature as potential starting points for further exploration, whereas they may not be.
- 77Alt, S.; Mitchenall, L. A.; Maxwell, A.; Heide, L. Inhibition of DNA gyrase and DNA topoisomerase IV of Staphylococcus aureus and Escherichia coli by aminocoumarin antibiotics. J. Antimicrob. Chemother. 2011, 66, 2061– 2069, DOI: 10.1093/jac/dkr247Google Scholar77https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVeitLvE&md5=ee96fa3dfea838f59610bd5ac1cce584Inhibition of DNA gyrase and DNA topoisomerase IV of Staphylococcus aureus and Escherichia coli by aminocoumarin antibioticsAlt, Silke; Mitchenall, Lesley A.; Maxwell, Anthony; Heide, LutzJournal of Antimicrobial Chemotherapy (2011), 66 (9), 2061-2069CODEN: JACHDX; ISSN:0305-7453. (Oxford University Press)Aminocoumarin antibiotics are potent inhibitors of bacterial DNA gyrase. We investigated the inhibitory and antibacterial activity of naturally occurring aminocoumarin antibiotics and six structural analogs (novclobiocins) against DNA gyrase and DNA topoisomerase IV from Escherichia coli and Staphylococcus aureus as well as the effect of potassium and sodium glutamate on the activity of these enzymes. The inhibitory concns. of the aminocoumarins were detd. in gyrase supercoiling assays and topoisomerase IV decatenation assays. Both subunits of S. aureus topoisomerase IV were purified as His-Tag proteins in E. coli. The MIC was tested in vivo for the control organisms E. coli ATCC 25922 and S. aureus ATCC 29213. DNA gyrase is the primary target in vitro of all investigated aminocoumarins. With the exception of simocyclinone D8, all other aminocoumarins inhibited S. aureus gyrase on av. 6-fold more effectively than E. coli gyrase. Potassium glutamate is essential for the activity of S. aureus gyrase and increases the sensitivity of E. coli gyrase to aminocoumarins ≥10-fold. The antibacterial activity of the tested compds. mirrored their relative activities against topoisomerases. The study provides insights about the substituents that are important for the inhibitory activity of aminocoumarins against the target enzymes, which will facilitate the rational design of improved antibiotics.
- 78Bisacchi, G. S.; Manchester, J. I. A New-Class Antibacterial-Almost. Lessons in Drug Discovery and Development: A Critical Analysis of More than 50 Years of Effort toward ATPase Inhibitors of DNA Gyrase and Topoisomerase IV. ACS. Infect. Dis. 2015, 1, 4– 41, DOI: 10.1021/id500013tGoogle ScholarThere is no corresponding record for this reference.
- 79Duma, R. J.; Warner, J. F. In Vitro Activity of Coumermycin A1 Against Mycobacterium tuberculosis var. hominis. Appl. Microbiol. 1969, 18, 404– 405, DOI: 10.1128/am.18.3.404-405.1969Google ScholarThere is no corresponding record for this reference.
- 80Broeck, A. V.; McEwen, A. G.; Chebaro, Y.; Potier, N.; Lamour, V. Structural Basis for DNA Gyrase Interaction with Coumermycin A1. J. Med. Chem. 2019, 62, 4225– 4231, DOI: 10.1021/acs.jmedchem.8b01928Google ScholarThere is no corresponding record for this reference.
- 81Gl, B.; Rajput, R.; Gupta, M.; Dahiya, P.; Thakur, J. K.; Bhatnagar, R.; Grover, A. Structure-based drug repurposing to inhibit the DNA gyrase of Mycobacterium tuberculosis. Biochem. J. 2020, 477, 4167– 4190, DOI: 10.1042/BCJ20200462Google Scholar81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s%252FovVGhsQ%253D%253D&md5=b16377b6b133bbe1ae523f52dd7e8725Structure-based drug repurposing to inhibit the DNA gyrase of Mycobacterium tuberculosisGl Balasubramani; Rajput Rinky; Grover Abhinav; Gupta Manish; Bhatnagar Rakesh; Dahiya Pradeep; Thakur Jitendra K; Bhatnagar RakeshThe Biochemical journal (2020), 477 (21), 4167-4190 ISSN:.Drug repurposing is an alternative avenue for identifying new drugs to treat tuberculosis (TB). Despite the broad-range of anti-tubercular drugs, the emergence of multi-drug-resistant and extensively drug-resistant strains of Mycobacterium tuberculosis (Mtb) H37Rv, as well as the significant death toll globally, necessitates the development of new and effective drugs to treat TB. In this study, we have employed a drug repurposing approach to address this drug resistance problem by screening the drugbank database to identify novel inhibitors of the Mtb target enzyme, DNA gyrase. The compounds were screened against the ATPase domain of the gyrase B subunit (MtbGyrB47), and the docking results showed that echinacoside, doxorubicin, epirubicin, and idarubicin possess high binding affinities against MtbGyrB47. Comprehensive assessment using fluorescence spectroscopy, surface plasmon resonance spectroscopy (SPR), and circular dichroism (CD) titration studies revealed echinacoside as a potent binder of MtbGyrB47. Furthermore, ATPase, and DNA supercoiling assays exhibited an IC50 values of 2.1-4.7 μM for echinacoside, doxorubicin, epirubicin, and idarubicin. Among these compounds, the least MIC90 of 6.3 and 12 μM were observed for epirubicin and echinacoside, respectively, against Mtb. Our findings indicate that echinacoside and epirubicin targets mycobacterial DNA gyrase, inhibit its catalytic cycle, and retard mycobacterium growth. Further, these compounds exhibit potential scaffolds for optimizing novel anti-mycobacterial agents that can act on drug-resistant strains.
- 82Sherer, B. A.; Hull, K.; Green, O.; Basarab, G.; Hauck, S.; Hill, P.; Loch, J. T., 3rd.; Mullen, G.; Bist, S.; Bryant, J.; Boriack-Sjodin, A.; Read, J.; DeGrace, N.; Uria-Nickelsen, M.; Illingworth, R. N.; Eakin, A. E. Pyrrolamide DNA gyrase inhibitors: Optimization of antibacterial activity and efficacy. Bioorg. Med. Chem. Lett. 2011, 21, 7416– 7420, DOI: 10.1016/j.bmcl.2011.10.010Google Scholar82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFSqtLnO&md5=0511dac22318e0af1337593e81f6d309Pyrrolamide DNA gyrase inhibitors: Optimization of antibacterial activity and efficacySherer, Brian A.; Hull, Kenneth; Green, Oluyinka; Basarab, Gregory; Hauck, Sheila; Hill, Pamela; Loch, James T., III; Mullen, George; Bist, Shanta; Bryant, Joanna; Boriack-Sjodin, Ann; Read, Jon; DeGrace, Nancy; Uria-Nickelsen, Maria; Illingworth, Ruth N.; Eakin, Ann E.Bioorganic & Medicinal Chemistry Letters (2011), 21 (24), 7416-7420CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The pyrrolamides are a new class of antibacterial agents targeting DNA gyrase, an essential enzyme across bacterial species and inhibition results in the disruption of DNA synthesis and subsequently, cell death. The optimization of biochem. activity and other drug-like properties through substitutions to the pyrrole, piperidine, and heterocycle portions of the mol. resulted in pyrrolamides with improved cellular activity and in vivo efficacy.
- 83Durcik, M.; Nyerges, Á.; Skok, Ž.; Skledar, D. G.; Trontelj, J.; Zidar, N.; Ilaš, J.; Zega, A.; Cruz, C. D.; Tammela, P.; Welin, M.; Kimbung, Y. R.; Focht, D.; Benek, O.; Révész, T.; Draskovits, G.; Szili, PÉ; Daruka, L.; Pál, C.; Kikelj, D.; Mašič, L. P.; Tomašič, T. New dual ATP-competitive inhibitors of bacterial DNA gyrase and topoisomerase IV active against ESKAPE pathogens. Eur. J. Med. Chem. 2021, 213, 113200 DOI: 10.1016/j.ejmech.2021.113200Google Scholar83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjtVWisLY%253D&md5=cd3848925844b467ff2d46ab988c67c9New dual ATP-competitive inhibitors of bacterial DNA gyrase and topoisomerase IV active against ESKAPE pathogensDurcik, Martina; Nyerges, Akos; Skok, Ziga; Skledar, Darja Gramec; Trontelj, Jurij; Zidar, Nace; Ilas, Janez; Zega, Anamarija; Cruz, Cristina D.; Tammela, Paivi; Welin, Martin; Kimbung, Yengo R.; Focht, Dorota; Benek, Ondrej; Revesz, Tamas; Draskovits, Gabor; Szili, Petra Eva; Daruka, Lejla; Pal, Csaba; Kikelj, Danijel; Masic, Lucija Peterlin; Tomasic, TihomirEuropean Journal of Medicinal Chemistry (2021), 213 (), 113200CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Here, the design and structure-activity relationship anal. of balanced, low nanomolar inhibitors of bacterial DNA gyrase and topoisomerase IV that show potent antibacterial activities against the ESKAPE pathogens were reported. For inhibitor I•HCl (R = 2-(morpholin-4-yl)ethyl), a crystal structure in complex with Staphylococcus aureus DNA gyrase B was obtained that confirms the mode of action of these compds. The best inhibitor, I [R = thiophen-3-yl-Me (II)], does not show any in vitro cytotoxicity and has excellent potency against Gram-pos. (MICs: range, 0.0078-0.0625μg/mL) and Gram-neg. pathogens (MICs: range, 1-2μg/mL). Furthermore, II inhibits GyrB mutants that can develop resistance to other drugs. Based on these data, it was expected that structural derivs. of II will represent a step toward clin. efficacious multitargeting antimicrobials that are not impacted by existing antimicrobial resistance.
- 84Mesleh, M. F.; Cross, J. B.; Zhang, J.; Kahmann, J.; Andersen, O. A.; Barker, J.; Cheng, R. K.; Felicetti, B.; Wood, M.; Hadfield, A. T.; Scheich, C.; Moy, T. I.; Yang, Q.; Shotwell, J.; Nguyen, K.; Lippa, B.; Dolle, R.; Ryan, M. D. Fragment-based discovery of DNA gyrase inhibitors targeting the ATPase subunit of GyrB. Bioorg. Med. Chem. Lett. 2016, 26, 1314– 1318, DOI: 10.1016/j.bmcl.2016.01.009Google ScholarThere is no corresponding record for this reference.
- 85Ushiyama, F.; Amada, H.; Takeuchi, T.; Tanaka-Yamamoto, N.; Kanazawa, H.; Nakano, K.; Mima, M.; Masuko, A.; Takata, I.; Hitaka, K.; Iwamoto, K.; Sugiyama, H.; Ohtake, N. Lead Identification of 8-(Methylamino)-2-oxo-1,2-dihydroquinoline Derivatives as DNA Gyrase Inhibitors: Hit-to-Lead Generation Involving Thermodynamic Evaluation. ACS Omega 2022, 5, 10145– 10159, DOI: 10.1021/acsomega.0c00865Google ScholarThere is no corresponding record for this reference.
- 86Skok, Ž.; Barančoková, M.; Benek, O.; Cruz, C. D.; Tammela, P.; Tomašič, T.; Zidar, N.; Mašič, L. P.; Zega, A.; Stevenson, C. E. M.; Mundy, J. E. A.; Lawson, D. M.; Maxwell, A.; Kikelj, D.; Ilaš, J. Exploring the Chemical Space of Benzothiazole-Based DNA Gyrase B Inhibitors. ACS Med. Chem. Lett. 2020, 11, 2433– 2440, DOI: 10.1021/acsmedchemlett.0c00416Google Scholar86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVKhtL3J&md5=03957be06e8e796ec89e3a4e05d996b4Exploring the Chemical Space of Benzothiazole-Based DNA Gyrase B InhibitorsSkok, Ziga; Barancokova, Michaela; Benek, Ondrej; Cruz, Cristina Durante; Tammela, Paivi; Tomasic, Tihomir; Zidar, Nace; Masic, Lucija Peterlin; Zega, Anamarija; Stevenson, Clare E. M.; Mundy, Julia E. A.; Lawson, David M.; Maxwell, Anthony; Kikelj, Danijel; Ilas, JanezACS Medicinal Chemistry Letters (2020), 11 (12), 2433-2440CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)We designed and synthesized a series of inhibitors of the bacterial enzymes DNA gyrase and DNA topoisomerase IV, based on our recently published benzothiazole-based inhibitor bearing an oxalyl moiety. To improve the antibacterial activity and retain potent enzymic activity, we systematically explored the chem. space. Several strategies of modification were followed: varying substituents on the pyrrole carboxamide moiety, alteration of the central scaffold, including variation of substitution position and, most importantly, modification of the oxalyl moiety. Compds. with acidic, basic, and neutral properties were synthesized. To understand the mechanism of action and binding mode, we have obtained a crystal structure of compd. 16a, bearing a primary amino group, in complex with the N-terminal domain of E. coli gyrase B (24 kDa) (PDB: 6YD9). Compd. 15a, with a low mol. wt. of 383 Da, potent inhibitory activity on E. coli gyrase (IC50 = 9.5 nM), potent antibacterial activity on E. faecalis (MIC = 3.13μM), and efflux impaired E. coli strain (MIC = 0.78μM), is an important contribution for the development of novel gyrase and topoisomerase IV inhibitors in Gram-neg. bacteria.
- 87Brvar, M.; Perdih, A.; Renko, M.; Anderluh, G.; Turk, D.; Solmajer, T. Structure-based discovery of substituted 4,5′-bithiazoles as novel DNA gyrase inhibitors. J. Med. Chem. 2012, 55, 6413– 6426, DOI: 10.1021/jm300395dGoogle Scholar87https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XptF2ns78%253D&md5=3701c7c3442de99bcaf8e76c51933991Structure-Based Discovery of Substituted 4,5'-Bithiazoles as Novel DNA Gyrase InhibitorsBrvar, Matjaz; Perdih, Andrej; Renko, Miha; Anderluh, Gregor; Turk, Dusan; Solmajer, TomJournal of Medicinal Chemistry (2012), 55 (14), 6413-6426CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bacterial DNA gyrase is a well-established and validated target for the development of novel antibacterials. Starting from the available structural information about the binding of the natural product inhibitor, clorobiocin, we identified a novel series of 4'-methyl-N2-phenyl-[4,5'-bithiazole]-2,2'-diamine inhibitors of gyrase B with a low micromolar inhibitory activity by implementing a two-step structure-based design procedure. This novel class of DNA gyrase inhibitors was extensively investigated by various techniques (differential scanning fluorimetry, surface plasmon resonance, and microscale thermophoresis). The binding mode of the potent inhibitor 18 was revealed by X-ray crystallog., confirming our initial in silico binding model. Furthermore, the high resoln. of the complex structure allowed for the placement of the Gly97-Ser108 flexible loop, thus revealing its role in binding of this class of compds. The crystal structure of the complex protein G24 and inhibitor 18 provides valuable information for further optimization of this novel class of DNA gyrase B inhibitors.
- 88Manchester, J. I.; Dussault, D. D.; Rose, J. A.; Boriack-Sjodin, P. A.; Uria-Nickelsen, M.; Ioannidis, G.; Bist, S.; Fleming, P.; Hull, K. G. Discovery of a novel azaindole class of antibacterial agents targeting the ATPase domains of DNA gyrase and Topoisomerase IV. Bioorg. Med. Chem. Lett. 2012, 22, 5150– 5156, DOI: 10.1016/j.bmcl.2012.05.128Google Scholar88https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVOqt7bL&md5=7c2e42ef7a21c7805736065e84d86a49Discovery of a novel azaindole class of antibacterial agents targeting the ATPase domains of DNA gyrase and Topoisomerase IVManchester, John I.; Dussault, Daemian D.; Rose, Jonathan A.; Boriack-Sjodin, P. Ann; Uria-Nickelsen, Maria; Ioannidis, Georgine; Bist, Shanta; Fleming, Paul; Hull, Kenneth G.Bioorganic & Medicinal Chemistry Letters (2012), 22 (15), 5150-5156CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The discovery and optimization of a series of bacterial topoisomerase inhibitors, e.g., I, is disclosed. Starting from a virtual screening hit, activity was optimized through a combination of structure-based design and phys. property optimization. Synthesis of fewer than a dozen compds. was required to achieve inhibition of the growth of methicillin-resistant Staphyloccus aureus at compd. concns. of 1.56 μM. These compds. simultaneously inhibit DNA gyrase and Topoisomerase IV at similar nanomolar concns., reducing the likelihood of the spontaneous occurrence of target-based mutations resulting in antibiotic resistance, an increasing threat in the treatment of serious infections.
- 89Gross, C. H.; Parsons, J. D.; Grossman, T. H.; Charifson, P. S.; Bellon, S.; Jernee, J.; Dwyer, M.; Chambers, S. P.; Markland, W.; Botfield, M.; Raybuck, S. A. Active–site residues of Escherichia coli DNA gyrase required in coupling ATP hydrolysis to DNA supercoiling and amino acid substitutions leading to novobiocin resistance. Antimicrob. Agents Chemother. 2003, 47, 1037– 1046, DOI: 10.1128/AAC.47.3.1037-1046.2003Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 3 publications.
- Iqball Faheem, Valakunja Nagaraja. Multifunctional Mycobacterial Topoisomerases with Distinctive Features. ACS Infectious Diseases 2025, Article ASAP.
- K. Rajakumari, K. Aravind, M. Balamugundhan, Manjunathan Jagadeesan, Ambiga Somasundaram, Parthiban Brindha Devi, Pasiyappazham Ramasamy. Comprehensive review of DNA gyrase as enzymatic target for drug discovery and development. European Journal of Medicinal Chemistry Reports 2024, 12 , 100233. https://doi.org/10.1016/j.ejmcr.2024.100233
- Balazs Balega, Michael Beer, James Spencer, Giorgio Colombo, Stefano A. Serapian, A. Sofia F. Oliveira, Adrian J. Mulholland. Dynamical nonequilibrium molecular dynamics simulations reveal allosteric networks, signal transduction mechanisms, and sites associated with drug resistance in biomolecular systems. Molecular Physics 2024, 6 https://doi.org/10.1080/00268976.2024.2428350
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 89 other publications.
- 1WHO. Global tuberculosis report 2023. https://www.who.int/publications/i/item/9789240083851 (accessed January 2024).There is no corresponding record for this reference.
- 2Aubry, A.; Fisher, L. M.; Jarlier, V.; Cambau, E. First functional characterization of a singly expressed bacterial type II topoisomerase: the enzyme from Mycobacterium tuberculosis. Biochem. Biophys. Res. Commun. 2006, 348, 158– 165, DOI: 10.1016/j.bbrc.2006.07.0172https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xot1Wqtbg%253D&md5=763bef9f46308f23af208f537e82c1bdFirst functional characterization of a singly expressed bacterial type II topoisomerase: The enzyme from Mycobacterium tuberculosisAubry, Alexandra; Mark Fisher, L.; Jarlier, Vincent; Cambau, EmmanuelleBiochemical and Biophysical Research Communications (2006), 348 (1), 158-165CODEN: BBRCA9; ISSN:0006-291X. (Elsevier)Genome deciphering revealed that Mycobacterium tuberculosis encodes a single type II topoisomerase contrary to common bacteria harboring two type II topoisomerases (DNA gyrase and topoisomerase IV). Functions of the M. tuberculosis type II topoisomerase were explored after cloning and expressing the subunits encoding genes in Escherichia coli. M. tuberculosis type II topoisomerase supercoiled relaxed pBR322 with a specific activity close to that of DNA gyrases of common bacteria whereas it exhibited DNA relaxation and formation of cleavable complexes with activities significantly higher than other DNA gyrases. Intermol. passage activity evaluated by the decatenation of kinetoplast DNA was 25-fold lower than that of the topoisomerase IV from Streptococcus pneumoniae, but was markedly higher than that of the E. coli gyrase. Overall, the type II topoisomerase of M. tuberculosis exhibits classical polyvalent activities of DNA gyrase for supercoiling but enhanced relaxation, cleavage, and decatenation activities.
- 3Cole, S. T.; Brosch, R.; Parkhill, J.; Garnier, T.; Churcher, C.; Harris, D.; Gordon, S. V.; Eiglmeier, K.; Gas, S.; Barry, C. E., 3rd; Tekaia, F.; Badcock, K.; Basham, D.; Brown, D.; Chillingworth, T.; Connor, R.; Davies, R.; Devlin, K.; Feltwell, T.; Gentles, S.; Hamlin, N.; Holroyd, S.; Hornsby, T.; Jagels, K.; Krogh, A.; McLean, J.; Moule, S.; Murphy, L.; Oliver, K.; Osborne, J.; Quail, M. A.; Rajandream, M. A.; Rogers, J.; Rutter, S.; Seeger, K.; Skelton, J.; Squares, R.; Squares, S.; Sulston, J. E.; Taylor, K.; Whitehead, S.; Barrell, B. G. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature. 1998, 393, 537– 544, DOI: 10.1038/311593https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXjvV2ksrY%253D&md5=44d23e7674bfeb1a3e30b55abf5268d2Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequenceCole, S. T.; Brosch, R.; Parkhill, J.; Garnier, T.; Churcher, C.; Harris, D.; Gordon, S. V.; Eiglmeier, K.; Gas, S.; Barry, C. E., III; Tekaia, F.; Badcock, K.; Basham, D.; Brown, D.; Chillingworth, T.; Connor, R.; Davies, R.; Devlin, K.; Feltwell, T.; Gentles, S.; Hamlin, N.; Holroyd, S.; Hornsby, T.; Jagels, K.; Krogh, A.; McLean, J.; Moule, S.; Murphy, L.; Oliver, K.; Osborne, J.; Quail, M. A.; Rajandream, M.-A.; Rogers, J.; Rutter, S.; Seeger, K.; Skelton, J.; Squares, R.; Squares, S.; Sulston, J. E.; Taylor, K.; Whitehead, S.; Barrell, B. G.Nature (London) (1998), 393 (6685), 537-544CODEN: NATUAS; ISSN:0028-0836. (Macmillan Magazines)Countless millions of people have died from tuberculosis, a chronic infectious disease caused by the tubercle bacillus. The complete genome sequence of the best-characterized strain of Mycobacterium tuberculosis, H37Rv, was detd. and analyzed in order to improve our understanding of the biol. of this slow-growing pathogen and to help the conception of new prophylactic and therapeutic interventions. The genome comprises 4,411,529 base pairs, contains around 4000 genes, and has a very high G+C content that is reflected in the biased amino acid content of the proteins. M. tuberculosis differs radically from other bacteria in that a very large portion of its coding capacity is devoted to the prodn. of enzymes involved in lipogenesis and lipolysis, and to 2 new families of glycine-rich proteins with a repetitive structure that may represent a source of antigenic variation.
- 4Bush, N. G.; Evans-Roberts, K.; Maxwell, A. DNA Topoisomerases. EcoSal Plus 2015, 6, 1– 34, DOI: 10.1128/ecosalplus.esp-0010-2014There is no corresponding record for this reference.
- 5Asif, M.; Siddiqui, A. A.; Husain, A. Quinolone derivatives as antitubercular drugs. Med. Chem. Res. 2013, 22, 1029– 1042, DOI: 10.1007/s00044-012-0101-3There is no corresponding record for this reference.
- 6Facchinetti, V.; Gomes, C. R. B.; de Souza, M. V. N.; Vasconcelos, T. R. A. Perspectives on the development of novel potentially active quinolones against tuberculosis and cancer. Mini-Rev. Med. Chem. 2012, 12, 866– 874, DOI: 10.2174/138955712800959099There is no corresponding record for this reference.
- 7Kathrotiya, H. G.; Patel, M. P. Synthesis and identification of β–aryloxyquinoline based diversely fluorine substituted N–aryl quinolone derivatives as a new class of antimicrobial, antituberculosis and antioxidant agents. Eur. J. Med. Chem. 2013, 63, 675– 684, DOI: 10.1016/j.ejmech.2013.03.017There is no corresponding record for this reference.
- 8Kim, O. K.; Ohemeng, K.; Barrett, J. F. Advances in DNA gyrase inhibitors. Expert Opin. Investig. Drugs. 2001, 10, 199– 212, DOI: 10.1517/13543784.10.2.199There is no corresponding record for this reference.
- 9Liu, K. L.; Teng, F.; Xiong, L.; Li, X.; Gao, C.; Yu, L. T. Discovery of quinolone derivatives as antimycobacterial agents. RSC Adv. 2021, 11, 24095– 24115, DOI: 10.1039/D0RA09250AThere is no corresponding record for this reference.
- 10Sriram, D.; Aubry, A.; Yogeeswari, P.; Fisher, L. M. Gatifloxacin derivatives: synthesis, antimycobacterial activities, and inhibition of Mycobacterium tuberculosis DNA gyrase. Bioorg. Med. Chem. Lett. 2006, 16, 2982– 2985, DOI: 10.1016/j.bmcl.2006.02.065There is no corresponding record for this reference.
- 11Huang, T. S.; Kunin, C. M.; Lee, S. S. J.; Chen, Y. S.; Tu, H. Z.; Liu, Y. C. Trends in fluoroquinolone resistance of Mycobacterium tuberculosis complex in a Taiwanese medical centre: 1995–2003. J. Antimicrob. Chemother. 2005, 56, 1058– 1062, DOI: 10.1093/jac/dki35311https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlagtLfE&md5=84f10b39609dd05e8a35c5e73b1ddec1Trends in fluoroquinolone resistance of Mycobacterium tuberculosis complex in a Taiwanese medical centre: 1995-2003Huang, Tsi-Shu; Kunin, Calvin M.; Lee, Susan Shin-Jung; Chen, Yao-Shen; Tu, Hui-Zin; Liu, Yung-ChingJournal of Antimicrobial Chemotherapy (2005), 56 (6), 1058-1062CODEN: JACHDX; ISSN:0305-7453. (Oxford University Press)Fluoroquinolones are being used more frequently for the treatment of multidrug-resistant (MDR) strains of Mycobacterium tuberculosis complex (MTB). This study was designed to det. the frequency of the emergence of fluoroquinolone-resistant strains in Taiwan and to assess whether this might be due to use of fluoroquinolones for treatment of patients with MDR or because of increased use of fluoroquinolones in the community for treatment of other infections. The authors also sought to det. whether there might be clonal spread of fluoroquinolone resistance. A total of 3497 clin. isolates of M. tuberculosis complex were obtained during 1995-2003, of which 141 were selected. They consisted of 62 isolates fully susceptible to four first-line drugs, 33 isolates resistant to rifampicin and isoniazid (MDR), and 46 isolates with a variety of any drug resistant patterns other than MDR (combination group). The MICs were detd. for ciprofloxacin, ofloxacin and levofloxacin. Results: An increase in the MIC90 and rates of resistance to ciprofloxacin, ofloxacin and levofloxacin were noted only in the MDR group. The rates were higher among strains isolated between 1998-2003 compared with those obtained between 1995-1997 (rate of resistance, 20% vs. 7.7%; MIC ≥ 4 mg/L vs. 1-2 mg/L). Among the 10 fluoroquinolone-resistant isolates, 5 (50%) possessed mutations other than S95T in the gyrA gene. No gyrB mutation was found in any of the clin. isolates. These findings suggest that fluoroquinolone resistance is the result of treatment of patients with MDR strains rather than from use in the general community in Taiwan. The emergence of fluoroquinolone resistance among MDR strains reinforces the need for routine fluoroquinolone susceptibility testing whenever these drugs might be used.
- 12Lee, A. S.; Tang, L. L.; Lim, I. H.; Wong, S. Y. Characterization of pyrazinamide and ofloxacin resistance among drug resistant Mycobacterium tuberculosis isolates from Singapore. Int. J. Infect. Dis. 2002, 6, 48– 51, DOI: 10.1016/s1201-9712(02)90136-012https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD38zntFWltQ%253D%253D&md5=783085804dfb8525f2d9a17d41c27f55Characterization of pyrazinamide and ofloxacin resistance among drug resistant Mycobacterium tuberculosis isolates from SingaporeLee Ann S G; Tang Lynn L H; Lim Irene H K; Wong Sin YewInternational journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases (2002), 6 (1), 48-51 ISSN:1201-9712.OBJECTIVES: To evaluate rapid molecular approaches for the detection of pyrazinamide (PZA) and ofloxacin resistance, by screening 100 known drug-resistant Mycobacterium tuberculosis isolates. METHODS: Mycobacterium tuberculosis isolates were tested for phenotypic resistance to pyrazinamide and ofloxacin using the BACTEC 460 radiometric method and the E-test, respectively. Mutation screening was done by amplifying the pncA, gyrA, and gyrB genes by the polymerase chain reaction (PCR) and direct automated sequencing. RESULTS: Twelve isolates were PZA-resistant and 8 of 12 (66.7%) isolates had missense mutations or deletions at the pncA gene, suggesting that mutation or deletion at the pncA gene is the major molecular mechanism of PZA resistance among the Singaporean isolates. Using the E-test, 48 isolates were resistant to ofloxacin, with minimum inhibitory concentrations of 4 microg/mL or higher. No mutations were observed at the quinolone resistance-determining region (QRDR) of gyrA in all isolates. At the QRDR of gyrB, mutations were present in 1 of 48 ofloxacin-resistant isolates and 0 of 19 ofloxacin-susceptible isolates. CONCLUSIONS: In Singapore, genotypic analysis of resistance to PZA and ofloxacin is inadequate and should be complemented by conventional methods.
- 13Maruri, F.; Sterling, T. R.; Kaiga, A. W.; Blackman, A.; van der Heijden, Y. F.; Mayer, C.; Cambau, E.; Aubry, A. A systematic review of gyrase mutations associated with fluoroquinolone–resistant Mycobacterium tuberculosis and a proposed gyrase numbering system. J. Antimicrob. Chemother. 2012, 67, 819– 831, DOI: 10.1093/jac/dkr56613https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xkt1ektbc%253D&md5=dcba60297069b8863c4e390e1098dffdA systematic review of gyrase mutations associated with fluoroquinolone-resistant Mycobacterium tuberculosis and a proposed gyrase numbering systemMaruri, Fernanda; Sterling, Timothy R.; Kaiga, Anne W.; Blackman, Amondrea; van der Heijden, Yuri F.; Mayer, Claudine; Cambau, Emmanuelle; Aubry, AlexandraJournal of Antimicrobial Chemotherapy (2012), 67 (4), 819-831CODEN: JACHDX; ISSN:0305-7453. (Oxford University Press)Fluoroquinolone resistance in Mycobacterium tuberculosis has become increasingly important. A review of mutations in DNA gyrase, the fluoroquinolone target, is needed to improve the mol. detection of resistance. The authors performed a systematic review of studies reporting mutations in DNA gyrase genes in clin. M. tuberculosis isolates. From 42 studies that met inclusion criteria, 1220 fluoroquinolone-resistant M. tuberculosis isolates underwent sequencing of the quinolone resistance-detg. region (QRDR) of gyrA; 780 (64%) had mutations. The QRDR of gyrB was sequenced in 534 resistant isolates; 17 (3%) had mutations. Mutations at gyrA codons 90, 91 or 94 were present in 654/1220 (54%) resistant isolates. Four different GyrB numbering systems were reported, resulting in mutation location discrepancies. The authors propose a consensus numbering system. Most fluoroquinolone-resistant M. tuberculosis isolates had mutations in DNA gyrase, but a substantial proportion did not. The proposed consensus numbering system can improve mol. detection of resistance and identification of novel mutations.
- 14Pitaksajjakul, P.; Wongwit, W.; Punprasit, W.; Eampokalap, B.; Peacock, S.; Ramasoota, P. Mutations in the GyrA and GyrB genes of fluoroquinolone–resistant Mycobacterium tuberculosis from TB patients in Thailand. Southeast Asian J. Trop. Med. Public Health 2005, 36, 228– 23714https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFyht78%253D&md5=db80a73a8e5ab2e6f9bb89813c2fac6dMutations in the gyrA and gyrB genes of fluoroquinolone-resistant Mycobacterium tuberculosis from TB patients in ThailandPitaksajjakul, Pannamthip; Wongwit, Waranya; Punprasit, Wantanee; Eampokalap, Boonchuy; Peacock, Sharon; Ramasoota, PongramaSoutheast Asian Journal of Tropical Medicine and Public Health (2005), 36 (Suppl. 4), 228-237CODEN: SJTMAK; ISSN:0125-1562. (SEAMEO-TROPMED Network)Among fluoroquinolone-resistant Mycobacterium tuberculosis (FQr-MTB) isolates, mutation at positions 90, 91, and 94 in gyrA gene and at positions 495, 516, and 533 in gyrB gene have been frequently reported. In this study, 35 isolates of FQr-MTB were collected from Siriraj Hospital and Chest Disease Institute. The quinolone-resistance-detg. regions (QRDR) of gyrA and gyrB genes in all 35 FQr-MTB isolates and from the H37Ra MTB strain were amplified using polymerase chain reaction (PCR). DNA-sequencing and single-strand conformation polymorphism (SSCP) were further utilized for characterization of the mutations in the QRDR of gyrA and gyrB genes and mutation screening, resp. From DNA-sequencing, 21 of 35 (60%) exhibited single-point mutations in different positions, at Ala90Val, Ser91Pro, and Asp94(Gly/Ala/His/Asn); and one novel mutation position at Gly88Cys in the gyrA gene and Asp495Asn in the gyrB gene. These positions were previously frequently reported to be responsible for FQr-MTB. The other 14 FQr-MTB isolates (40%) had no mutation. This study is the first report of mutation occurring only in the QRDR of the gyrB gene, without prior mutation in the gyrA QRDR among FQr-MTB isolates. By SSCP anal. for screening of the mutant FQr-MTB, the SSCP patterns of mutated FQr-MTB isolates were clearly differentiated from the SSCP patterns of FQs-MTB.
- 15Pitaksajjakul, P.; Worakhunpiset, S.; Chaiprasert, A.; Boonyasopun, J.; Ramasoota, P. GyrA and GyrB mutations in ofloxacin–resistant Mycobacterium tuberculosis clinical isolates in Thailand. Southeast Asian J. Trop. Med. Public Health. 2011, 42, 1163– 116715https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlansbfI&md5=c0a8d3f3b0538afe15fcf095a91c32c2gyrA and gyrB mutations in ofloxacin-resistant Mycobacterium tuberculosis clinical isolates in ThailandPitaksajjakul, Pannamthip; Worakhunpiset, Suwalee; Chaiprasert, Angkana; Boonyasopun, Jirakarn; Ramasoota, PongramaSoutheast Asian Journal of Tropical Medicine and Public Health (2011), 42 (5), 1163-1167CODEN: SJTMAK; ISSN:0125-1562. (SEAMEO-TROPMED Network)In order to identify mutations in gyrA and gyrB genes in 92 ofloxacin-resistant Mycobacterium tuberculosis (OFXr-MTB) clin. isolates collected from Siriraj Hospital, Mahidol University and Chest Disease Institute, Thailand. The quinolone resistance-detg. regions (QRDR) of gyrA and gyrB in all 92 OFXr-MTB isolates were amplified using polymerase chain reaction and sequenced. There were 70 isolates with point mutations assocd. with ofloxacin resistance. In gyrA QRDR, 69 isolates had mutations in gyrA Gly88 (Ala/(75), Ala90 (Val), Ser91 (Pro) and Asp94 (Gly/Ala/His/Asn), the latter being the most common (42%). Only one isolate was found with mutation at position Asp495 (Asn). The other 22 isolates had no mutations in both gyrA and gyrB QRDR. Thus, point mutations in gyrA and gyrB QRDR were responsible for OFXr-MTB clin. isolates in Thailand.
- 16Soudani, A.; Hadjfredj, S.; Zribi, M.; Messaoud, T.; Masmoudi, A.; Majed, B.; Fendri, C. First report of molecular characterization of fluoroquinolone–resistant Mycobacterium tuberculosis isolates from a Tunisian hospital. Clin. Microbiol. Infect. 2010, 16, 1454– 1457, DOI: 10.1111/j.1469-0691.2010.03087.xThere is no corresponding record for this reference.
- 17Medapi, B.; Renuka, J.; Saxena, S.; Sridevi, J. P.; Medishetti, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Design and synthesis of novel quinoline–aminopiperidine hybrid analogues as Mycobacterium tuberculosis DNA gyrase B inhibitors. Bioorg. Med. Chem. 2015, 23, 2062– 2078, DOI: 10.1016/j.bmc.2015.03.004There is no corresponding record for this reference.
- 18Medapi, B.; Suryadevara, P.; Renuka, J.; Sridevi, J. P.; Yogeeswari, P.; Sriram, D. 4–Aminoquinoline derivatives as novel Mycobacterium tuberculosis GyrB inhibitors: Structural optimization, synthesis and biological evaluation. Eur. J. Med. Chem. 2015, 103, 1– 16, DOI: 10.1016/j.ejmech.2015.06.032There is no corresponding record for this reference.
- 19Jeankumar, V. U.; Reshma, R. S.; Vats, R.; Janupally, R.; Saxena, S.; Yogeeswari, P.; Sriram, D. Engineering another class of anti–tubercular lead: Hit to lead optimization of an intriguing class of gyrase ATPase inhibitors. Eur. J. Med. Chem. 2016, 122, 216– 231, DOI: 10.1016/j.ejmech.2016.06.042There is no corresponding record for this reference.
- 20Jeankumar, V. U.; Renuka, J.; Pulla, V. K.; Soni, V.; Sridevi, J. P.; Suryadevara, P.; Shravan, M.; Medishetti, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Development of novel N–linked aminopiperidine–based mycobacterial DNA gyrase B inhibitors: scaffold hopping from known antibacterial leads. Int. J. Antimicrob Agents. 2014, 43, 269– 278, DOI: 10.1016/j.ijantimicag.2013.12.006There is no corresponding record for this reference.
- 21Jeankumar, V. U.; Kotagiri, S.; Janupally, R.; Suryadevara, P.; Sridevi, J. P.; Medishetti, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Exploring the gyrase ATPase domain for tailoring newer anti–tubercular drugs: hit to lead optimization of a novel class of thiazole inhibitors. Bioorg. Med. Chem. 2015, 23, 588– 601, DOI: 10.1016/j.bmc.2014.12.001There is no corresponding record for this reference.
- 22Omar, M. A.; Masaret, G. S.; Abbas, E. M. H.; Abdel Aziz, M. M.; Harras, M. F.; Farghaly, T. A. Novel anti–tubercular and antibacterial based benzosuberone–thiazole moieties: synthesis, molecular docking analysis, DNA gyrase supercoiling and ATPase activity. Bioorg Chem. 2020, 104, 104316 DOI: 10.1016/j.bioorg.2020.104316There is no corresponding record for this reference.
- 23Jeankumar, V. U.; Renuka, J.; Kotagiri, S.; Saxena, S.; Kakan, S. S.; Sridevi, J. P.; Yellanki, S.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Gyrase ATPase domain as an antitubercular drug discovery platform: structure–based design and lead optimization of nitrothiazolyl carboxamide analogues. Chem. Med. Chem. 2014, 9, 1850– 1859, DOI: 10.1002/cmdc.201402035There is no corresponding record for this reference.
- 24P, S. H.; Solapure, S.; Mukherjee, K.; Nandi, V.; Waterson, D.; Shandil, R.; Balganesh, M.; Sambandamurthy, V. K.; Raichurkar, A. K.; Deshpande, A.; Ghosh, A.; Awasthy, D.; Shanbhag, G.; Sheikh, G.; McMiken, H.; Puttur, J.; Reddy, J.; Werngren, J.; Read, J.; Kumar, M.; M, R.; Chinnapattu, M.; Madhavapeddi, P.; Manjrekar, P.; Basu, R.; Gaonkar, S.; Sharma, S.; Hoffner, S.; Humnabadkar, V.; Subbulakshmi, V.; Panduga, V. Optimization of pyrrolamides as mycobacterial GyrB ATPase inhibitors: structure–activity relationship and in vivo efficacy in a mouse model of tuberculosis. Antimicrob. Agents Chemother. 2014, 58, 61– 70, DOI: 10.1128/AAC.01751-13There is no corresponding record for this reference.
- 25Reddy, K. I.; Srihari, K.; Renuka, J.; Sree, K. S.; Chuppala, A.; Jeankumar, V. U.; Sridevi, J. P.; Babu, K. S.; Yogeeswari, P.; Sriram, D. An efficient synthesis and biological screening of benzofuran and benzo[d]isothiazole derivatives for Mycobacterium tuberculosis DNA GyrB inhibition. Bioorg. Med. Chem. 2014, 22, 6552– 6563, DOI: 10.1016/j.bmc.2014.10.01625https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVSls7fO&md5=35443a5527173e382e720eec298aa5d6An efficient synthesis and biological screening of benzofuran and benzo[d]isothiazole derivatives for Mycobacterium tuberculosis DNA GyrB inhibitionReddy, Kummetha Indrasena; Srihari, Konduri; Renuka, Janupally; Sree, Komanduri Shruthi; Chuppala, Aruna; Jeankumar, Variam Ullas; Sridevi, Jonnalagadda Padma; Babu, Kondra Sudhakar; Yogeeswari, Perumal; Sriram, DharmarajanBioorganic & Medicinal Chemistry (2014), 22 (23), 6552-6563CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A series of twenty eight mols. of Et 5-(piperazin-1-yl)benzofuran-2-carboxylate and 3-(piperazin-1-yl)benzo[d]isothiazole were designed by mol. hybridization of thiazole aminopiperidine core and carbamide side chain in eight steps and were screened for their in vitro Mycobacterium smegmatis (MS) GyrB ATPase assay, Mycobacterium tuberculosis (MTB) DNA gyrase super coiling assay, antitubercular activity, cytotoxicity and protein-inhibitor interaction assay through differential scanning fluorometry. Also the orientation and the ligand-protein interactions of the top hit mols. with MS DNA gyrase B subunit active site were investigated applying extra precision mode (XP) of Glide. Among the compds. studied, 4-(benzo[d]isothiazol-3-yl)-N-(4-chlorophenyl)piperazine-1-carboxamide (26) was found to be the most promising inhibitor with an MS GyrB IC50 of 1.77 ± 0.23 μM, 0.42 ± 0.23 against MTB DNA gyrase, MTB MIC of 3.64 μM, and was not cytotoxic in eukaryotic cells at 100 μM. Moreover the interaction of protein-ligand complex was stable and showed a pos. shift of 3.5 °C in differential scanning fluorimetric evaluations.
- 26Renuka, J.; Reddy, K. I.; Srihari, K.; Jeankumar, V. U.; Shravan, M.; Sridevi, J. P.; Yogeeswari, P.; Babu, K. S.; Sriram, D. Design, synthesis, biological evaluation of substituted benzofurans as DNA gyrase B inhibitors of Mycobacterium tuberculosis. Bioorg. Med. Chem. 2014, 22, 4924– 4934, DOI: 10.1016/j.bmc.2014.06.04126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlGmu7bM&md5=4e84e0c1d803e5579a10314a199872d4Design, synthesis, biological evaluation of substituted benzofurans as DNA gyrase B inhibitors of Mycobacterium tuberculosisRenuka, Janupally; Reddy, Kummetha Indrasena; Srihari, Konduri; Jeankumar, Variam Ullas; Shravan, Morla; Sridevi, Jonnalagadda Padma; Yogeeswari, Perumal; Babu, Kondra Sudhakar; Sriram, DharmarajanBioorganic & Medicinal Chemistry (2014), 22 (17), 4924-4934CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)DNA gyrase of Mycobacterium tuberculosis (MTB) is a type II topoisomerase and is a well established and validated target for the development of novel therapeutics. The authors present the discovery and optimization of an Et 5-(piperazin-1-yl) benzofuran-2-carboxylate series of mycobacterial DNA gyrase B inhibitors, selected from the Birla Institute of Technol. and Science (BITS) database chem. library of about 3000 mols. When these compds. were tested for their biol. activity, the compd. 22 emerged as the most active potent lead with an IC50 of 3.2 ± 0.15 μM against Mycobacterium smegmatis DNA gyrase B enzyme and 0.81 ± 0.24 μM in MTB supercoiling activity. The binding of this most active compd. to the DNA gyrase B enzyme and its thermal stability was further characterized using a differential scanning fluorimetry method.
- 27Jeankumar, V. U.; Saxena, S.; Vats, R.; Reshma, R. S.; Janupally, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Structure–guided discovery of antitubercular agents that target the gyrase ATPase domain. Chem. Med. Chem. 2016, 11, 539– 548, DOI: 10.1002/cmdc.201500556There is no corresponding record for this reference.
- 28Saxena, S.; Samala, G.; Renuka, J.; Sridevi, J. P.; Yogeeswari, P.; Sriram, D. Development of 2–amino–5–phenylthiophene–3–carboxamide derivatives as novel inhibitors of Mycobacterium tuberculosis DNA GyrB domain. Bioorg. Med. Chem. 2015, 23, 1402– 1412, DOI: 10.1016/j.bmc.2015.02.032There is no corresponding record for this reference.
- 29Saxena, S.; Renuka, J.; Yogeeswari, P.; Sriram, D. Discovery of novel mycobacterial DNA gyrase B inhibitors: In silico and in vitro biological evaluation. Mol. Inform. 2014, 33, 597– 609, DOI: 10.1002/minf.201400058There is no corresponding record for this reference.
- 30McGarry, D. H.; Cooper, I. R.; Walker, R.; Warrilow, C. E.; Pichowicz, M.; Ratcliffe, A. J.; Salisbury, A. M.; Savage, V. J.; Moyo, E.; Maclean, J.; Smith, A.; Charrier, C.; Stokes, N. R.; Lindsay, D. M.; Kerr, W. J. Design, synthesis and antibacterial properties of pyrimido[4,5–b]indol–8–amine inhibitors of DNA gyrase. Bioorg. Med. Chem. Lett. 2018, 28, 2998– 3003, DOI: 10.1016/j.bmcl.2018.05.049There is no corresponding record for this reference.
- 31Henderson, S. R.; Stevenson, C. E. M.; Malone, B.; Zholnerovych, Y.; Mitchenall, L. A.; Pichowicz, M.; McGarry, D. H.; Cooper, I. R.; Charrier, C.; Salisbury, A. M.; Lawson, D. M.; Maxwell, A. Structural and mechanistic analysis of ATPase inhibitors targeting mycobacterial DNA gyrase. J. Antimicrob. Chemother. 2020, 75, 2835– 2842, DOI: 10.1093/jac/dkaa28631https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitl2jtrs%253D&md5=78dddb492907ec5f0215e19f55655990Structural and mechanistic analysis of ATPase inhibitors targeting mycobacterial DNA gyraseHenderson, Sara R.; Stevenson, Clare E. M.; Malone, Brandon; Zholnerovych, Yelyzaveta; Mitchenall, Lesley A.; Pichowicz, Mark; McGarry, David H.; Cooper, Ian R.; Charrier, Cedric; Salisbury, Anne-Marie; Lawson, David M.; Maxwell, AnthonyJournal of Antimicrobial Chemotherapy (2020), 75 (10), 2835-2842CODEN: JACHDX; ISSN:1460-2091. (Oxford University Press)To evaluate the efficacy of two novel compds. against mycobacteria and det. the mol. basis of their action on DNA gyrase using structural and mechanistic approaches. Redx03863 and Redx04739 were tested in antibacterial assays, and also against their target, DNA gyrase, using DNA supercoiling and ATPase assays. X-ray crystallog. was used to det. the structure of the gyrase B protein ATPase sub-domain from Mycobacterium smegmatis complexed with the aminocoumarin drug novobiocin, and structures of the same domain from Mycobacterium thermoresistibile complexed with novobiocin, and also with Redx03863. Both compds., Redx03863 and Redx04739, were active against selected Gram-pos. and Gram-neg. species, with Redx03863 being the more potent, and Redx04739 showing selectivity against M. smegmatis. Both compds. were potent inhibitors of the supercoiling and ATPase reactions of DNA gyrase, but did not appreciably affect the ATP-independent relaxation reaction. The structure of Redx03863 bound to the gyrase B protein ATPase sub-domain from M. thermoresistibile shows that it binds at a site adjacent to the ATP- and novobiocin-binding sites. We found that most of the mutations that we made in the Redx03863-binding pocket, based on the structure, rendered gyrase inactive. Redx03863 and Redx04739 inhibit gyrase by preventing the binding of ATP. The fact that the Redx03863-binding pocket is distinct from that of novobiocin, coupled with the lack of activity of resistant mutants, suggests that such compds. could have potential to be further exploited as antibiotics.
- 32Kamsri, B.; Pakamwong, B.; Thongdee, P.; Phusi, N.; Kamsri, P.; Punkvang, A.; Ketrat, S.; Saparpakorn, P.; Hannongbua, S.; Sangswan, J.; Suttisintong, K.; Sureram, S.; Kittakoop, P.; Hongmanee, P.; Santanirand, P.; Leanpolchareanchai, J.; Goudar, K. E.; Spencer, J.; Mulholland, A. J.; Pungpo, P. Bioisosteric Design Identifies Inhibitors of Mycobacterium tuberculosis DNA Gyrase ATPase Activity. J. Chem. Inf Model. 2023, 63, 2707– 2718, DOI: 10.1021/acs.jcim.2c01376There is no corresponding record for this reference.
- 33Pakamwong, B.; Thongdee, P.; Kamsri, B.; Phusi, N.; Kamsri, P.; Punkvang, A.; Ketrat, S.; Saparpakorn, P.; Hannongbua, S.; Ariyachaokun, K.; Suttisintong, K.; Sureram, S.; Kittakoop, P.; Hongmanee, P.; Santanirand, P.; Spencer, J.; Mulholland, A. J.; Pungpo, P. Identification of Potent DNA Gyrase Inhibitors Active against Mycobacterium tuberculosis. J. Chem. Inf Model. 2022, 62, 1680– 1690, DOI: 10.1021/acs.jcim.1c0139033https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XosFeqsbc%253D&md5=60557f6d45a2d8df0a071a3d37afe03fIdentification of Potent DNA Gyrase Inhibitors Active against Mycobacterium tuberculosisPakamwong, Bongkochawan; Thongdee, Paptawan; Kamsri, Bundit; Phusi, Naruedon; Kamsri, Pharit; Punkvang, Auradee; Ketrat, Sombat; Saparpakorn, Patchreenart; Hannongbua, Supa; Ariyachaokun, Kanchiyaphat; Suttisintong, Khomson; Sureram, Sanya; Kittakoop, Prasat; Hongmanee, Poonpilas; Santanirand, Pitak; Spencer, James; Mulholland, Adrian J.; Pungpo, PornpanJournal of Chemical Information and Modeling (2022), 62 (7), 1680-1690CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Mycobacterium tuberculosis DNA gyrase manipulates the DNA topol. using controlled breakage and religation of DNA driven by ATP hydrolysis. DNA gyrase has been validated as the enzyme target of fluoroquinolones (FQs), second-line antibiotics used for the treatment of multidrug-resistant tuberculosis. Mutations around the DNA gyrase DNA-binding site result in the emergence of FQ resistance in M. tuberculosis; inhibition of DNA gyrase ATPase activity is one strategy to overcome this. Here, virtual screening, subsequently validated by biol. assays, was applied to select candidate inhibitors of the M. tuberculosis DNA gyrase ATPase activity from the Specs compd. library (www.specs.net). Thirty compds. were identified and selected as hits for in vitro biol. assays, of which two compds., G24 and G26, inhibited the growth of M. tuberculosis H37Rv with a minimal inhibitory concn. of 12.5 μg/mL. The two compds. inhibited DNA gyrase ATPase activity with IC50 values of 2.69 and 2.46 μM, resp., suggesting this to be the likely basis of their antitubercular activity. Models of complexes of compds. I and II bound to the M. tuberculosis DNA gyrase ATP-binding site, generated by mol. dynamics simulations followed by pharmacophore mapping anal., showed hydrophobic interactions of inhibitor hydrophobic headgroups and electrostatic and hydrogen bond interactions of the polar tails, which are likely to be important for their inhibition. Decreasing compd. lipophilicity by increasing the polarity of these tails then presents a likely route to improving the soly. and activity. Thus, compds. I and II provide attractive starting templates for the optimization of antitubercular agents that act by targeting DNA gyrase.
- 34Maxwell, A. The interaction between coumarin drugs and DNA gyrase. Mol. Microbiol. 1993, 9, 681– 686, DOI: 10.1111/j.1365-2958.1993.tb01728.x34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXmsVCjsr8%253D&md5=936de9ec8f1f199de584ec2cedb941b6The interaction between coumarin drugs and DNA gyraseMaxwell, AnthonyMolecular Microbiology (1993), 9 (4), 681-6CODEN: MOMIEE; ISSN:0950-382X.A review with 49 refs. The coumarin group of antibiotics have as their target bacterial DNA gyrase (I). The drugs bind to the B subunit of I and inhibit DNA supercoiling by blocking the ATPase activity. Recent data show that the binding site for the drugs lies within the N-terminal part of the B protein, and individual amino acids involved in coumarin interaction are being identified. The mode of inhibition of the I ATPase reaction by coumarins is unlikely to be simple competitive inhibition, and the drugs may act by stabilizing a conformation of the enzyme with low affinity for ATP.
- 35Grossman, S.; Fishwick, C. W. G.; McPhillie, M. J. Developments in non–intercalating bacterial topoisomerase inhibitors: allosteric and ATPase inhibitors of DNA gyrase and topoisomerase IV. Pharm. 2023, 16, 261, DOI: 10.3390/ph16020261There is no corresponding record for this reference.
- 36Kar, S.; Roy, K. How far can virtual screening take us in drug discovery?. Expert Opin Drug Discovery 2013, 8, 245– 261, DOI: 10.1517/17460441.2013.761204There is no corresponding record for this reference.
- 37Lionta, E.; Spyrou, G.; Vassilatis, D. K.; Cournia, Z. Structure–based virtual screening for drug discovery: principles, applications and recent advances. Curr. Top Med. Chem. 2014, 14, 1923– 1938, DOI: 10.2174/156802661466614092912444537https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVGgt7fJ&md5=0550a396dd011adf52699b091548345bStructure-Based Virtual Screening for Drug Discovery: Principles, Applications and Recent AdvancesLionta, Evanthia; Spyrou, George; Vassilatis, Demetrios K.; Cournia, ZoeCurrent Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2014), 14 (16), 1923-1938CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)A review. Structure-based drug discovery (SBDD) is becoming an essential tool in assisting fast and cost-efficient lead discovery and optimization. The application of rational, structure-based drug design is proven to be more efficient than the traditional way of drug discovery since it aims to understand the mol. basis of a disease and utilizes the knowledge of the three-dimensional structure of the biol. target in the process. In this review, we focus on the principles and applications of Virtual Screening (VS) within the context of SBDD and examine different procedures ranging from the initial stages of the process that include receptor and library pre-processing, to docking, scoring and post-processing of top scoring hits. Recent improvements in structure-based virtual screening (SBVS) efficiency through ensemble docking, induced fit and consensus docking are also discussed. The review highlights advances in the field within the framework of several success studies that have led to nM inhibition directly from VS and provides recent trends in library design as well as discusses limitations of the method. Applications of SBVS in the design of substrates for engineered proteins that enable the discovery of new metabolic and signal transduction pathways and the design of inhibitors of multifunctional proteins are also reviewed. Finally, we contribute two promising VS protocols recently developed by us that aim to increase inhibitor selectivity. In the first protocol, we describe the discovery of micromolar inhibitors through SBVS designed to inhibit the mutant H1047R PI3Kα kinase. Second, we discuss a strategy for the identification of selective binders for the RXRα nuclear receptor. In this protocol, a set of target structures is constructed for ensemble docking based on binding site shape characterization and clustering, aiming to enhance the hit rate of selective inhibitors for the desired protein target through the SBVS process.
- 38Oliveira, T. A. D.; Silva, M. P. D.; Maia, E. H. B.; Silva, A. M. D.; Taranto, A. G. Virtual screening algorithms in drug discovery: a review focused on machine and deep learning methods. Drugs Drug Candidates. 2023, 2, 311– 334, DOI: 10.3390/ddc2020017There is no corresponding record for this reference.
- 39Thongdee, P.; Hanwarinroj, C.; Pakamwong, B.; Kamsri, P.; Punkvang, A.; Leanpolchareanchai, J.; Ketrat, S.; Saparpakorn, P.; Hannongbua, S.; Ariyachaokun, K.; Suttisintong, K.; Sureram, S.; Kittakoop, P.; Hongmanee, P.; Santanirand, P.; Mukamolova, G. V.; Blood, R. A.; Takebayashi, Y.; Spencer, J.; Mulholland, A. J.; Pungpo, P. Virtual screening identifies novel and potent inhibitors of Mycobacterium tuberculosis PknB with antibacterial activity. J. Chem. Inf Model. 2022, 62, 6508– 6518, DOI: 10.1021/acs.jcim.2c00531There is no corresponding record for this reference.
- 40Arica Sosa, A.; Alcántara, R.; Jiménez Avalos, G.; Zimic, M.; Milón, P.; Quiliano, M. Identifying RO9021 as a potential inhibitor of PknG from Mycobacterium tuberculosis: combinative computational and in vitro studies. ACS Omega. 2022, 7, 20204– 20218, DOI: 10.1021/acsomega.2c02093There is no corresponding record for this reference.
- 4141. Sheikh, B. A.; Bhat, B. A.; Rizvi, M. A.; Ahmad, Z.; Almilaibary, A.; Alkhanani, M.; Mir, M. A. Computational studies to identify potential inhibitors targeting the DprE1 protein in Mycobacterium tuberculosis. Int. J. Pharm. Investig. 2023, 13, 129– 138, DOI: 10.5530/223097131750There is no corresponding record for this reference.
- 42Verma, A.; Kumar, V.; Naik, B.; Khan, J. M.; Singh, P.; Saris, P. E. J.; Gupta, S. Screening and molecular dynamics simulation of compounds inhibiting MurB enzyme of drug–resistant Mycobacterium tuberculosis: An in–silico approach. Saudi J. Biol. Sci. 2023, 30, 103730 DOI: 10.1016/j.sjbs.2023.103730There is no corresponding record for this reference.
- 43Kumar, G. S.; Dubey, A.; Panda, S. P.; Alawi, M. M.; Sindi, A. A.; Azhar, E. I.; Dwivedi, V. D.; Agrawal, S. Repurposing of antibacterial compounds for suppression of Mycobacterium tuberculosis dormancy reactivation by targeting resuscitation–promoting factors B. J. Biomol. Struct. Dyn. 2023, 1– 13, DOI: 10.1080/07391102.2023.2245059There is no corresponding record for this reference.
- 44Johannsen, S.; Gierse, R. M.; Olshanova, A.; Smerznak, E.; Laggner, C.; Eschweiler, L.; Adeli, Z.; Hamid, R.; Alhayek, A.; Reiling, N.; Haupenthal, J.; Hirsch, A. K. H. Not every hit–identification technique works on 1–deoxy–d–xylulose 5–phosphate synthase (DXPS): making the most of a virtual screening campaign. ChemMedChem 2023, 18, e202200590 DOI: 10.1002/cmdc.202200590There is no corresponding record for this reference.
- 45Damera, T.; Kondaparthi, V.; Bingi, M.; Mustyala, K. K.; Malkhed, V. Identification of novel scaffolds to inhibit Mycobacterium tuberculosis PimA protein–A computational approach. J. Cell Biochem. 2023, 124, 836– 848, DOI: 10.1002/jcb.30412There is no corresponding record for this reference.
- 46Varon, H. A.; Santos, P.; Lopez-Vallejo, F.; Soto, C. Y. Novel scaffolds targeting Mycobacterium tuberculosis plasma membrane Ca2+ transporter CtpF by structure–based strategy. Bioorg. Chem. 2023, 138, 106648 DOI: 10.1016/j.bioorg.2023.106648There is no corresponding record for this reference.
- 47Daina, A.; Michielin, O.; Zoete, V. SwissADME: a free web tool to evaluate pharmacokinetics, drug–likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717, DOI: 10.1038/srep4271747https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czisFSrtg%253D%253D&md5=9715b8cb8a34b17c4c73ff69a5a8cc50SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small moleculesDaina Antoine; Michielin Olivier; Zoete Vincent; Michielin Olivier; Michielin OlivierScientific reports (2017), 7 (), 42717 ISSN:.To be effective as a drug, a potent molecule must reach its target in the body in sufficient concentration, and stay there in a bioactive form long enough for the expected biologic events to occur. Drug development involves assessment of absorption, distribution, metabolism and excretion (ADME) increasingly earlier in the discovery process, at a stage when considered compounds are numerous but access to the physical samples is limited. In that context, computer models constitute valid alternatives to experiments. Here, we present the new SwissADME web tool that gives free access to a pool of fast yet robust predictive models for physicochemical properties, pharmacokinetics, drug-likeness and medicinal chemistry friendliness, among which in-house proficient methods such as the BOILED-Egg, iLOGP and Bioavailability Radar. Easy efficient input and interpretation are ensured thanks to a user-friendly interface through the login-free website http://www.swissadme.ch. Specialists, but also nonexpert in cheminformatics or computational chemistry can predict rapidly key parameters for a collection of molecules to support their drug discovery endeavours.
- 48Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B. A.; Thiessen, P. A.; Yu, B.; Zaslavsky, L.; Zhang, J.; Bolton, E. E. PubChem in 2021: new data content and improved web interfaces. Nucleic Acids Res. 2021, 49, D1388– D1395, DOI: 10.1093/nar/gkaa97148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXntFCit7Y%253D&md5=5bbf4c2b84fc02bbb043cbcc75d4b948PubChem in 2021: new data content and improved web interfacesKim, Sunghwan; Chen, Jie; Cheng, Tiejun; Gindulyte, Asta; He, Jia; He, Siqian; Li, Qingliang; Shoemaker, Benjamin A.; Thiessen, Paul A.; Yu, Bo; Zaslavsky, Leonid; Zhang, Jian; Bolton, Evan E.Nucleic Acids Research (2021), 49 (D1), D1388-D1395CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)PubChem is a popular chem. information resource that serves the scientific community as well as the general public, with millions of unique users per mo. In the past 2 yr, PubChem made substantial improvements. Data from >100 new data sources were added to PubChem, including chem.-literature links from Thieme Chem., chem. and phys. property links from SpringerMaterials, and patent links from the World Intellectual Properties Organization (WIPO). PubChem's homepage and individual record pages were updated to help users find desired information faster. This update involved a data model change for the data objects used by these pages as well as by programmatic users. Several new services were introduced, including the PubChem Periodic Table and Element pages, Pathway pages, and Knowledge panels. Addnl., in response to the coronavirus disease 2019 (COVID-19) outbreak, PubChem created a special data collection that contains PubChem data related to COVID-19 and the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- 49Backman, T. W. H.; Cao, Y.; Girke, T. ChemMine tools: an online service for analyzing and clustering small molecules. Nucleic Acids Res. 2011, 39, W486– W491, DOI: 10.1093/nar/gkr32049https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXosVOntbk%253D&md5=99a8d31fed2f8e47c0c81ffd192078a7ChemMine tools: an online service for analyzing and clustering small moleculesBackman, Tyler W. H.; Cao, Yiqun; Girke, ThomasNucleic Acids Research (2011), 39 (Web Server), W486-W491CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChemMine Tools is an online service for small mol. data anal. It provides a web interface to a set of chemoinformatics and data mining tools that are useful for various anal. routines performed in chem. genomics and drug discovery. The service also offers programmable access options via the R library ChemmineR. The primary functionalities of ChemMine Tools fall into five major application areas: data visualization, structure comparisons, similarity searching, compd. clustering and prediction of chem. properties. First, users can upload compd. data sets to the online Compd. Workbench. Numerous utilities are provided for compd. viewing, structure drawing and format interconversion. Second, pairwise structural similarities among compds. can be quantified. Third, interfaces to ultra-fast structure similarity search algorithms are available to efficiently mine the chem. space in the public domain. These include fingerprint and embedding/indexing algorithms. Fourth, the service includes a Clustering Toolbox that integrates chemoinformatic algorithms with data mining utilities to enable systematic structure and activity based analyses of custom compd. sets. Fifth, physicochem. property descriptors of custom compd. sets can be calcd. These descriptors are important for assessing the bioactivity profile of compds. in silico and quant. structure-activity relationship (QSAR) analyses. ChemMine Tools is available at: http://chemmine.ucr.edu.
- 50Studier, F. W.; Moffatt, B. A. Use of bacteriophage T7 RNA polymerase to direct selective high–level expression of cloned genes. J. Mol. Biol. 1986, 189, 113– 130, DOI: 10.1016/0022-2836(86)90385-250https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XktlKrsr4%253D&md5=3219300bc2f640fe9830c7518eb99bfcUse of bacteriophage T7 RNA polymerase to direct selective high-level expression of cloned genesStudier, F. William; Moffatt, Barbara A.Journal of Molecular Biology (1986), 189 (1), 113-30CODEN: JMOBAK; ISSN:0022-2836.A gene expression system based on phage T7 RNA polymerase [9014-24-8] was developed. T7 RNA polymerase is highly selective for its own promoters, which do not occur naturally in Escherichia coli. A relatively small amt. of T7 RNA polymerase provided from a cloned copy of T7 gene 1 is sufficient to direct high-level transcription from a T7 promoter in a multicopy plasmid. Such transcription can proceed several times around the plasmid without terminating and can be so active that transcription by E. coli RNA polymerase is greatly decreased. When a cleavage site for RNase III is introduced, discrete RNAs of plasmid length can accumulate. The natural transcription terminator from T7 DNA also works effectively in the plasmid. Both the rate of synthesis and the accumulation of RNA directed by T7 RNA polymerase can reach levels comparable with those for rRNAs in a normal cell. These high levels of accumulation suggest that the RNAs are relatively stable, perhaps in part because their great length and(or) stem-and-loop structures at their 3' ends help to protect them against exonucleolytic degrdn. Apparently, a specific mRNA produced by T7 RNA polymerase can rapidly sat. the translational machinery of E. coli, so that the rate of protein synthesis from such an mRNA will depend primarily on the efficiency of its translation. When the mRNA is efficiently translated, a target protein can accumulate to >50% of the total cell protein in ≤3 h. Two ways were used to deliver active T7 RNA polymerase to the cell: (1) infection by a λ deriv. that carried gene 1; or (2) induction of a chromosomal copy of gene 1 under control of the lacUV5 promoter. When gene 1 is delivered by infection, very toxic target genes can be maintained silently in the cell until T7 RNA polymerase is introduced, when they rapidly become expressed at high levels. When gene 1 is resident in the chromosome, even the very low basal levels of T7 RNA polymerase present in the uninduced cell can prevent the establishment of plasmids carrying toxic target genes, or make the plasmid unstable. But if the target plasmid can be maintained, induction of chromosomal gene 1 can be a convenient way to produce large amts. of target RNA and(or) protein. T7 RNA polymerase seems to be capable of transcribing almost any DNA linked to a T7 promoter, so the T7 expression system should be capable of transcribing almost any gene or its complement in E. coli. Comparable T7 expression systems can be developed in other types of cells.
- 51Laemmli, U. K. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970, 227, 680– 685, DOI: 10.1038/227680a051https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXlsFags7s%253D&md5=fff3e668784b8bb3669f854be60a216bCleavage of structural proteins during the assembly of the head of bacteriophage T4Laemmli, U. K.Nature (London, United Kingdom) (1970), 227 (5259), 680-685CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Using an improved method of polyacrylamide gel electrophoresis based on the capability of SDS to break down proteins into their individual polypeptide chains, many previously unknown proteins have been found in bacteriophage T4 and some of these have been identified with specific gene products. Four major components of the head are cleaved during the process of assembly, apparently after the precursor proteins have assembled into some large intermediate structure.
- 52Wang, X. D.; Meng, M. X.; Gao, L. B.; Liu, T.; Xu, Q.; Zeng, S. Permeation of astilbin and taxifolin in Caco–2 cell and their effects on the P–gp. Int. J. Pharm. 2009, 378, 1– 8, DOI: 10.1016/j.ijpharm.2009.05.02252https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXovFKqtLw%253D&md5=59d129ebdfbe70949d731fdfce20abfdPermeation of astilbin and taxifolin in Caco-2 cell and their effects on the P-gpWang, Xiao-dan; Meng, Min-xin; Gao, Ling-bo; Liu, Ting; Xu, Qiang; Zeng, SuInternational Journal of Pharmaceutics (2009), 378 (1-2), 1-8CODEN: IJPHDE; ISSN:0378-5173. (Elsevier B.V.)This study was designed to understand the transport profiles of astilbin and taxifolin in Caco-2 cell model and their effects on the function and expression of P-glycoprotein. The transport studies were examd. using Caco-2 cells cultured on Transwell inserts. Their effects on the function and expression of P-glycoprotein were detected using Western Blot and RT-PCR. The transport was concn. and temp. dependent. The apparent permeability (P app) of these two compds. in the secretory direction was larger than that in the absorptive direction in the concn. range of 10-1000 μM. Those compds. had no effects on the P-glycoprotein-mediated transport of Rhodamine 123. Caco-2 cells exposed to astilbin or taxifolin for 36 h exhibited higher P-glycoprotein activity through up-regulating P-glycoprotein expression at protein and mRNA levels. These results indicated that P-glycoprotein and Multidrug Resistance Protein 2 might play important roles in limiting the bioavailability of those compds. Drugs which are the inhibitors of P-glycoprotein or Multidrug Resistance Protein 2 may increase the oral bioavailability of astilbin or taxifolin and the possibility of unwanted drug-food interactions. The increased expression of P-glycoprotein in Caco-2 cells may serve as an adaptation and defense mechanism in limiting the entry of xenobiotics into the body.
- 53Faralli, A.; Shekarforoush, E.; Ajalloueian, F.; Mendes, A. C.; Chronakis, I. S. In vitro permeability enhancement of curcumin across Caco–2 cells monolayers using electrospun xanthan–chitosan nanofibers. Carbohydr. Polym. 2019, 206, 38– 47, DOI: 10.1016/j.carbpol.2018.10.073There is no corresponding record for this reference.
- 54Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessmen of docking accuracy. J. Med. Chem. 2004, 47, 1739– 1749, DOI: 10.1021/jm030643054https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit74%253D&md5=8cc2f0022318b12dd972e9c493375bf9Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessment of docking accuracyFriesner, Richard A.; Banks, Jay L.; Murphy, Robert B.; Halgren, Thomas A.; Klicic, Jasna J.; Mainz, Daniel T.; Repasky, Matthew P.; Knoll, Eric H.; Shelley, Mee; Perry, Jason K.; Shaw, David E.; Francis, Perry; Shenkin, Peter S.Journal of Medicinal Chemistry (2004), 47 (7), 1739-1749CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Unlike other methods for docking ligands to the rigid 3D structure of a known protein receptor, Glide approximates a complete systematic search of the conformational, orientational, and positional space of the docked ligand. In this search, an initial rough positioning and scoring phase that dramatically narrows the search space is followed by torsionally flexible energy optimization on an OPLS-AA nonbonded potential grid for a few hundred surviving candidate poses. The very best candidates are further refined via a Monte Carlo sampling of pose conformation; in some cases, this is crucial to obtaining an accurate docked pose. Selection of the best docked pose uses a model energy function that combines empirical and force-field-based terms. Docking accuracy is assessed by redocking ligands from 282 cocrystd. PDB complexes starting from conformationally optimized ligand geometries that bear no memory of the correctly docked pose. Errors in geometry for the top-ranked pose are less than 1 Å in nearly half of the cases and are greater than 2 Å in only about one-third of them. Comparisons to published data on rms deviations show that Glide is nearly twice as accurate as GOLD and more than twice as accurate as FlexX for ligands having up to 20 rotatable bonds. Glide is also found to be more accurate than the recently described Surflex method.
- 55Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra Precision Glide: Docking and scoring incorporating a model of hydrophobic enclosure for protein–ligand complexes. J. Med. Chem. 2006, 49, 6177– 6196, DOI: 10.1021/jm051256o55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpvVGmurg%253D&md5=ea428c82ead0d8c27f8c1a7b694a1edfExtra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 56Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. J. Med. Chem. 2004, 47, 1750– 1759, DOI: 10.1021/jm030644s56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit78%253D&md5=33d68dd968e65626b449df61e44e37beGlide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screeningHalgren, Thomas A.; Murphy, Robert B.; Friesner, Richard A.; Beard, Hege S.; Frye, Leah L.; Pollard, W. Thomas; Banks, Jay L.Journal of Medicinal Chemistry (2004), 47 (7), 1750-1759CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Glide's ability to identify active compds. in a database screen is characterized by applying Glide to a diverse set of nine protein receptors. In many cases, two, or even three, protein sites are employed to probe the sensitivity of the results to the site geometry. To make the database screens as realistic as possible, the screens use sets of "druglike" decoy ligands that have been selected to be representative of what we believe is likely to be found in the compd. collection of a pharmaceutical or biotechnol. company. Results are presented for releases 1.8, 2.0, and 2.5 of Glide. The comparisons show that av. measures for both "early" and "global" enrichment for Glide 2.5 are 3 times higher than for Glide 1.8 and more than 2 times higher than for Glide 2.0 because of better results for the least well-handled screens. This improvement in enrichment stems largely from the better balance of the more widely parametrized GlideScore 2.5 function and the inclusion of terms that penalize ligand-protein interactions that violate established principles of phys. chem., particularly as it concerns the exposure to solvent of charged protein and ligand groups. Comparisons to results for the thymidine kinase and estrogen receptors published by Rognan and co-workers (J. Med. Chem. 2000, 43, 4759-4767) show that Glide 2.5 performs better than GOLD 1.1, FlexX 1.8, or DOCK 4.01.
- 57Petrella, S.; Capton, E.; Raynal, B.; Giffard, C.; Thureau, A.; Bonneté, F.; Alzari, P. M.; Aubry, A.; Mayer, C. Overall Structures of Mycobacterium tuberculosis DNA Gyrase Reveal the Role of a Corynebacteriales GyrB-Specific Insert in ATPase Activity. Structure. 2019, 27, 579– 589, DOI: 10.1016/j.str.2019.01.00457https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisl2lurw%253D&md5=492e76347e4646be2fb760e0655c4d14Overall Structures of Mycobacterium tuberculosis DNA Gyrase Reveal the Role of a Corynebacteriales GyrB-Specific Insert in ATPase ActivityPetrella, Stephanie; Capton, Estelle; Raynal, Bertrand; Giffard, Clement; Thureau, Aurelien; Bonnete, Francoise; Alzari, Pedro M.; Aubry, Alexandra; Mayer, ClaudineStructure (Oxford, United Kingdom) (2019), 27 (4), 579-589.e5CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)Despite sharing common features, previous studies have shown that gyrases from different species have been modified throughout evolution to modulate their properties. Here, we report two crystal structures of Mycobacterium tuberculosis DNA gyrase, an apo and AMPPNP-bound form at 2.6-Å and 3.3-Å resoln., resp. These structures provide high-resoln. structural data on the quaternary organization and interdomain connections of a gyrase (full-length GyrB-GyrA57)2 thus providing crucial inputs on this essential drug target. Together with small-angle X-ray scattering studies, they revealed an "extremely open" N-gate state, which persists even in the DNA-free gyrase-AMPPNP complex and an unexpected connection between the ATPase and cleavage core domains mediated by two Corynebacteriales-specific motifs, resp. the C-loop and DEEE-loop. We show that the C-loop participates in the stabilization of this open conformation, explaining why this gyrase has a lower ATPase activity. Our results image a conformational state which might be targeted for drug discovery.
- 58Agrawal, A.; Roue, M.; Spitzfaden, C.; Petrella, S.; Aubry, A.; Hann, M.; Bax, B.; Mayer, C. Mycobacterium tuberculosis DNA gyrase ATPase domain structures suggest a dissociative mechanism that explains how ATP hydrolysis is coupled to domain motion. Biochem. J. 2013, 456, 263– 273, DOI: 10.1042/BJ2013053858https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCmur3F&md5=761b89faa80294350062b3cbf31ab137Mycobacterium tuberculosis DNA gyrase ATPase domain structures suggest a dissociative mechanism that explains how ATP hydrolysis is coupled to domain motionAgrawal, Alka; Roue, Melanie; Spitzfaden, Claus; Petrella, Stephanie; Aubry, Alexandra; Hann, Michael; Bax, Benjamin; Mayer, ClaudineBiochemical Journal (2013), 456 (2), 263-273CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)DNA gyrase, a type II topoisomerase, regulates DNA topol. by creating a double-stranded break in one DNA duplex and transporting another DNA duplex [T-DNA (transported DNA)] through this break. The ATPase domains dimerize, in the presence of ATP, to trap the T-DNA segment. Hydrolysis of only one of the two ATPs, and release of the resulting Pi, is ratelimiting in DNA strand passage. A long unresolved puzzle is how the non-hydrolysable ATP analog AMP-PNP (adenosine 5'-[β,γ-imido]triphosphate) can catalyze one round of DNA strand passage without Pi release. In the present paper we discuss two crystal structures of the Mycobacterium tuberculosis DNA gyrase ATPase domain: one complexed with AMP-PCP (adenosine 5'-[β,γ-methylene]triphosphate) was unexpectedly monomeric, the other, an AMP-PNP complex, crystd. as a dimer. In the AMP-PNP structure, the unprotonated nitrogen (P-N=P imino) accepts hydrogen bonds from a well-ordered 'ATP lid', which is known to be required for dimerization. The equivalent CH2 group, in AMP-PCP, cannot accept hydrogen bonds, leaving the 'ATP lid' region disordered. Further anal. suggested that AMP-PNP can be converted from the imino (P-N=P) form into the imido form (P-NH-P) during the catalytic cycle. A main-chain NH is proposed to move to either protonate AMP-P-N=P to AMP-P-NH-P, or to protonate ATP to initiate ATP hydrolysis. This suggests a novel dissociative mechanism for ATP hydrolysis that could be applicable not only to GHKL phosphotransferases, but also to unrelated ATPases and GTPases such as Ras. On the basis of the domain orientation in our AMP-PCP structure we propose a mechanochem. scheme to explain how ATP hydrolysis is coupled to domain motion.
- 59Shirude, P. S.; Madhavapeddi, P.; Tucker, J. A.; Murugan, K.; Patil, V.; Basavarajappa, H.; Raichurkar, A. V.; Humnabadkar, V.; Hussein, S.; Sharma, S.; Ramya, V. K.; Narayan, C. B.; Balganesh, T. S.; Sambandamurthy, V. K. Aminopyrazinamides: novel and specific GyrB inhibitors that kill replicating and nonreplicating Mycobacterium tuberculosis. ACS Chem. Biol. 2013, 8, 519– 523, DOI: 10.1021/cb300510w59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvV2gsLrI&md5=4f8053bd1721c40019c70fab7f6147beAminopyrazinamides: novel and specific GyrB inhibitors that kill replicating and nonreplicating Mycobacterium tuberculosisShirude, Pravin S.; Madhavapeddi, Prashanti; Tucker, Julie A.; Murugan, Kannan; Patil, Vikas; Basavarajappa, Halesha; Raichurkar, Anandkumar V.; Humnabadkar, Vaishali; Hussein, Syeed; Sharma, Sreevalli; Ramya, V. K.; Narayan, Chandan B.; Balganesh, Tanjore S.; Sambandamurthy, Vasan K.ACS Chemical Biology (2013), 8 (3), 519-523CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Aminopyrazinamides originated from a high throughput screen targeting the Mycobacterium smegmatis (Msm) GyrB ATPase. This series displays chem. tractability, robust structure-activity relationship, and potent antitubercular activity. The crystal structure of Msm GyrB in complex with one of the aminopyrazinamides revealed promising attributes of specificity against other broad spectrum pathogens and selectivity against eukaryotic kinases due to novel interactions at hydrophobic pocket, unlike other known GyrB inhibitors. The aminopyrazinamides display excellent mycobacterial kill under in vitro, intracellular, and hypoxic conditions.
- 60Greenwood, J. R.; Calkins, D.; Sullivan, A. P.; Shelley, J. C. Towards the comprehensive, rapid, and accurate prediction of the favorable tautomeric states of drug–like molecules in aqueous solution. J. Comput. Aided Mol. Des. 2010, 24, 591– 604, DOI: 10.1007/s10822-010-9349-160https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnsFGqtbo%253D&md5=1d7bc0f966ca793d6be80554868367b8Towards the comprehensive, rapid, and accurate prediction of the favorable tautomeric states of drug-like molecules in aqueous solutionGreenwood, Jeremy R.; Calkins, David; Sullivan, Arron P.; Shelley, John C.Journal of Computer-Aided Molecular Design (2010), 24 (6-7), 591-604CODEN: JCADEQ; ISSN:0920-654X. (Springer)A review. Generating the appropriate protonation states of drug-like mols. in soln. is important for success in both ligand- and structure-based virtual screening. Screening collections of millions of compds. requires a method for detg. tautomers and their energies that is sufficiently rapid, accurate, and comprehensive. To maximize enrichment, the lowest energy tautomers must be detd. from heterogeneous input, without over-enumerating unfavorable states. While computationally expensive, the d. functional theory (DFT) method M06-2X/aug-cc-pVTZ(-f) [PB-SCRF] provides accurate energies for enumerated model tautomeric systems. The empirical Hammett-Taft methodol. can very rapidly extrapolate substituent effects from model systems to drug-like mols. via the relationship between pKT and pKa. Combining the 2 complementary approaches transforms the tautomer problem from a scientific challenge to one of engineering scale-up, and avoids issues that arise due to the very limited no. of measured pKT values, esp. for the complicated heterocycles often favored by medicinal chemists for their novelty and versatility. Several hundreds of pre-calcd. tautomer energies and substituent pKa effects are tabulated in databases for use in structural adjustment by the program Epik, which treats tautomers as a subset of the larger problem of the protonation states in aq. ensembles and their energy penalties. Accuracy and coverage is continually improved and expanded by parameterizing new systems of interest using DFT and exptl. data. Recommendations are made for how to best incorporate tautomers in mol. design and virtual screening workflows.
- 61Shelley, J. C.; Cholleti, A.; Frye, L. L.; Greenwood, J. R.; Timlin, M. R.; Uchimaya, M. Epik: a software program for pKa prediction and protonation state generation for drug–like molecules. J. Comput. Aided Mol. Des. 2007, 21, 681– 691, DOI: 10.1007/s10822-007-9133-z61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVKrtbzP&md5=f4f429ea3894e1ad2519cdf3333a5645Epik: a software program for pKa prediction and protonation state generation for drug-like moleculesShelley, John C.; Cholleti, Anuradha; Frye, Leah L.; Greenwood, Jeremy R.; Timlin, Mathew R.; Uchimaya, MakotoJournal of Computer-Aided Molecular Design (2007), 21 (12), 681-691CODEN: JCADEQ; ISSN:0920-654X. (Springer)Epik is a computer program for predicting pKa values for drug-like mols. Epik can use this capability in combination with technol. for tautomerization to adjust the protonation state of small drug-like mols. to automatically generate one or more of the most probable forms for use in further mol. modeling studies. Many medicinal chems. can exchange protons with their environment, resulting in various ionization and tautomeric states, collectively known as protonation states. The protonation state of a drug can affect its soly. and membrane permeability. In modeling, the protonation state of a ligand will also affect which conformations are predicted for the mol., as well as predictions for binding modes and ligand affinities based upon protein-ligand interactions. Despite the importance of the protonation state, many databases of candidate mols. used in drug development do not store reliable information on the most probable protonation states. Epik is sufficiently rapid and accurate to process large databases of drug-like mols. to provide this information. Several new technologies are employed. Extensions to the well-established Hammett and Taft approaches are used for pKa prediction, namely, mesomer standardization, charge cancellation, and charge spreading to make the predicted results reflect the nature of the mol. itself rather just for the particular Lewis structure used on input. In addn., a new iterative technol. for generating, ranking and culling the generated protonation states is employed.
- 62Case, D. A.; Aktulga, H. M.; Belfon, K.; Ben Shalom, I. Y.; Brozell, S. R.; Cerutti, D. S.; Cheatham, III, T. E.; Cisneros, G. A.; Cruzeiro, V. W. D.; Darden, T. A.; Duke, R. E.; Giambasu, G.; Gilson, M. K.; Gohlke, H.; Goetz, A. W.; Harris, R.; Izadi, S.; Izmailov, S. A.; Jin, C.; Kasavajhala, K.; Kaymak, M. C.; King, E.; Kovalenko, A.; Kurtzman, T.; Lee, T. S.; LeGrand, S.; Li, P.; Lin, C.; Liu, J.; Luchko, T.; Luo, R.; Machado, M.; Man, V.; Manathunga, M.; Merz, K. M.; Miao, Y.; Mikhailovskii, O.; Monard, G.; Nguyen, H.; O’Hearn, K. A.; Onufriev, A.; Pan, F.; Pantano, S.; Qi, R.; Rahnamoun, A.; Roe, D. R.; Roitberg, A.; Sagui, C.; Schott Verdugo, S.; Shen, J.; Simmerling, C. L.; Skrynnikov, N. R.; Smith, J.; Swails, J.; Walker, R. C.; Wang, J.; Wei, H.; Wolf, R. M.; Wu, X.; Xue, Y.; York, D. M.; Zhao, S.; Kollman, P. A. Amber 2020; University of California: San Francisco, 2021.There is no corresponding record for this reference.
- 63Wang, J.; Wolf, R. M.; Caldwell, J. W.; Kollman, P. A.; Case, D. A. Development and testing of a general amber force field. J. Comput. Chem. 2004, 25, 1157– 1174, DOI: 10.1002/jcc.2003563https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksFakurc%253D&md5=2992017a8cf51f89290ae2562403b115Development and testing of a general Amber force fieldWang, Junmei; Wolf, Romain M.; Caldwell, James W.; Kollman, Peter A.; Case, David A.Journal of Computational Chemistry (2004), 25 (9), 1157-1174CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)We describe here a general Amber force field (GAFF) for org. mols. GAFF is designed to be compatible with existing Amber force fields for proteins and nucleic acids, and has parameters for most org. and pharmaceutical mols. that are composed of H, C, N, O, S, P, and halogens. It uses a simple functional form and a limited no. of atom types, but incorporates both empirical and heuristic models to est. force consts. and partial at. charges. The performance of GAFF in test cases is encouraging. In test I, 74 crystallog. structures were compared to GAFF minimized structures, with a root-mean-square displacement of 0.26 Å, which is comparable to that of the Tripos 5.2 force field (0.25 Å) and better than those of MMFF 94 and CHARMm (0.47 and 0.44 Å, resp.). In test II, gas phase minimizations were performed on 22 nucleic acid base pairs, and the minimized structures and intermol. energies were compared to MP2/6-31G* results. The RMS of displacements and relative energies were 0.25 Å and 1.2 kcal/mol, resp. These data are comparable to results from Parm99/RESP (0.16 Å and 1.18 kcal/mol, resp.), which were parameterized to these base pairs. Test III looked at the relative energies of 71 conformational pairs that were used in development of the Parm99 force field. The RMS error in relative energies (compared to expt.) is about 0.5 kcal/mol. GAFF can be applied to wide range of mols. in an automatic fashion, making it suitable for rational drug design and database searching.
- 64Bayly, C. I.; Cieplak, P.; Cornell, W. D.; Kollman, P. A. A Well behaved electrostatic potential based method using charge restraints for deriving atomic charges: The RESP model. J. Phys. Chem. 1993, 97, 10269– 10280, DOI: 10.1021/j100142a00464https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXlvVyqsLs%253D&md5=e65c6a556ffc174df4f327687912a0bdA well-behaved electrostatic potential based method using charge restraints for deriving atomic charges: the RESP modelBayly, Christopher I.; Cieplak, Piotr; Cornell, Wendy; Kollman, Peter A.Journal of Physical Chemistry (1993), 97 (40), 10269-80CODEN: JPCHAX; ISSN:0022-3654.The authors present a new approach to generating electrostatic potential (ESP) derived charges for mols. The major strength of electrostatic potential derived charges is that they optimally reproduce the intermol. interaction properties of mols. with a simple two-body additive potential, provided, of course, that a suitably accurate level of quantum mech. calcn. is used to derive the ESP around the mol. Previously, the major weaknesses of these charges have been that they were not easily transferably between common functional groups in related mols., they have often been conformationally dependent, and the large charges that frequently occur can be problematic for simulating intramol. interactions. Introducing restraints in the form of a penalty function into the fitting process considerably reduces the above problems, with only a minor decrease in the quality of the fit to the quantum mech. ESP. Several other refinements in addn. to the restrained electrostatic potential (RESP) fit yield a general and algorithmic charge fitting procedure for generating atom-centered point charges. This approach can thus be recommended for general use in mol. mechanics, mol. dynamics, and free energy calcns. for any org. or bioorg. system.
- 65Cieplak, P.; Cornell, W. D.; Bayly, C.; Kollman, P. A. Application of the multimolecule and multiconformational RESP methodology to biopolymers: Charge derivation for DNA, RNA, and proteins. J. Comput. Chem. 1995, 16, 1357– 1377, DOI: 10.1002/jcc.54016110665https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXovVKqtrY%253D&md5=27e75f5f3f53d777737661e591e957cdApplication of the multimolecule and multiconformational RESP methodology to biopolymers: charge derivation for DNA, RNA, and proteinsCieplak, Piotr; Cornell, Wendy D.; Bayly, Christopher; Kollman, Peter A.Journal of Computational Chemistry (1995), 16 (11), 1357-77CODEN: JCCHDD; ISSN:0192-8651. (Wiley)The authors present the derivation of charges of ribo- and deoxynucleosides, nucleotides, and peptide fragments using electrostatic potentials obtained from ab initio calcns. with the 6-31G* basis set. For the nucleic acid fragments, the authors used electrostatic potentials of the four deoxyribonucleoside (A, G, C, T) and four ribonucleosides (A, G, C, U) and dimethylphosphate. The charges for the deoxyribose nucleosides and nucleotides are derived using multiple-mol. fitting and restrained electrostatic potential (RESP) fits, with Lagrangian multipliers ensuring a net charge of 0 or. The authors suggest that the preferred approach for deriving charges for nucleosides and nucleotides involves allowing only C1' and H1' of the sugar to vary as the nucleic acid base, with the remainder of sugar and backbone atoms forced to be equiv. For peptide fragments, the authors have combined multiple conformation fitting, previously employed by Williams and Reynolds et al., with the RESP approach to derive charges for blocked dipeptides appropriate for each of the 20 naturally occurring amino acids. Based on the results for Pr amine, the authors suggest that two conformations for each peptide suffice to give charges that represent well the conformationally dependent electrostatic properties of mols., provided that these two conformations contain different values of the dihedral angles that terminate in heteroatoms or hydrogens attached to heteroatoms or hydrogens attached to heteroatoms. In these blocked dipeptide models, it is useful to require equiv. N-H and C=O charges for all amino acids with a given net charge (except proline), and this is accomplished in a straightforward fashion with multiple-mol. fitting. Finally, the application of multiple Lagrangian constraints allows for the derivation of monomeric residues with the appropriate net charge from a chem. blocked version of the residue. The multiple Lagrange constraints also enable charges from two or more mols. to be spliced together in a well-defined fashion. Thus, the combined use of multiple mols., multiple conformations, multiple Lagrangian constraints, and RESP fitting is shown to be a powerful approach to deriving electrostatic charges for biopolymers.
- 66Fox, T.; Kollman, P. A. Application of the RESP methodology in the parametrization of organic solvents. J. Phys. Chem. B 1998, 102, 8070– 8079, DOI: 10.1021/jp971765566https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXmtVelu78%253D&md5=848186c21337d0ff82cafd35a3a526a4Application of the RESP Methodology in the Parametrization of Organic SolventsFox, Thomas; Kollman, Peter A.Journal of Physical Chemistry B (1998), 102 (41), 8070-8079CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)We present parameterizations for the nonaq. solvents DMSO, ethanol, CCl4, CHCl3, and CH2Cl2 that are compatible with the recent AMBER force field by Cornell et al. (J. Am. Chem. Soc. 1995, 117, 5179-5197). With the general procedure for generating new parameters and the RESP approach to obtain the at. charges, we achieve flexible all-atom solvent models whose d., heat of vaporization, diffusion const., and rotational correlation times are-esp. for a generic force field-in good agreement with available exptl. data.
- 67Jorgensen, W. L.; Chandrasekhar, J.; Madura, J. D.; Impey, R. W.; Klein, M. L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 1983, 79, 926– 935, DOI: 10.1063/1.44586967https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3sXksF2htL4%253D&md5=a1161334e381746be8c9b15a5e56f704Comparison of simple potential functions for simulating liquid waterJorgensen, William L.; Chandrasekhar, Jayaraman; Madura, Jeffry D.; Impey, Roger W.; Klein, Michael L.Journal of Chemical Physics (1983), 79 (2), 926-35CODEN: JCPSA6; ISSN:0021-9606.Classical Monte Carlo simulations were carried out for liq. H2O in the NPT ensemble at 25° and 1 atm using 6 of the simpler intermol. potential functions for the dimer. Comparisons were made with exptl. thermodn. and structural data including the neutron diffraction results of Thiessen and Narten (1982). The computed densities and potential energies agree with expt. except for the original Bernal-Fowler model, which yields an 18% overest. of the d. and poor structural results. The discrepancy may be due to the correction terms needed in processing the neutron data or to an effect uniformly neglected in the computations. Comparisons were made for the self-diffusion coeffs. obtained from mol. dynamics simulations.
- 68Roe, D. R.; Cheatham, T., III. PTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory Data. J. Chem. Theory Comput. 2013, 9, 3084– 3095, DOI: 10.1021/ct400341p68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFehtr8%253D&md5=6f1bee934f13f180bd7e1feb6b78036dPTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory DataRoe, Daniel R.; Cheatham, Thomas E.Journal of Chemical Theory and Computation (2013), 9 (7), 3084-3095CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We describe PTRAJ and its successor CPPTRAJ, two complementary, portable, and freely available computer programs for the anal. and processing of time series of three-dimensional at. positions (i.e., coordinate trajectories) and the data therein derived. Common tools include the ability to manipulate the data to convert among trajectory formats, process groups of trajectories generated with ensemble methods (e.g., replica exchange mol. dynamics), image with periodic boundary conditions, create av. structures, strip subsets of the system, and perform calcns. such as RMS fitting, measuring distances, B-factors, radii of gyration, radial distribution functions, and time correlations, among other actions and analyses. Both the PTRAJ and CPPTRAJ programs and source code are freely available under the GNU General Public License version 3 and are currently distributed within the AmberTools 12 suite of support programs that make up part of the Amber package of computer programs (see http://ambermd.org). This overview describes the general design, features, and history of these two programs, as well as algorithmic improvements and new features available in CPPTRAJ.
- 69Wang, E.; Sun, H.; Wang, J.; Wang, Z.; Liu, H.; Zhang, J. Z. H.; Hou, T. End–point binding free energy calculation with MM/PBSA and MM/GBSA: Strategies and applications in drug design. Chem. Rev. 2019, 119, 9478– 9508, DOI: 10.1021/acs.chemrev.9b00055There is no corresponding record for this reference.
- 70Hou, T.; Yu, R. Molecular dynamics and free energy studies on the wild–type and double mutant HIV–1 protease complexed with amprenavir and two amprenavir–related inhibitors: mechanism for binding and drug resistance. J. Med. Chem. 2007, 50, 1177– 1188, DOI: 10.1021/jm060916270https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhs1Ghtbk%253D&md5=e13654603bac0ccfc57e8720e98cf8eeMolecular Dynamics and Free Energy Studies on the Wild-type and Double Mutant HIV-1 Protease Complexed with Amprenavir and Two Amprenavir-Related Inhibitors: Mechanism for Binding and Drug ResistanceHou, Tingjun; Yu, RonJournal of Medicinal Chemistry (2007), 50 (6), 1177-1188CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The V82F/I84V double mutation is considered as the key residue mutation of the HIV-1 protease drug resistance because it can significantly lower the binding affinity of protease inhibitors in clin. uses. In the current work, the binding of amprenavir to both of the wild-type and the drug-resistant V82F/I84V mutant of the HIV-1 protease was investigated by mol. dynamics (MD) simulations and was compared to those of two inhibitors in development, TMC126 and TMC114. Abs. binding free energies were calcd. by mol. mechanics/Poisson-Boltzmann surface area (MM/ PBSA) methodol. The predicted binding affinities give a good explanation of structure-affinity relation (SAR) of three studied inhibitors. Furthermore, in the 18 ns MD simulations on the free wild-type and the mutated proteases, we obsd. that the free mutated protease shows similar dynamic characteristics of the flap opening and a little higher structural stability than the free wild-type protease. This suggests that the effect of the mutations may not significantly affect the equil. between the semi-open and the closed conformations. Finally, decompn. anal. of binding free energies and the further structural anal. indicate that the dominating effect of the V82F/I84V double mutation is to distort the geometry of the binding site and hence weaken the interactions of inhibitors pre-shaped to the wild-type binding site.
- 71Li, W.; Zhang, J.; Guo, L.; Wang, Q. Importance of three–body problems and protein–protein interactions in proteolysis–targeting chimera modeling: Insights from molecular dynamics simulations. J. Chem. Inf. Model. 2022, 62, 523– 532, DOI: 10.1021/acs.jcim.1c01150There is no corresponding record for this reference.
- 72Miller, B. R.; McGee, T. D.; Swails, J. M.; Homeyer, N.; Gohlke, H.; Roitberg, A. E. MMPBSA.py: An efficient program for end–state free energy calculations. J. Chem. Theory Comput. 2012, 8, 3314– 3321, DOI: 10.1021/ct300418h72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2gtrzP&md5=cc4148bd8f70c7cad94fd3ec6f580e52MMPBSA.py: An Efficient Program for End-State Free Energy CalculationsMiller, Bill R., III; McGee, T. Dwight, Jr.; Swails, Jason M.; Homeyer, Nadine; Gohlke, Holger; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (9), 3314-3321CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)MM-PBSA is a post-processing end-state method to calc. free energies of mols. in soln. MMPBSA.py is a program written in Python for streamlining end-state free energy calcns. using ensembles derived from mol. dynamics (MD) or Monte Carlo (MC) simulations. Several implicit solvation models are available with MMPBSA.py, including the Poisson-Boltzmann Model, the Generalized Born Model, and the Ref. Interaction Site Model. Vibrational frequencies may be calcd. using normal mode or quasi-harmonic anal. to approx. the solute entropy. Specific interactions can also be dissected using free energy decompn. or alanine scanning. A parallel implementation significantly speeds up the calcn. by dividing frames evenly across available processors. MMPBSA.py is an efficient, user-friendly program with the flexibility to accommodate the needs of users performing end-state free energy calcns. The source code can be downloaded at http://ambermd.org/ with AmberTools, released under the GNU General Public License.
- 73Onufriev, A.; Bashford, D.; Case, D. A. Exploring protein native states and large–scale conformational changes with a modified generalized born model. Proteins. 2004, 55, 383– 394, DOI: 10.1002/prot.2003373https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjtFKhs78%253D&md5=06bab80e7df579625cb29a4ae7260d0cExploring protein native states and large-scale conformational changes with a modified Generalized Born modelOnufriev, Alexey; Bashford, Donald; Case, David A.Proteins: Structure, Function, and Bioinformatics (2004), 55 (2), 383-394CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)Implicit solvation models provide, for many applications, a reasonably accurate and computationally effective way to describe the electrostatics of aq. solvation. Here, a popular anal. Generalized Born (GB) solvation model is modified to improve its accuracy in calcg. the solvent polarization part of free energy changes in large-scale conformational transitions, such as protein folding. In contrast to an earlier GB model (implemented in the AMBER-6 program), the improved version does not overstabilize the native structures relative to the finite-difference Poisson-Boltzmann continuum treatment. In addn. to improving the energy balance between folded and unfolded conformers, the algorithm (available in the AMBER-7 and NAB mol. modeling packages) is shown to perform well in more than 50 ns of native-state mol. dynamics (MD) simulations of thioredoxin, protein-A, and ubiquitin, as well as in a simulation of Barnase/Barstar complex formation. For thioredoxin, various combinations of input parameters have been explored, such as the underlying gas-phase force fields and the at. radii. The best performance is achieved with a previously proposed modification to the torsional potential in the Amber ff99 force field, which yields stable native trajectories for all of the tested proteins, with back-bone root-mean-square deviations from the native structures being ∼ 1.5 Å after 6 ns of simulation time. The structure of Barnase/Barstar complex is regenerated, starting from an unbound state, to within 1.9 Å relative to the crystal structure of the complex.
- 74Weiser, J.; Shenkin, P. S.; Still, W. C. Approximate atomic surfaces from linear combinations of pairwise overlaps (LCPO). J. Comput. Chem. 1999, 20, 217– 230, DOI: 10.1002/(SICI)1096-987X(19990130)20:2<217::AID-JCC4>3.0.CO;2-A74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXltVCltQ%253D%253D&md5=1c6cf923fe089cf61a1952fdcf55f788Approximate atomic surfaces from linear combinations of pairwise overlaps (LCPO)Weiser, Jorg; Shenkin, Peter S.; Still, W. ClarkJournal of Computational Chemistry (1999), 20 (2), 217-230CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)A fast anal. formula was derived for the calcn. of approx. at. and mol. van der Waals (vdWSA), and solvent-accessible surface areas (SASAs), as well as the first and second derivs. of these quantities with respect to at. coordinates. This method makes use of linear combinations of terms composed from pairwise overlaps of hard spheres; therefore, we term this the LCPO method for linear combination of pairwise overlaps. For higher performance, neighbor-list redn. (NLR) was applied as a preprocessing step. Eighteen compds. of different sizes (8-2366 atoms) and classes (org., proteins, DNA, and various complexes) were chosen as representative test cases. LCPO/NLR computed the SASA and first derivs. of penicillopepsin, a protein with 2366 atoms, in 0.87 s (0.22 s for the creation of the neighbor list, 0.35 s for NLR, and 0.30 s for SASA and first derivs.) on an SGI R10000/194 Mhz processor. This appears comparable to or better than timings reported previously for other algorithms. The vdWSAs were in good agreement with the numerical results: relative errors for total mol. surface areas ranged from 0.1 to 2.0% and av. abs. at. surface area deviations from 0.3 to 0.7 Å2. For SASAs without NLR, the LCPO method exhibited relative errors in the range of 0.4-9.2% for total mol. surface areas and av. abs. at. surface area deviations of 2.0-2.7 Å2; with NLR the relative mol. errors ranged from 0.1 to 7.8% and the av. abs. at. surface area deviation from 1.6 to 3.0 Å2.
- 75Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 2001, 46, 3– 26, DOI: 10.1016/S0169-409X(00)00129-075https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXitVOhs7o%253D&md5=c60bb89da68f051c0ee7ac4c0468a0e4Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settingsLipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J.Advanced Drug Delivery Reviews (2001), 46 (1-3), 3-26CODEN: ADDREP; ISSN:0169-409X. (Elsevier Science Ireland Ltd.)A review with 50 refs. Exptl. and computational approaches to est. soly. and permeability in discovery and development settings are described. In the discovery setting 'the rule of 5' predicts that poor absorption or permeation is more likely when there are more than 5 H-bond donors, 10 H-bond acceptors, the mol. wt. (MWT) is greater than 500 and the calcd. Log P (CLogP) is greater than 5 (or MlogP >4.15). Computational methodol. for the rule-based Moriguchi Log P (MLogP) calcn. is described. Turbidimetric soly. measurement is described and applied to known drugs. High throughput screening (HTS) leads tend to have higher MWT and Log P and lower turbidimetric soly. than leads in the pre-HTS era. In the development setting, soly. calcns. focus on exact value prediction and are difficult because of polymorphism. Recent work on linear free energy relationships and Log P approaches are critically reviewed. Useful predictions are possible in closely related analog series when coupled with exptl. thermodn. soly. measurements.
- 76Baell, J. B.; Holloway, G. A. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J. Med. Chem. 2010, 53, 2719– 2740, DOI: 10.1021/jm901137j76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsF2qsLw%253D&md5=fbf397aa4910753c550425708c866fd2New Substructure Filters for Removal of Pan Assay Interference Compounds (PAINS) from Screening Libraries and for Their Exclusion in BioassaysBaell, Jonathan B.; Holloway, Georgina A.Journal of Medicinal Chemistry (2010), 53 (7), 2719-2740CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)This report describes a no. of substructural features which can help to identify compds. that appear as frequent hitters (promiscuous compds.) in many biochem. high throughput screens. The compds. identified by such substructural features are not recognized by filters commonly used to identify reactive compds. Even though these substructural features were identified using only one assay detection technol., such compds. have been reported to be active from many different assays. In fact, these compds. are increasingly prevalent in the literature as potential starting points for further exploration, whereas they may not be.
- 77Alt, S.; Mitchenall, L. A.; Maxwell, A.; Heide, L. Inhibition of DNA gyrase and DNA topoisomerase IV of Staphylococcus aureus and Escherichia coli by aminocoumarin antibiotics. J. Antimicrob. Chemother. 2011, 66, 2061– 2069, DOI: 10.1093/jac/dkr24777https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVeitLvE&md5=ee96fa3dfea838f59610bd5ac1cce584Inhibition of DNA gyrase and DNA topoisomerase IV of Staphylococcus aureus and Escherichia coli by aminocoumarin antibioticsAlt, Silke; Mitchenall, Lesley A.; Maxwell, Anthony; Heide, LutzJournal of Antimicrobial Chemotherapy (2011), 66 (9), 2061-2069CODEN: JACHDX; ISSN:0305-7453. (Oxford University Press)Aminocoumarin antibiotics are potent inhibitors of bacterial DNA gyrase. We investigated the inhibitory and antibacterial activity of naturally occurring aminocoumarin antibiotics and six structural analogs (novclobiocins) against DNA gyrase and DNA topoisomerase IV from Escherichia coli and Staphylococcus aureus as well as the effect of potassium and sodium glutamate on the activity of these enzymes. The inhibitory concns. of the aminocoumarins were detd. in gyrase supercoiling assays and topoisomerase IV decatenation assays. Both subunits of S. aureus topoisomerase IV were purified as His-Tag proteins in E. coli. The MIC was tested in vivo for the control organisms E. coli ATCC 25922 and S. aureus ATCC 29213. DNA gyrase is the primary target in vitro of all investigated aminocoumarins. With the exception of simocyclinone D8, all other aminocoumarins inhibited S. aureus gyrase on av. 6-fold more effectively than E. coli gyrase. Potassium glutamate is essential for the activity of S. aureus gyrase and increases the sensitivity of E. coli gyrase to aminocoumarins ≥10-fold. The antibacterial activity of the tested compds. mirrored their relative activities against topoisomerases. The study provides insights about the substituents that are important for the inhibitory activity of aminocoumarins against the target enzymes, which will facilitate the rational design of improved antibiotics.
- 78Bisacchi, G. S.; Manchester, J. I. A New-Class Antibacterial-Almost. Lessons in Drug Discovery and Development: A Critical Analysis of More than 50 Years of Effort toward ATPase Inhibitors of DNA Gyrase and Topoisomerase IV. ACS. Infect. Dis. 2015, 1, 4– 41, DOI: 10.1021/id500013tThere is no corresponding record for this reference.
- 79Duma, R. J.; Warner, J. F. In Vitro Activity of Coumermycin A1 Against Mycobacterium tuberculosis var. hominis. Appl. Microbiol. 1969, 18, 404– 405, DOI: 10.1128/am.18.3.404-405.1969There is no corresponding record for this reference.
- 80Broeck, A. V.; McEwen, A. G.; Chebaro, Y.; Potier, N.; Lamour, V. Structural Basis for DNA Gyrase Interaction with Coumermycin A1. J. Med. Chem. 2019, 62, 4225– 4231, DOI: 10.1021/acs.jmedchem.8b01928There is no corresponding record for this reference.
- 81Gl, B.; Rajput, R.; Gupta, M.; Dahiya, P.; Thakur, J. K.; Bhatnagar, R.; Grover, A. Structure-based drug repurposing to inhibit the DNA gyrase of Mycobacterium tuberculosis. Biochem. J. 2020, 477, 4167– 4190, DOI: 10.1042/BCJ2020046281https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s%252FovVGhsQ%253D%253D&md5=b16377b6b133bbe1ae523f52dd7e8725Structure-based drug repurposing to inhibit the DNA gyrase of Mycobacterium tuberculosisGl Balasubramani; Rajput Rinky; Grover Abhinav; Gupta Manish; Bhatnagar Rakesh; Dahiya Pradeep; Thakur Jitendra K; Bhatnagar RakeshThe Biochemical journal (2020), 477 (21), 4167-4190 ISSN:.Drug repurposing is an alternative avenue for identifying new drugs to treat tuberculosis (TB). Despite the broad-range of anti-tubercular drugs, the emergence of multi-drug-resistant and extensively drug-resistant strains of Mycobacterium tuberculosis (Mtb) H37Rv, as well as the significant death toll globally, necessitates the development of new and effective drugs to treat TB. In this study, we have employed a drug repurposing approach to address this drug resistance problem by screening the drugbank database to identify novel inhibitors of the Mtb target enzyme, DNA gyrase. The compounds were screened against the ATPase domain of the gyrase B subunit (MtbGyrB47), and the docking results showed that echinacoside, doxorubicin, epirubicin, and idarubicin possess high binding affinities against MtbGyrB47. Comprehensive assessment using fluorescence spectroscopy, surface plasmon resonance spectroscopy (SPR), and circular dichroism (CD) titration studies revealed echinacoside as a potent binder of MtbGyrB47. Furthermore, ATPase, and DNA supercoiling assays exhibited an IC50 values of 2.1-4.7 μM for echinacoside, doxorubicin, epirubicin, and idarubicin. Among these compounds, the least MIC90 of 6.3 and 12 μM were observed for epirubicin and echinacoside, respectively, against Mtb. Our findings indicate that echinacoside and epirubicin targets mycobacterial DNA gyrase, inhibit its catalytic cycle, and retard mycobacterium growth. Further, these compounds exhibit potential scaffolds for optimizing novel anti-mycobacterial agents that can act on drug-resistant strains.
- 82Sherer, B. A.; Hull, K.; Green, O.; Basarab, G.; Hauck, S.; Hill, P.; Loch, J. T., 3rd.; Mullen, G.; Bist, S.; Bryant, J.; Boriack-Sjodin, A.; Read, J.; DeGrace, N.; Uria-Nickelsen, M.; Illingworth, R. N.; Eakin, A. E. Pyrrolamide DNA gyrase inhibitors: Optimization of antibacterial activity and efficacy. Bioorg. Med. Chem. Lett. 2011, 21, 7416– 7420, DOI: 10.1016/j.bmcl.2011.10.01082https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFSqtLnO&md5=0511dac22318e0af1337593e81f6d309Pyrrolamide DNA gyrase inhibitors: Optimization of antibacterial activity and efficacySherer, Brian A.; Hull, Kenneth; Green, Oluyinka; Basarab, Gregory; Hauck, Sheila; Hill, Pamela; Loch, James T., III; Mullen, George; Bist, Shanta; Bryant, Joanna; Boriack-Sjodin, Ann; Read, Jon; DeGrace, Nancy; Uria-Nickelsen, Maria; Illingworth, Ruth N.; Eakin, Ann E.Bioorganic & Medicinal Chemistry Letters (2011), 21 (24), 7416-7420CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The pyrrolamides are a new class of antibacterial agents targeting DNA gyrase, an essential enzyme across bacterial species and inhibition results in the disruption of DNA synthesis and subsequently, cell death. The optimization of biochem. activity and other drug-like properties through substitutions to the pyrrole, piperidine, and heterocycle portions of the mol. resulted in pyrrolamides with improved cellular activity and in vivo efficacy.
- 83Durcik, M.; Nyerges, Á.; Skok, Ž.; Skledar, D. G.; Trontelj, J.; Zidar, N.; Ilaš, J.; Zega, A.; Cruz, C. D.; Tammela, P.; Welin, M.; Kimbung, Y. R.; Focht, D.; Benek, O.; Révész, T.; Draskovits, G.; Szili, PÉ; Daruka, L.; Pál, C.; Kikelj, D.; Mašič, L. P.; Tomašič, T. New dual ATP-competitive inhibitors of bacterial DNA gyrase and topoisomerase IV active against ESKAPE pathogens. Eur. J. Med. Chem. 2021, 213, 113200 DOI: 10.1016/j.ejmech.2021.11320083https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjtVWisLY%253D&md5=cd3848925844b467ff2d46ab988c67c9New dual ATP-competitive inhibitors of bacterial DNA gyrase and topoisomerase IV active against ESKAPE pathogensDurcik, Martina; Nyerges, Akos; Skok, Ziga; Skledar, Darja Gramec; Trontelj, Jurij; Zidar, Nace; Ilas, Janez; Zega, Anamarija; Cruz, Cristina D.; Tammela, Paivi; Welin, Martin; Kimbung, Yengo R.; Focht, Dorota; Benek, Ondrej; Revesz, Tamas; Draskovits, Gabor; Szili, Petra Eva; Daruka, Lejla; Pal, Csaba; Kikelj, Danijel; Masic, Lucija Peterlin; Tomasic, TihomirEuropean Journal of Medicinal Chemistry (2021), 213 (), 113200CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Here, the design and structure-activity relationship anal. of balanced, low nanomolar inhibitors of bacterial DNA gyrase and topoisomerase IV that show potent antibacterial activities against the ESKAPE pathogens were reported. For inhibitor I•HCl (R = 2-(morpholin-4-yl)ethyl), a crystal structure in complex with Staphylococcus aureus DNA gyrase B was obtained that confirms the mode of action of these compds. The best inhibitor, I [R = thiophen-3-yl-Me (II)], does not show any in vitro cytotoxicity and has excellent potency against Gram-pos. (MICs: range, 0.0078-0.0625μg/mL) and Gram-neg. pathogens (MICs: range, 1-2μg/mL). Furthermore, II inhibits GyrB mutants that can develop resistance to other drugs. Based on these data, it was expected that structural derivs. of II will represent a step toward clin. efficacious multitargeting antimicrobials that are not impacted by existing antimicrobial resistance.
- 84Mesleh, M. F.; Cross, J. B.; Zhang, J.; Kahmann, J.; Andersen, O. A.; Barker, J.; Cheng, R. K.; Felicetti, B.; Wood, M.; Hadfield, A. T.; Scheich, C.; Moy, T. I.; Yang, Q.; Shotwell, J.; Nguyen, K.; Lippa, B.; Dolle, R.; Ryan, M. D. Fragment-based discovery of DNA gyrase inhibitors targeting the ATPase subunit of GyrB. Bioorg. Med. Chem. Lett. 2016, 26, 1314– 1318, DOI: 10.1016/j.bmcl.2016.01.009There is no corresponding record for this reference.
- 85Ushiyama, F.; Amada, H.; Takeuchi, T.; Tanaka-Yamamoto, N.; Kanazawa, H.; Nakano, K.; Mima, M.; Masuko, A.; Takata, I.; Hitaka, K.; Iwamoto, K.; Sugiyama, H.; Ohtake, N. Lead Identification of 8-(Methylamino)-2-oxo-1,2-dihydroquinoline Derivatives as DNA Gyrase Inhibitors: Hit-to-Lead Generation Involving Thermodynamic Evaluation. ACS Omega 2022, 5, 10145– 10159, DOI: 10.1021/acsomega.0c00865There is no corresponding record for this reference.
- 86Skok, Ž.; Barančoková, M.; Benek, O.; Cruz, C. D.; Tammela, P.; Tomašič, T.; Zidar, N.; Mašič, L. P.; Zega, A.; Stevenson, C. E. M.; Mundy, J. E. A.; Lawson, D. M.; Maxwell, A.; Kikelj, D.; Ilaš, J. Exploring the Chemical Space of Benzothiazole-Based DNA Gyrase B Inhibitors. ACS Med. Chem. Lett. 2020, 11, 2433– 2440, DOI: 10.1021/acsmedchemlett.0c0041686https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVKhtL3J&md5=03957be06e8e796ec89e3a4e05d996b4Exploring the Chemical Space of Benzothiazole-Based DNA Gyrase B InhibitorsSkok, Ziga; Barancokova, Michaela; Benek, Ondrej; Cruz, Cristina Durante; Tammela, Paivi; Tomasic, Tihomir; Zidar, Nace; Masic, Lucija Peterlin; Zega, Anamarija; Stevenson, Clare E. M.; Mundy, Julia E. A.; Lawson, David M.; Maxwell, Anthony; Kikelj, Danijel; Ilas, JanezACS Medicinal Chemistry Letters (2020), 11 (12), 2433-2440CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)We designed and synthesized a series of inhibitors of the bacterial enzymes DNA gyrase and DNA topoisomerase IV, based on our recently published benzothiazole-based inhibitor bearing an oxalyl moiety. To improve the antibacterial activity and retain potent enzymic activity, we systematically explored the chem. space. Several strategies of modification were followed: varying substituents on the pyrrole carboxamide moiety, alteration of the central scaffold, including variation of substitution position and, most importantly, modification of the oxalyl moiety. Compds. with acidic, basic, and neutral properties were synthesized. To understand the mechanism of action and binding mode, we have obtained a crystal structure of compd. 16a, bearing a primary amino group, in complex with the N-terminal domain of E. coli gyrase B (24 kDa) (PDB: 6YD9). Compd. 15a, with a low mol. wt. of 383 Da, potent inhibitory activity on E. coli gyrase (IC50 = 9.5 nM), potent antibacterial activity on E. faecalis (MIC = 3.13μM), and efflux impaired E. coli strain (MIC = 0.78μM), is an important contribution for the development of novel gyrase and topoisomerase IV inhibitors in Gram-neg. bacteria.
- 87Brvar, M.; Perdih, A.; Renko, M.; Anderluh, G.; Turk, D.; Solmajer, T. Structure-based discovery of substituted 4,5′-bithiazoles as novel DNA gyrase inhibitors. J. Med. Chem. 2012, 55, 6413– 6426, DOI: 10.1021/jm300395d87https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XptF2ns78%253D&md5=3701c7c3442de99bcaf8e76c51933991Structure-Based Discovery of Substituted 4,5'-Bithiazoles as Novel DNA Gyrase InhibitorsBrvar, Matjaz; Perdih, Andrej; Renko, Miha; Anderluh, Gregor; Turk, Dusan; Solmajer, TomJournal of Medicinal Chemistry (2012), 55 (14), 6413-6426CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bacterial DNA gyrase is a well-established and validated target for the development of novel antibacterials. Starting from the available structural information about the binding of the natural product inhibitor, clorobiocin, we identified a novel series of 4'-methyl-N2-phenyl-[4,5'-bithiazole]-2,2'-diamine inhibitors of gyrase B with a low micromolar inhibitory activity by implementing a two-step structure-based design procedure. This novel class of DNA gyrase inhibitors was extensively investigated by various techniques (differential scanning fluorimetry, surface plasmon resonance, and microscale thermophoresis). The binding mode of the potent inhibitor 18 was revealed by X-ray crystallog., confirming our initial in silico binding model. Furthermore, the high resoln. of the complex structure allowed for the placement of the Gly97-Ser108 flexible loop, thus revealing its role in binding of this class of compds. The crystal structure of the complex protein G24 and inhibitor 18 provides valuable information for further optimization of this novel class of DNA gyrase B inhibitors.
- 88Manchester, J. I.; Dussault, D. D.; Rose, J. A.; Boriack-Sjodin, P. A.; Uria-Nickelsen, M.; Ioannidis, G.; Bist, S.; Fleming, P.; Hull, K. G. Discovery of a novel azaindole class of antibacterial agents targeting the ATPase domains of DNA gyrase and Topoisomerase IV. Bioorg. Med. Chem. Lett. 2012, 22, 5150– 5156, DOI: 10.1016/j.bmcl.2012.05.12888https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVOqt7bL&md5=7c2e42ef7a21c7805736065e84d86a49Discovery of a novel azaindole class of antibacterial agents targeting the ATPase domains of DNA gyrase and Topoisomerase IVManchester, John I.; Dussault, Daemian D.; Rose, Jonathan A.; Boriack-Sjodin, P. Ann; Uria-Nickelsen, Maria; Ioannidis, Georgine; Bist, Shanta; Fleming, Paul; Hull, Kenneth G.Bioorganic & Medicinal Chemistry Letters (2012), 22 (15), 5150-5156CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The discovery and optimization of a series of bacterial topoisomerase inhibitors, e.g., I, is disclosed. Starting from a virtual screening hit, activity was optimized through a combination of structure-based design and phys. property optimization. Synthesis of fewer than a dozen compds. was required to achieve inhibition of the growth of methicillin-resistant Staphyloccus aureus at compd. concns. of 1.56 μM. These compds. simultaneously inhibit DNA gyrase and Topoisomerase IV at similar nanomolar concns., reducing the likelihood of the spontaneous occurrence of target-based mutations resulting in antibiotic resistance, an increasing threat in the treatment of serious infections.
- 89Gross, C. H.; Parsons, J. D.; Grossman, T. H.; Charifson, P. S.; Bellon, S.; Jernee, J.; Dwyer, M.; Chambers, S. P.; Markland, W.; Botfield, M.; Raybuck, S. A. Active–site residues of Escherichia coli DNA gyrase required in coupling ATP hydrolysis to DNA supercoiling and amino acid substitutions leading to novobiocin resistance. Antimicrob. Agents Chemother. 2003, 47, 1037– 1046, DOI: 10.1128/AAC.47.3.1037-1046.2003There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.4c00511.
Hit compound cytotoxicity against Caco-2 cells; cells were incubated with different concentrations of compounds (1, 2, 3, 8, 11, 14, G24, and G26) for 24 h and viability measured by MTT assay; all-atom RMSD plots for MD simulations of M. tuberculosis GyrB:inhibitor complexes; per-residue analysis of inhibitor:GyrB interactions; hydrogen bond interactions of known ATPase inhibitors with Asp79 in the M. smegmatis DNA gyrase ATP binding site; hits and leads obtained from three different virtual screening methods; previously characterized compounds identified from virtual similarity screening; details of fit parameters for IC50 determinations; hydrogen bonding of most active compounds (R-stereomers) to GyrB Asp79; and calculated binding free energies for binding of compounds 8, 11, and 14 to the wild type and mutant (D79A) GyrB subunits (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.