1
|
Ashwath P, Osiecki P, Weiner D, Via LE, Sarathy JP. Role of DNA Double-Strand Break Formation in Gyrase Inhibitor-Mediated Killing of Nonreplicating Persistent Mycobacterium tuberculosis in Caseum. ACS Infect Dis 2024; 10:3631-3639. [PMID: 39315541 PMCID: PMC11474946 DOI: 10.1021/acsinfecdis.4c00499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Tuberculosis is the leading cause of mortality by infectious agents worldwide. The necrotic debris, known as caseum, which accumulates in the center of pulmonary lesions and cavities is home to nonreplicating drug-tolerant Mycobacterium tuberculosis that presents a significant hurdle to achieving a fast and durable cure. Fluoroquinolones such as moxifloxacin are highly effective at killing this nonreplicating persistent bacterial population and boosting TB lesion sterilization. Fluoroquinolones target bacterial DNA gyrase, which catalyzes the negative supercoiling of DNA and relaxes supercoils ahead of replication forks. In this study, we investigated the potency of several other classes of gyrase inhibitors against M. tuberculosis in different states of replication. In contrast to fluoroquinolones, many other gyrase inhibitors kill only replicating bacterial cultures but produce negligible cidal activity against M. tuberculosis in ex vivo rabbit caseum. We demonstrate that while these inhibitors are capable of inhibiting M. tuberculosis gyrase DNA supercoiling activity, fluoroquinolones are unique in their ability to cleave double-stranded DNA at low micromolar concentrations. We hypothesize that double-strand break formation is an important driver of gyrase inhibitor-mediated bactericidal potency against nonreplicating persistent M. tuberculosis populations in the host. This study provides general insight into the lesion sterilization potential of different gyrase inhibitor classes and informs the development of more effective chemotherapeutic options against persistent mycobacterial infections.
Collapse
Affiliation(s)
- Priyanka Ashwath
- Center
for Discovery and Innovation, Hackensack
Meridian Health, 111 Ideation Way, Nutley, New Jersey 07110, United States
| | - Paulina Osiecki
- Center
for Discovery and Innovation, Hackensack
Meridian Health, 111 Ideation Way, Nutley, New Jersey 07110, United States
| | - Danielle Weiner
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, 33 North Drive, Bethesda, Maryland 20892, United States
- Tuberculosis
Imaging Program (TBIP), Division of Intramural Research, NIAID, NIH, 33 North Drive, Building 33, Bethesda, Maryland 20892, United States
| | - Laura E. Via
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, 33 North Drive, Bethesda, Maryland 20892, United States
- Tuberculosis
Imaging Program (TBIP), Division of Intramural Research, NIAID, NIH, 33 North Drive, Building 33, Bethesda, Maryland 20892, United States
| | - Jansy P. Sarathy
- Center
for Discovery and Innovation, Hackensack
Meridian Health, 111 Ideation Way, Nutley, New Jersey 07110, United States
- Department
of Medical Sciences, Hackensack Meridian
School of Medicine, 123
Metro Blvd, Nutley 07110 New Jersey, United
States
| |
Collapse
|
2
|
Bakker AT, Kotsogianni I, Avalos M, Punt JM, Liu B, Piermarini D, Gagestein B, Slingerland CJ, Zhang L, Willemse JJ, Ghimire LB, van den Berg RJHBN, Janssen APA, Ottenhoff THM, van Boeckel CAA, van Wezel GP, Ghilarov D, Martin NI, van der Stelt M. Discovery of isoquinoline sulfonamides as allosteric gyrase inhibitors with activity against fluoroquinolone-resistant bacteria. Nat Chem 2024; 16:1462-1472. [PMID: 38898213 PMCID: PMC11374673 DOI: 10.1038/s41557-024-01516-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/22/2024] [Indexed: 06/21/2024]
Abstract
Bacteria have evolved resistance to nearly all known antibacterials, emphasizing the need to identify antibiotics that operate via novel mechanisms. Here we report a class of allosteric inhibitors of DNA gyrase with antibacterial activity against fluoroquinolone-resistant clinical isolates of Escherichia coli. Screening of a small-molecule library revealed an initial isoquinoline sulfonamide hit, which was optimized via medicinal chemistry efforts to afford the more potent antibacterial LEI-800. Target identification studies, including whole-genome sequencing of in vitro selected mutants with resistance to isoquinoline sulfonamides, unanimously pointed to the DNA gyrase complex, an essential bacterial topoisomerase and an established antibacterial target. Using single-particle cryogenic electron microscopy, we determined the structure of the gyrase-LEI-800-DNA complex. The compound occupies an allosteric, hydrophobic pocket in the GyrA subunit and has a mode of action that is distinct from the clinically used fluoroquinolones or any other gyrase inhibitor reported to date. LEI-800 provides a chemotype suitable for development to counter the increasingly widespread bacterial resistance to fluoroquinolones.
Collapse
Affiliation(s)
- Alexander T Bakker
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Mariana Avalos
- Department of Molecular Biotechnology, Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Jeroen M Punt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Bing Liu
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Diana Piermarini
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Berend Gagestein
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Cornelis J Slingerland
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Le Zhang
- Department of Molecular Biotechnology, Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Joost J Willemse
- Department of Molecular Biotechnology, Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Leela B Ghimire
- Department of Molecular Microbiology, John Innes Centre, Norwich, UK
| | | | - Antonius P A Janssen
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Constant A A van Boeckel
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Gilles P van Wezel
- Department of Molecular Biotechnology, Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Dmitry Ghilarov
- Department of Molecular Microbiology, John Innes Centre, Norwich, UK.
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology, Leiden University, Leiden, the Netherlands.
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
3
|
Bakchi B, Maddipatla S, Gupta K, Singampalli A, Saxena D, Maitra R, Agnivesh PK, Kalia NP, Nanduri S, Chopra S, Yaddanapudi VM. Facile one-pot synthesis of N-pyridinylaminonaphthol derivatives and their antibacterial evaluation against multidrug-resistant Staphylococcus aureus. Arch Pharm (Weinheim) 2024:e2400358. [PMID: 39102220 DOI: 10.1002/ardp.202400358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 08/06/2024]
Abstract
The escalating severity of the menace posed by bacterial resistance has rendered the existing antibiotics less effective, thus necessitating the discovery of new antibacterial agents. The current study reports the exploration of substituted N-pyridinylaminonaphthols produced by a straightforward, one-pot multicomponent reaction process as antibacterial agents. The synthesized derivatives were assessed in vitro for their antibacterial properties against a panel of bacterial pathogens. The analogs 4b, 4g, 4h, 4i, 4j, 4l, 4r, and 4t exhibited potent inhibitory activity with minimum inhibitory concentration (MIC) values of 1-2 µg/mL. Notably, 4b, 4l, and 4t displayed an excellent selectivity index. Additionally, they were active against the multidrug-resistant bacterial strains, with 4l exhibiting the best activity against methicillin-resistant Staphylococcus aureus and vancomycin resistant staphylococcus aureus with a MIC of 1 µg/mL. 4l showed synergism with gentamycin and showed bactericidal property in a concentration-dependent manner. Furthermore, the molecule 4l inhibited the DNA gyrase supercoiling activity. Absorption, distribution, metabolism, excretion/toxicity parameters and pharmacokinetic properties were assessed via in silico techniques, which elucidate the potential mode of action. These findings demonstrate the potential of the N-pyridinylaminonaphthol derivatives as antibacterial agents against multidrug-resistant S. aureus.
Collapse
Affiliation(s)
- Bulti Bakchi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Sarvan Maddipatla
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Khushi Gupta
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Anuradha Singampalli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Deepanshi Saxena
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Rahul Maitra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Puja K Agnivesh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Nitin P Kalia
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Siddharth Chopra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Venkata M Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| |
Collapse
|
4
|
Collins J, Osheroff N. Gyrase and Topoisomerase IV: Recycling Old Targets for New Antibacterials to Combat Fluoroquinolone Resistance. ACS Infect Dis 2024; 10:1097-1115. [PMID: 38564341 PMCID: PMC11019561 DOI: 10.1021/acsinfecdis.4c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Beyond their requisite functions in many critical DNA processes, the bacterial type II topoisomerases, gyrase and topoisomerase IV, are the targets of fluoroquinolone antibacterials. These drugs act by stabilizing gyrase/topoisomerase IV-generated DNA strand breaks and by robbing the cell of the catalytic activities of these essential enzymes. Since their clinical approval in the mid-1980s, fluoroquinolones have been used to treat a broad spectrum of infectious diseases and are listed among the five "highest priority" critically important antimicrobial classes by the World Health Organization. Unfortunately, the widespread use of fluoroquinolones has been accompanied by a rise in target-mediated resistance caused by specific mutations in gyrase and topoisomerase IV, which has curtailed the medical efficacy of this drug class. As a result, efforts are underway to identify novel antibacterials that target the bacterial type II topoisomerases. Several new classes of gyrase/topoisomerase IV-targeted antibacterials have emerged, including novel bacterial topoisomerase inhibitors, Mycobacterium tuberculosis gyrase inhibitors, triazaacenaphthylenes, spiropyrimidinetriones, and thiophenes. Phase III clinical trials that utilized two members of these classes, gepotidacin (triazaacenaphthylene) and zoliflodacin (spiropyrimidinetrione), have been completed with positive outcomes, underscoring the potential of these compounds to become the first new classes of antibacterials introduced into the clinic in decades. Because gyrase and topoisomerase IV are validated targets for established and emerging antibacterials, this review will describe the catalytic mechanism and cellular activities of the bacterial type II topoisomerases, their interactions with fluoroquinolones, the mechanism of target-mediated fluoroquinolone resistance, and the actions of novel antibacterials against wild-type and fluoroquinolone-resistant gyrase and topoisomerase IV.
Collapse
Affiliation(s)
- Jessica
A. Collins
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Neil Osheroff
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Medicine (Hematology/Oncology), Vanderbilt
University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
5
|
Ruan S, Tu CH, Bourne CR. Friend or Foe: Protein Inhibitors of DNA Gyrase. BIOLOGY 2024; 13:84. [PMID: 38392303 PMCID: PMC10886550 DOI: 10.3390/biology13020084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/20/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024]
Abstract
DNA gyrase is essential for the successful replication of circular chromosomes, such as those found in most bacterial species, by relieving topological stressors associated with unwinding the double-stranded genetic material. This critical central role makes gyrase a valued target for antibacterial approaches, as exemplified by the highly successful fluoroquinolone class of antibiotics. It is reasonable that the activity of gyrase could be intrinsically regulated within cells, thereby helping to coordinate DNA replication with doubling times. Numerous proteins have been identified to exert inhibitory effects on DNA gyrase, although at lower doses, it can appear readily reversible and therefore may have regulatory value. Some of these, such as the small protein toxins found in plasmid-borne addiction modules, can promote cell death by inducing damage to DNA, resulting in an analogous outcome as quinolone antibiotics. Others, however, appear to transiently impact gyrase in a readily reversible and non-damaging mechanism, such as the plasmid-derived Qnr family of DNA-mimetic proteins. The current review examines the origins and known activities of protein inhibitors of gyrase and highlights opportunities to further exert control over bacterial growth by targeting this validated antibacterial target with novel molecular mechanisms. Furthermore, we are gaining new insights into fundamental regulatory strategies of gyrase that may prove important for understanding diverse growth strategies among different bacteria.
Collapse
Affiliation(s)
- Shengfeng Ruan
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA
| | - Chih-Han Tu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA
| | - Christina R Bourne
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA
| |
Collapse
|
6
|
Ommi O, Naiyaz Ahmad M, Gajula SNR, Wanjari P, Sau S, Agnivesh PK, Sahoo SK, Kalia NP, Sonti R, Nanduri S, Dasgupta A, Chopra S, Yaddanapudi VM. Synthesis and pharmacological evaluation of 1,3-diaryl substituted pyrazole based (thio)urea derivatives as potent antimicrobial agents against multi-drug resistant Staphylococcus aureus and Mycobacterium tuberculosis. RSC Med Chem 2023; 14:1296-1308. [PMID: 37484564 PMCID: PMC10357928 DOI: 10.1039/d3md00079f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/08/2023] [Indexed: 07/25/2023] Open
Abstract
The urgent development of newer alternatives has been deemed a panacea for tackling emerging antimicrobial resistance effectively. Herein, we report the design, synthesis, and biological evaluation of 1,3-diaryl substituted pyrazole-based urea and thiourea derivatives as antimicrobial agents. Preliminary screening results revealed that compound 7a (3,4-dichlorophenyl derivative) exhibited potent activity against S. aureus (MIC = 0.25 μg mL-1) and compound 7j (2,4-difluorophenyl derivative) against Mycobacterium tuberculosis (MIC = 1 μg mL-1). Compounds 7a and 7j were non-toxic to Vero cells with a favorable selectivity index of 40 and 200, respectively, and demonstrated good microsomal stability. Compound 7a exhibited equipotent activity (MIC = 0.25 μg mL-1) against various multidrug-resistant strains of S. aureus, which include various strains of MRSA and VRSA, and elicited bacteriostatic properties. In an enzymatic assay, 7a effectively inhibited DNA gyrase supercoiling activity at a concentration of 8 times MIC. Further, molecular modeling studies suggested that compound 7a binds at the active site of DNA gyrase with good affinity.
Collapse
Affiliation(s)
- Ojaswitha Ommi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Mohammad Naiyaz Ahmad
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 UP India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Parita Wanjari
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Shashikanta Sau
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Puja Kumari Agnivesh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Santosh Kumar Sahoo
- Department of Pharmaceutical Chemistry, GITAM School of Pharmacy, GITAM University Visakhapatnam 530045 India
| | - Nitin Pal Kalia
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| | - Arunava Dasgupta
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 UP India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Sidharth Chopra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road Lucknow 226031 UP India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Balanagar Hyderabad 500037 Telangana India
| |
Collapse
|
7
|
Qi Y, Liu Y, Zhang B, Wang M, Cao L, Song L, Jin N, Zhang H. Comparative antibacterial analysis of the anthraquinone compounds based on the AIM theory, molecular docking, and dynamics simulation analysis. J Mol Model 2022; 29:16. [PMID: 36547745 DOI: 10.1007/s00894-022-05406-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/04/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Hydroxyanthraquinones and anthraquinone glucoside derivatives are always considered as the active antibacterial components. METHODS Comparison of structure characteristics and antibacterial effect of these compounds was performed by applying quantum chemical calculations, atoms in molecules theory, molecular docking, and dynamics simulation procedure. Density functional theory calculation with B3LYP using 6-31G (d, p) basis set has been used to determine ground state molecular geometries. RESULTS The molecular geometric stability, electrostatic potential, frontier orbital energies, and topological properties were analyzed at the active site. Once glucose ring is introduced into the hydroxyanthraquinone rings, almost all of the positive molecular potentials are distributed among the hydroxyl hydrogen atoms of the glucose rings. In addition, low electron density ρ (r) and positive Laplacian value of the O-H bond of the anthraquinone glucoside are the evidences of the highly polarized and covalently decreased bonding interactions. The anthraquinone glucoside compounds have generally higher intermolecular binding energies than the corresponding aglycones due to the strong interaction between the glucose rings and the surrounding amino acids. Molecular dynamics simulations further explored the stability and dynamic behavior of the anthraquinone compound and protein complexes through RMSD, RMSF, SASA, and Rg. CONCLUSION The type of carboxyl, hydroxyl, and hydroxymethyl groups on phenyl ring and the substituent glucose rings is important to the interactions with the topoisomerase type II enzyme DNA gyrase B.
Collapse
Affiliation(s)
- Yanjiao Qi
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, 730000, Lanzhou, People's Republic of China.,Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in Universities of Gansu Province, 730000, Lanzhou, People's Republic of China
| | - Yue Liu
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in Universities of Gansu Province, 730000, Lanzhou, People's Republic of China
| | - Bo Zhang
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in Universities of Gansu Province, 730000, Lanzhou, People's Republic of China
| | - Mingyang Wang
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, 730000, Lanzhou, People's Republic of China
| | - Long Cao
- Department of Chemical Engineering, Northwest Minzu University, 730124, Lanzhou, People's Republic of China
| | - Li Song
- Gansu Hualing Dairy Co. LTD., Lanzhou, People's Republic of China
| | - Nengzhi Jin
- Gansu Province Computing Center, 730000, Lanzhou, People's Republic of China
| | - Hong Zhang
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, 730000, Lanzhou, People's Republic of China.
| |
Collapse
|
8
|
Alfonso EE, Troche R, Deng Z, Annamalai T, Chapagain P, Tse-Dinh YC, Leng F. Potent Inhibition of Bacterial DNA Gyrase by Digallic Acid and Other Gallate Derivatives. ChemMedChem 2022; 17:e202200301. [PMID: 36161274 PMCID: PMC9742164 DOI: 10.1002/cmdc.202200301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/23/2022] [Indexed: 01/14/2023]
Abstract
Bacterial DNA gyrase, an essential enzyme, is a validated target for discovering and developing new antibiotics. Here we screened a pool of polyphenols and discovered that digallic acid is a potent DNA gyrase inhibitor. We also found that several food additives based on gallate, such as dodecyl gallate, potently inhibit bacterial DNA gyrase. Interestingly, the IC50 of these gallate derivatives against DNA gyrase is correlated with the length of hydrocarbon chain connecting to the gallate. These new bacterial DNA gyrase inhibitors are ATP competitive inhibitors of DNA gyrase. Our results also show that digallic acid and certain gallate derivatives potently inhibit E. coli DNA topoisomerase IV. Several gallate derivatives have strong antimicrobial activities against Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA). This study provides a solid foundation for the design and synthesis of gallate-based DNA gyrase inhibitors that may be used to combat antibacterial resistance.
Collapse
Affiliation(s)
- Eddy E Alfonso
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Rogelio Troche
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Zifang Deng
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Thirunavukkarasu Annamalai
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Prem Chapagain
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
- Department of Physics, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Yuk-Ching Tse-Dinh
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| | - Fenfei Leng
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL, 33199, USA
| |
Collapse
|
9
|
Alfonso EE, Deng Z, Boaretto D, Hood BL, Vasile S, Smith LH, Chambers JW, Chapagain P, Leng F. Novel and Structurally Diversified Bacterial DNA Gyrase Inhibitors Discovered through a Fluorescence-Based High-Throughput Screening Assay. ACS Pharmacol Transl Sci 2022; 5:932-944. [PMID: 36268121 PMCID: PMC9578135 DOI: 10.1021/acsptsci.2c00113] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Indexed: 12/25/2022]
Abstract
Bacterial DNA gyrase, a type IIA DNA topoisomerase that plays an essential role in bacterial DNA replication and transcription, is a clinically validated target for discovering and developing new antibiotics. In this article, based on a supercoiling-dependent fluorescence quenching (SDFQ) method, we developed a high-throughput screening (HTS) assay to identify inhibitors targeting bacterial DNA gyrase and screened the National Institutes of Health's Molecular Libraries Small Molecule Repository library containing 370,620 compounds in which 2891 potential gyrase inhibitors have been identified. According to these screening results, we acquired 235 compounds to analyze their inhibition activities against bacterial DNA gyrase using gel- and SDFQ-based DNA gyrase inhibition assays and discovered 155 new bacterial DNA gyrase inhibitors with a wide structural diversity. Several of them have potent antibacterial activities. These newly discovered gyrase inhibitors include several DNA gyrase poisons that stabilize the gyrase-DNA cleavage complexes and provide new chemical scaffolds for the design and synthesis of bacterial DNA gyrase inhibitors that may be used to combat multidrug-resistant bacterial pathogens. Additionally, this HTS assay can be applied to screen inhibitors against other DNA topoisomerases.
Collapse
Affiliation(s)
- Eddy E. Alfonso
- Biomolecular
Sciences Institute, Florida International
University, Miami, Florida 33199, United States
- Department
of Chemistry and Biochemistry, Florida International
University, Miami, Florida 33199, United
States
| | - Zifang Deng
- Biomolecular
Sciences Institute, Florida International
University, Miami, Florida 33199, United States
- Department
of Chemistry and Biochemistry, Florida International
University, Miami, Florida 33199, United
States
| | - Daniel Boaretto
- Biomolecular
Sciences Institute, Florida International
University, Miami, Florida 33199, United States
- Department
of Chemistry and Biochemistry, Florida International
University, Miami, Florida 33199, United
States
| | - Becky L. Hood
- Conrad
Prebys Center for Chemical Genomics, Sanford
Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Stefan Vasile
- Conrad
Prebys Center for Chemical Genomics, Sanford
Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Layton H. Smith
- Conrad
Prebys Center for Chemical Genomics, Sanford
Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Jeremy W. Chambers
- Biomolecular
Sciences Institute, Florida International
University, Miami, Florida 33199, United States
- Department
of Environmental Health Sciences, Florida
International University, Miami, Florida 33199, United States
| | - Prem Chapagain
- Biomolecular
Sciences Institute, Florida International
University, Miami, Florida 33199, United States
- Department
of Physics, Florida International University, Miami, Florida 33199, United States
| | - Fenfei Leng
- Biomolecular
Sciences Institute, Florida International
University, Miami, Florida 33199, United States
- Department
of Chemistry and Biochemistry, Florida International
University, Miami, Florida 33199, United
States
| |
Collapse
|
10
|
New potent ciprofloxacin-uracil conjugates as DNA gyrase and topoisomerase IV inhibitors against methicillin-resistant Staphylococcus aureus. Bioorg Med Chem 2022; 73:117004. [PMID: 36148773 DOI: 10.1016/j.bmc.2022.117004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022]
Abstract
A series of ciprofloxacin-uracil conjugates 5a-t were synthesized and identified by 1H NMR, 13C NMR, mass spectroscopy and elemental analyses. The antibacterial results revealed that the new derivatives exhibited better activity against Gram-positive than the Gram-negative strains; most of the target compounds exhibited good activities against S. aureus ATCC 6538. Compounds 5b and 5g possess the highest activities with MICs of 1.25 and 2.37 µM, respectively, which are more potent than the parent drug ciprofloxacin, MIC, 7.58 µM. In addition, they also exhibited potent activities against MRSA AUMC 261 with MICs, 0.031 and 0.046 µM respectively, higher than ciprofloxacin with MIC, 0.57 µM. Moreover, compounds 5b and 5g showed potent inhibitory activities against DNA gyrase (IC50 = 1.72 and 5.72 µM) and topoisomerase IV (4.36 and 7.77 µM) compared to ciprofloxacin with IC50 values 0.66 and 8.16 µM, respectively. The molecular docking study revealed that compounds 5b and 5g may formed stable interaction with the active sites of DNA gyrase and topoisomerase IV similar to ciprofloxacin. Hence, 5b and 5g are considered promising antibacterial candidated against MRSA AUMC 261 strains that requires further optimization.
Collapse
|
11
|
Juliana Martins F, Savacini Sagrillo F, Josiane Vinturelle Medeiros R, Gonçalves de Souza A, Rodrigues Pinto Costa A, Silva Novais J, Alves Miceli L, R Campos V, Marie Sá Figueiredo A, Claudia Cunha A, Lidmar von Ranke N, Lamim Bello M, de A Abrahim-Vieira B, M T De Souza A, A Ratcliffe N, da Costa Santos Boechat F, Cecília Bastos Vieira de Souza M, Rangel Rodrigues C, Carla Castro H. Evaluation of biological activities of quinone-4-oxoquinoline derivatives against pathogens of clinical importance. Curr Top Med Chem 2022; 22:973-991. [PMID: 35524665 DOI: 10.2174/1568026622666220504124710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Microbial resistance has become a worldwide public health problem, and may lead to morbidity and mortality in affected patients. OBJECTIVE Therefore, this work aimed to evaluate the antibacterial activity of quinone-4-oxoquinoline derivatives. METHOD These derivatives were evaluated against Gram-positive and Gram-negative bacteria by their antibacterial activity, anti-biofilm, and hemolytic activities and by in silico assays. RESULTS The quinone-4-oxoquinoline derivatives presented broad-spectrum antibacterial activities, and in some cases were more active than commercially available reference drugs. These compounds also inhibited bacterial adhesion and the assays revealed seven non-hemolytic derivatives. The derivatives seem to cause damage to the bacterial cell membrane and those containing the carboxyl group at the C-3 position of the 4-quinolonic nucleus were more active than those containing a carboxyethyl group. CONCLUSION The isoquinoline-5,8-dione nucleus also favored antimicrobial activity. The study showed that the target of the derivatives must be a non-conventional hydrophobic allosteric binding pocket on the DNA gyrase enzyme.
Collapse
Affiliation(s)
- Francislene Juliana Martins
- Federal Fluminense University, Biology Institute, Postgraduate Program in Science and Biotechnology, Niterói, Rio de Janeiro, Brazil
| | - Fernanda Savacini Sagrillo
- Federal Fluminense University, Chemistry Institute, Department of Organic Chemistry, Niterói, Rio de Janeiro, Brazil
| | | | - Alan Gonçalves de Souza
- Federal Fluminense University, Chemistry Institute, Department of Organic Chemistry, Niterói, Rio de Janeiro, Brazil
| | - Amanda Rodrigues Pinto Costa
- Federal Fluminense University, Chemistry Institute, Department of Organic Chemistry, Niterói, Rio de Janeiro, Brazil
| | - Juliana Silva Novais
- Federal Fluminense University, Medical School, Postgraduate in Pathology, Niterói, Rio de Janeiro, Brazil.,Universidade Estácio de Sá (UNESA), Faculdade de Farmácia, São Gonçalo, Rio de Janeiro, Brazil
| | - Leonardo Alves Miceli
- Federal University of Rio de Janeiro, Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Rio de Janeiro, Brazil
| | - Vinícius R Campos
- Federal Fluminense University, Chemistry Institute, Department of Organic Chemistry, Niterói, Rio de Janeiro, Brazil
| | - Agnes Marie Sá Figueiredo
- Federal University of Rio de Janeiro, Microbiology Institute Professor Paulo Goes, Department of Medical Microbiology, Rio de Janeiro, Brazil
| | - Anna Claudia Cunha
- Federal Fluminense University, Chemistry Institute, Department of Organic Chemistry, Niterói, Rio de Janeiro, Brazil
| | - Natalia Lidmar von Ranke
- Federal University of Rio de Janeiro, Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Rio de Janeiro, Brazil
| | - Murilo Lamim Bello
- Federal University of Rio de Janeiro, Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Rio de Janeiro, Brazil
| | - Bárbara de A Abrahim-Vieira
- Federal University of Rio de Janeiro, Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Rio de Janeiro, Brazil
| | - Alessandra M T De Souza
- Federal University of Rio de Janeiro, Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Rio de Janeiro, Brazil
| | - Norman A Ratcliffe
- Department of Biosciences, College of Science Swansea University, SA2 8PP. UK
| | | | | | - Carlos Rangel Rodrigues
- Federal University of Rio de Janeiro, Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Rio de Janeiro, Brazil
| | - Helena Carla Castro
- Federal Fluminense University, Biology Institute, Postgraduate Program in Science and Biotechnology, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Purushothaman M, Dhar SK, Natesh R. Role of unique loops in oligomerization and ATPase function of Plasmodium falciparum gyrase B. Protein Sci 2022; 31:323-332. [PMID: 34716632 PMCID: PMC8820116 DOI: 10.1002/pro.4217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 02/03/2023]
Abstract
DNA gyrase is an ATP dependent Type IIA topoisomerase that is unique to prokaryotes. Interestingly DNA gyrase has also been found in the apicoplasts of apicomplexan parasites like Plasmodium falciparum (Pf) the causative agent of Malaria. Gyrase B (GyrB), a subunit of gyrase A2 B2 complex has an N-terminal domain (GyrBN) which is endowed with ATPase activity. We reported earlier that PfGyrB exhibits ATP-independent dimerization unlike its bacterial counterparts. Here we report the role of two unique regions (L1 and L2) identified in PfGyrBN. Deletions of L1 alone (PfGyrBNΔL1), or L1 and L2 together (PfGyrBNΔL1ΔL2) have indicated that these regions may play an important role in ATPase activity and the oligomeric state of PfGyrBN. Our experiments show that the deletion of L1 region disrupts the dimer interface of PfGyrBN and reduces its ATPase activity. Further through ITC experiments we show that the binding affinity of ATP to PfGyrBN is reduced upon the deletion of L1 region. We have observed a reduction in ATPase activity for of all three proteins PfGyrBN, PfGyrBNΔL1, and PfGyrBNΔL1ΔL2 in presence of coumermycin. Our results suggests that L1 region of PfGyrBN is likely to be functionally important and may provide a unique dimer interface that affects its enzymatic activity. Since deletion of L1 region decreases the affinity of ATP to the protein, this region can be targeted toward designing novel inhibitors of ATP hydrolysis.
Collapse
Affiliation(s)
- Monica Purushothaman
- School of BiologyIndian Institute of Science Education and Research ThiruvananthapuramThiruvananthapuramKeralaIndia
| | - Suman Kumar Dhar
- Special Centre of Molecular MedicineJawaharlal Nehru UniversityNew DelhiIndia
| | - Ramanathan Natesh
- School of BiologyIndian Institute of Science Education and Research ThiruvananthapuramThiruvananthapuramKeralaIndia
| |
Collapse
|