1
|
Yazdanpanah S, Shafiekhani M, Emami M, Khodadadi H, Pakshir K, Zomorodian K. Exploring the anti-biofilm and gene regulatory effects of anti-inflammatory drugs on Candida albicans. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03727-y. [PMID: 39731595 DOI: 10.1007/s00210-024-03727-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024]
Abstract
Researchers have repurposed several existing anti-inflammatory drugs as potential antifungal agents in recent years. So, this study aimed to investigate the effects of anti-inflammatory drugs on the growth, biofilm formation, and expression of genes related to morphogenesis and pathogenesis in Candida albicans. The minimum inhibitory concentration (MIC) of anti-inflammatory drugs was assessed using the broth microdilution method. Biofilm formation in C. albicans was evaluated using XTT reduction assay following exposure to different concentrations of drugs. Additionally, the expression of adhesin-related genes (ALS1, ALS3), hyphal cell wall specific genes (EAP1, HWP1), secreted aspartyl proteinase (SAP4, SAP6), and morphogenesis pathway regulatory gene (EFG1) was analyzed using quantitative RT-PCR. Betamethasone and dexamethasone markedly inhibited C. albicans biofilm formation by up to 80% at a concentration of 2 mg/mL. Moreover, the inhibition of C. albicans biofilm formation was significant at concentrations ranging from 0.6 to 10 mg/mL for piroxicam and from 0.75 to 12 mg/mL for diclofenac. The expression of key genes involved in biofilm formation including EFG1, HWP1, and ALS3 was all downregulated under hyphae-inducing conditions. Moreover, the expression proteinase genes of C. albicans were upregulated following exposure with corticosteroids. The data obtained provides valuable insights into the antifungal potential of anti-inflammatory drugs. Our novel findings indicate the downregulation of several Candida genes that are crucial for morphogenesis, pathogenesis, and biofilm formation. However, further research is necessary to fully elucidate the clinical applications and effectiveness of anti-inflammatory drugs as alternative or adjunctive therapies for Candida infections.
Collapse
Affiliation(s)
- Somayeh Yazdanpanah
- Department of Medical Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Shafiekhani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Emami
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Khodadadi
- Department of Medical Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Keyvan Pakshir
- Department of Medical Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Basic Sciences in Infectious Diseases Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamiar Zomorodian
- Department of Medical Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Basic Sciences in Infectious Diseases Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Butler MS, Vollmer W, Goodall ECA, Capon RJ, Henderson IR, Blaskovich MAT. A Review of Antibacterial Candidates with New Modes of Action. ACS Infect Dis 2024; 10:3440-3474. [PMID: 39018341 PMCID: PMC11474978 DOI: 10.1021/acsinfecdis.4c00218] [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: 03/17/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
There is a lack of new antibiotics to combat drug-resistant bacterial infections that increasingly threaten global health. The current pipeline of clinical-stage antimicrobials is primarily populated by "new and improved" versions of existing antibiotic classes, supplemented by several novel chemical scaffolds that act on traditional targets. The lack of fresh chemotypes acting on previously unexploited targets (the "holy grail" for new antimicrobials due to their scarcity) is particularly unfortunate as these offer the greatest opportunity for innovative breakthroughs to overcome existing resistance. In recognition of their potential, this review focuses on this subset of high value antibiotics, providing chemical structures where available. This review focuses on candidates that have progressed to clinical trials, as well as selected examples of promising pioneering approaches in advanced stages of development, in order to stimulate additional research aimed at combating drug-resistant infections.
Collapse
Affiliation(s)
- Mark S. Butler
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Waldemar Vollmer
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Emily C. A. Goodall
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Robert J. Capon
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Ian R. Henderson
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Mark A. T. Blaskovich
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
3
|
Mazumder S, Bindu S, Debsharma S, Bandyopadhyay U. Induction of mitochondrial toxicity by non-steroidal anti-inflammatory drugs (NSAIDs): The ultimate trade-off governing the therapeutic merits and demerits of these wonder drugs. Biochem Pharmacol 2024; 228:116283. [PMID: 38750902 DOI: 10.1016/j.bcp.2024.116283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are most extensively used over-the-counter FDA-approved analgesic medicines for treating inflammation, musculoskeletal pain, arthritis, pyrexia and menstrual cramps. Moreover, aspirin is widely used against cardiovascular complications. Owing to their non-addictive nature, NSAIDs are also commissioned as safer opioid-sparing alternatives in acute trauma and post-surgical treatments. In fact, therapeutic spectrum of NSAIDs is expanding. These "wonder-drugs" are now repurposed against lung diseases, diabetes, neurodegenerative disorders, fungal infections and most notably cancer, due to their efficacy against chemoresistance, radio-resistance and cancer stem cells. However, prolonged NSAID treatment accompany several adverse effects. Mechanistically, apart from cyclooxygenase inhibition, NSAIDs directly target mitochondria to induce cell death. Interestingly, there are also incidences of dose-dependent effects where NSAIDs are found to improve mitochondrial health thereby suggesting plausible mitohormesis. While mitochondria-targeted effects of NSAIDs are discretely studied, a comprehensive account emphasizing the multiple dimensions in which NSAIDs affect mitochondrial structure-function integrity, leading to cell death, is lacking. This review discusses the current understanding of NSAID-mitochondria interactions in the pathophysiological background. This is essential for assessing the risk-benefit trade-offs of NSAIDs for judiciously strategizing NSAID-based approaches to manage pain and inflammation as well as formulating effective anti-cancer strategies. We also discuss recent developments constituting selective mitochondria-targeted NSAIDs including theranostics, mitocans, chimeric small molecules, prodrugs and nanomedicines that rationally optimize safer application of NSAIDs. Thus, we present a comprehensive understanding of therapeutic merits and demerits of NSAIDs with mitochondria at its cross roads. This would help in NSAID-based disease management research and drug development.
Collapse
Affiliation(s)
- Somnath Mazumder
- Department of Zoology, Raja Peary Mohan College, 1 Acharya Dhruba Pal Road, Uttarpara, West Bengal 712258, India
| | - Samik Bindu
- Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal 736101, India
| | - Subhashis Debsharma
- Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Uday Bandyopadhyay
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, West Bengal, India.
| |
Collapse
|
4
|
Okpala OE, Rondevaldova J, Osei-Owusu H, Kudera T, Kokoskova T, Kokoska L. Susceptibility of Staphylococcus aureus to Anti-Inflammatory Drugs with a Focus on the Combinatory Effect of Celecoxib with Oxacillin In Vitro. Molecules 2024; 29:3665. [PMID: 39125072 PMCID: PMC11314137 DOI: 10.3390/molecules29153665] [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: 06/19/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Musculoskeletal infections (MIs) are among the most difficult-to-treat staphylococcal diseases due to antibiotic resistance. This has encouraged the development of innovative strategies, such as combination therapy, to combat MI. The aim of this study was to investigate the in vitro antistaphylococcal activity of anti-inflammatory drugs and the combined antimicrobial effect of celecoxib and oxacillin. The minimum inhibitory concentrations (MICs) of 17 anti-inflammatory drugs against standard strains and clinical isolates of S. aureus, including methicillin-resistant strains (MRSAs), were determined using the broth microdilution method. The fractional inhibitory concentration indices (FICIs) were evaluated using checkerboard assays. Celecoxib produced the most potent antistaphylococcal effect against all tested strains (MICs ranging from 32 to 64 mg/L), followed by that of diacerein against MRSA3 and MRSA ATCC 33592 (MIC 64 mg/L). Several synergistic effects were observed against the tested S. aureus strains, including MRSA (FICI ranging from 0.087 to 0.471). The strongest synergistic interaction (FICI 0.087) was against MRSA ATCC 33592 at a celecoxib concentration of 2 mg/L, with a 19-fold oxacillin MIC reduction (from 512 to 26.888 mg/L). This is the first report on the combined antistaphylococcal effect of celecoxib and oxacillin. These findings suggest celecoxib and its combination with oxacillin as perspective agents for research focused on the development of novel therapies for MI caused by S. aureus. This study further indicates that celecoxib could resensitize certain MRSA strains, in some cases, to be susceptible to β-lactams (e.g., oxacillin) that were not previously tested. It is essential to mention that the in vitro concentrations of anti-inflammatory drugs are higher than those typically obtained in patients. Therefore, an alternative option for its administration could be the use of a drug delivery system for the controlled slow release from an implant at the infection site.
Collapse
Affiliation(s)
- Onyedika Emmanuel Okpala
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| | - Johana Rondevaldova
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| | - Hayford Osei-Owusu
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| | - Tomas Kudera
- Drift-Food Research Centre, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic;
| | - Tersia Kokoskova
- Department of Animal Science and Food Processing, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic;
| | - Ladislav Kokoska
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamycka 129, Suchdol, 165 00 Prague, Czech Republic; (O.E.O.); (J.R.); (H.O.-O.)
| |
Collapse
|
5
|
Bakht P, Ijaz M, Iqbal MZ, Aslam HB, Rehman A. Repurposing of non-steroidal anti-inflammatory drugs for combination therapies to combat multidrug-resistant S. aureus of bovine reproductive tract origin. Vet Res Commun 2024; 48:1497-1510. [PMID: 38347266 DOI: 10.1007/s11259-024-10322-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/31/2024] [Indexed: 06/04/2024]
Abstract
Multidrug-resistant bacteria have become the predominant etiology in bovine female reproductive tract infections and thus require effective treatment approaches. The main goal of this study was the molecular detection of mecA, blaZ, tetK, and aacA-aphD genes in Staphylococcus aureus (S. aureus) responsible for methicillin, beta-lactam, tetracycline, and aminoglycoside resistance respectively. Phylogenetic analysis was conducted to check the homology of staphylococcal genes with NCBI sequences. The in-vitro efficacy of non-steroidal anti-inflammatory drugs (NSAIDs) in combination therapies against MDR S. aureus was evaluated using well diffusion assay and checkerboard method. Vaginal swab samples (n = 384) collected from bovines suffering from endometritis, pyometra, and retained placenta were tested for S. aureus. Results showed a 17.96% overall prevalence. Both phenotypic and genotypic resistance was observed among S. aureus isolates with 50.72% and 37.68% isolates being confirmed as methicillin-resistant (MRSA), 36.23% and 18.84% isolates exhibiting beta-lactam, 40.58%, and 27.54% isolates showing tetracycline, and 33.33% and 36.23% isolates showing aminoglycosides resistance based on disc diffusion and gene confirmation, respectively. Phylogenetic analysis indicated homology with previously reported Pakistani isolates suggesting the possibility of MDR S. aureus transmission within and between animals. Synergy testing indicated that combinations of ceftriaxone-ketoprofen (153.77%), ceftriaxone-meloxicam (149.55%), amoxiclav-flunixin meglumine (106.06%), and oxytetracycline-flunixin meglumine (104.47%) showed synergy on well diffusion assay. Based on the fractional inhibitory concentration index by checkerboard method, oxytetracycline-meloxicam and gentamicin-ketoprofen combinations exhibited synergistic interaction. In conclusion, MDR S. aureus resistance was mitigated in-vitro through the combination of antibiotics (oxytetracycline, gentamicin) with NSAIDs (meloxicam, ketoprofen) that could be used to create therapeutic strategies for bovine reproductive issues.
Collapse
Affiliation(s)
- Painda Bakht
- Department of Veterinary Medicine, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Ijaz
- Department of Veterinary Medicine, University of Veterinary and Animal Sciences, Lahore, Pakistan.
| | - Muhammad Zahid Iqbal
- Department of Veterinary Medicine, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Hassaan Bin Aslam
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Abdul Rehman
- Department of Theriogenology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| |
Collapse
|
6
|
Simonsen S, Søgaard CK, Olsen JG, Otterlei M, Kragelund BB. The bacterial DNA sliding clamp, β-clamp: structure, interactions, dynamics and drug discovery. Cell Mol Life Sci 2024; 81:245. [PMID: 38814467 PMCID: PMC11139829 DOI: 10.1007/s00018-024-05252-w] [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: 03/14/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
DNA replication is a tightly coordinated event carried out by a multiprotein replication complex. An essential factor in the bacterial replication complex is the ring-shaped DNA sliding clamp, β-clamp, ensuring processive DNA replication and DNA repair through tethering of polymerases and DNA repair proteins to DNA. β -clamp is a hub protein with multiple interaction partners all binding through a conserved clamp binding sequence motif. Due to its central role as a DNA scaffold protein, β-clamp is an interesting target for antimicrobial drugs, yet little effort has been put into understanding the functional interactions of β-clamp. In this review, we scrutinize the β-clamp structure and dynamics, examine how its interactions with a plethora of binding partners are regulated through short linear binding motifs and discuss how contexts play into selection. We describe the dynamic process of clamp loading onto DNA and cover the recent advances in drug development targeting β-clamp. Despite decades of research in β-clamps and recent landmark structural insight, much remains undisclosed fostering an increased focus on this very central protein.
Collapse
Affiliation(s)
- Signe Simonsen
- Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
- Structural Biology and NMR Laboratory, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Caroline K Søgaard
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Johan G Olsen
- Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
- Structural Biology and NMR Laboratory, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
- Department of Biology, REPIN, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Marit Otterlei
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Birthe B Kragelund
- Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark.
- Structural Biology and NMR Laboratory, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark.
- Department of Biology, REPIN, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark.
| |
Collapse
|
7
|
Soliman AM, El-Sagheir AMK, Thabet MM, Abdel Hakiem AF, Aboraia AS. Synthesis, characterization, molecular modeling studies, and biological evaluation of metal piroxicam complexes (M = Ni(II), Pt(IV), Pd(II), Ag(I)) as antibacterial and anticancer agents. Drug Dev Res 2024; 85:e22156. [PMID: 38355931 DOI: 10.1002/ddr.22156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/01/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Four piroxicam metal complexes; NiL2 , PtL2 , PdL2 , and AgL were synthesized and characterized by different techniques with enhanced antibacterial and anticancer activity. Regarding in vitro antimicrobial activity, complex NiL2 displayed potent antibacterial effect against Escherichia coli and Pseudomonas aeruginosa that was 1.9-folds higher than piroxicam (minimum inhibitory concentration [MIC] = 31.85, 65.32 µM), respectively. In case of G+ve bacteria, complex PtL2 had potent activity on Staphylococcus aureus which was 2.1-folds higher than piroxicam (MIC = 43.12 µM), while activity of complex AgL against Enterococcus faecalis was threefolds higher than piroxicam (MIC = 74.57 µM. Complexes PtL2 and PdL2 exhibited higher inhibition of DNA gyrase than piroxicam (IC50 = 6.21 µM) in the range of 1.9-1.7-folds. The in vitro antiproliferative activity depicted that all investigated complexes showed better cytotoxic effect than piroxicam, specifically Pt and Pd complexes which had lower IC50 values than piroxicam on human liver cancer cell line HepG2 by 1.8 and 1.7-folds, respectively. While Pd and Ag complexes showed 2 and 1.6-folds better effect on human colon cancer cell line HT-29 compared with piroxicam. Molecular modeling studies including docking on Stranded DNA Duplex (1juu) and DNA gyrase enzyme (1kzn) that gave good insight about interaction of complexes with target molecules, calculation of electrostatic potential map and global reactivity descriptors were performed.
Collapse
Affiliation(s)
- Aya M Soliman
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Ahmed M K El-Sagheir
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
- Division of Pharmaceutical Chemistry and Technology, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Momen M Thabet
- Department of Microbiology and Immunology, Faculty of Pharmacy, South Valley University, Qena, Egypt
| | | | - Ahmed S Aboraia
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
8
|
Compain G, Monsarrat C, Blagojevic J, Brillet K, Dumas P, Hammann P, Kuhn L, Martiel I, Engilberge S, Oliéric V, Wolff P, Burnouf DY, Wagner J, Guichard G. Peptide-Based Covalent Inhibitors Bearing Mild Electrophiles to Target a Conserved His Residue of the Bacterial Sliding Clamp. JACS AU 2024; 4:432-440. [PMID: 38425897 PMCID: PMC10900491 DOI: 10.1021/jacsau.3c00572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 03/02/2024]
Abstract
Peptide-based covalent inhibitors targeted to nucleophilic protein residues have recently emerged as new modalities to target protein-protein interactions (PPIs) as they may provide some benefits over more classic competitive inhibitors. Covalent inhibitors are generally targeted to cysteine, the most intrinsically reactive amino acid residue, and to lysine, which is more abundant at the surface of proteins but much less frequently to histidine. Herein, we report the structure-guided design of targeted covalent inhibitors (TCIs) able to bind covalently and selectively to the bacterial sliding clamp (SC), by reacting with a well-conserved histidine residue located on the edge of the peptide-binding pocket. SC is an essential component of the bacterial DNA replication machinery, identified as a promising target for the development of new antibacterial compounds. Thermodynamic and kinetic analyses of ligands bearing different mild electrophilic warheads confirmed the higher efficiency of the chloroacetamide compared to Michael acceptors. Two high-resolution X-ray structures of covalent inhibitor-SC adducts were obtained, revealing the canonical orientation of the ligand and details of covalent bond formation with histidine. Proteomic studies were consistent with a selective SC engagement by the chloroacetamide-based TCI. Finally, the TCI of SC was substantially more active than the parent noncovalent inhibitor in an in vitro SC-dependent DNA synthesis assay, validating the potential of the approach to design covalent inhibitors of protein-protein interactions targeted to histidine.
Collapse
Affiliation(s)
- Guillaume Compain
- Univ.
Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, 2 Rue Robert Escarpit, F-33607 Pessac, France
| | - Clément Monsarrat
- Univ.
Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, 2 Rue Robert Escarpit, F-33607 Pessac, France
| | - Julie Blagojevic
- Université
de Strasbourg, CNRS, FR1589, Plateforme Protéomique Strasbourg
Esplanade, 2 Allée K. Roentgen, 67084 Strasbourg, France
| | - Karl Brillet
- Université
de Strasbourg, CNRS, Architecture et Réactivité de l’ARN,
UPR 9002, Institut de Biologie Moléculaire et Cellulaire du
CNRS, 2 Allée
K. Roentgen, 67084 Strasbourg, France
| | - Philippe Dumas
- Department
of Integrative Structural Biology, IGBMC, Strasbourg University, ESBS, 1 Rue Laurent Fries, 67404 Illkirch, Cedex, France
| | - Philippe Hammann
- Université
de Strasbourg, CNRS, FR1589, Plateforme Protéomique Strasbourg
Esplanade, 2 Allée K. Roentgen, 67084 Strasbourg, France
| | - Lauriane Kuhn
- Université
de Strasbourg, CNRS, FR1589, Plateforme Protéomique Strasbourg
Esplanade, 2 Allée K. Roentgen, 67084 Strasbourg, France
| | - Isabelle Martiel
- Swiss
Light Source (SLS), Paul Scherrer Institute
(PSI), 5232 Villigen-PSI, Switzerland
| | - Sylvain Engilberge
- Swiss
Light Source (SLS), Paul Scherrer Institute
(PSI), 5232 Villigen-PSI, Switzerland
| | - Vincent Oliéric
- Swiss
Light Source (SLS), Paul Scherrer Institute
(PSI), 5232 Villigen-PSI, Switzerland
| | - Philippe Wolff
- Université
de Strasbourg, CNRS, Architecture et Réactivité de l’ARN,
UPR 9002, Institut de Biologie Moléculaire et Cellulaire du
CNRS, 2 Allée
K. Roentgen, 67084 Strasbourg, France
| | - Dominique Y. Burnouf
- Université
de Strasbourg, CNRS, Architecture et Réactivité de l’ARN,
UPR 9002, Institut de Biologie Moléculaire et Cellulaire du
CNRS, 2 Allée
K. Roentgen, 67084 Strasbourg, France
| | - Jérôme Wagner
- Université
de Strasbourg, CNRS, Architecture et Réactivité de l’ARN,
UPR 9002, Institut de Biologie Moléculaire et Cellulaire du
CNRS, 2 Allée
K. Roentgen, 67084 Strasbourg, France
| | - Gilles Guichard
- Univ.
Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, 2 Rue Robert Escarpit, F-33607 Pessac, France
| |
Collapse
|
9
|
Ghara S, Bera S, Dastidar P. Antibacterial Hydrogel as a Self-Drug-Delivery System Derived from Zn(II)-bis-imidazole/NSAID-Based Organic-Inorganic Hybrids. ACS APPLIED BIO MATERIALS 2023; 6:4749-4763. [PMID: 37864581 DOI: 10.1021/acsabm.3c00525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
A skin wound is prone to bacterial infection and growth. An antibacterial topical hydrogel that can act as a self-drug-delivery (SDD) system is reported here. Two bidentate ligands (L2/L1) derived from imidazole/benzimidazole derivatives when reacted with Zn(NO3)2 and a series of nonsteroidal-anti-inflammatory drugs (NSAIDs) produced crystalline products, which were characterized by single-crystal X-ray diffraction (SXRD). Simple mixing of the ingredients of the crystalline products (stoichiometry guided by the corresponding crystal structure) produced an aqueous gel (DMSO/water) when the bidentate ligand was water-insoluble L2, whereas water-soluble L1 readily produced hydrogels under similar conditions. Dynamic rheology and scanning electron microscopy (SEM) were employed to characterize the gels. Zone inhibition diameters, minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and hemolysis data suggested that among the hydrogelators, L1MEC derived from L1, meclofenac and Zn(NO3)2, was found to be the best against the Gram-negative bacteria Escherichia coli. The corresponding hydrogel L1MEC_HG and a piece of a dried cloth bandage coated with the hydrogel also showed appreciable activity against E. coli. The antibacterial property of L1MEC_HG against E. coli, thus demonstrated, is relevant in developing an antibacterial SDD system because E. coli is reported to be present in infected wounds.
Collapse
Affiliation(s)
- Sucharita Ghara
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Sourabh Bera
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Parthasarathi Dastidar
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| |
Collapse
|
10
|
Shao Z, Yang J, Gao Y, Zhang Y, Zhao X, Shao Q, Zhang W, Cao C, Liu H, Gan J. Structural and functional studies of PCNA from African swine fever virus. J Virol 2023; 97:e0074823. [PMID: 37534905 PMCID: PMC10506467 DOI: 10.1128/jvi.00748-23] [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: 05/19/2023] [Accepted: 06/16/2023] [Indexed: 08/04/2023] Open
Abstract
Proliferating cell nuclear antigen (PCNA) belongs to the DNA sliding clamp family. Via interacting with various partner proteins, PCNA plays critical roles in DNA replication, DNA repair, chromatin assembly, epigenetic inheritance, chromatin remodeling, and many other fundamental biological processes. Although PCNA and PCNA-interacting partner networks are conserved across species, PCNA of a given species is rarely functional in heterologous systems, emphasizing the importance of more representative PCNA studies. Here, we report two crystal structures of PCNA from African swine fever virus (ASFV), which is the only member of the Asfarviridae family. Compared to the eukaryotic and archaeal PCNAs and the sliding clamp structural homologs from other viruses, AsfvPCNA possesses unique sequences and/or conformations at several regions, such as the J-loop, interdomain-connecting loop (IDCL), P-loop, and C-tail, which are involved in partner recognition or modification of sliding clamps. In addition to double-stranded DNA binding, we also demonstrate that AsfvPCNA can modestly enhance the ligation activity of the AsfvLIG protein. The unique structural features of AsfvPCNA can serve as a potential target for the development of ASFV-specific inhibitors and help combat the deadly virus. IMPORTANCE Two high-resolution crystal structures of African swine fever virus proliferating cell nuclear antigen (AsfvPCNA) are presented here. Structural comparison revealed that AsfvPCNA is unique at several regions, such as the J-loop, the interdomain-connecting loop linker, and the P-loop, which may play important roles in ASFV-specific partner selection of AsfvPCNA. Unlike eukaryotic and archaeal PCNAs, AsfvPCNA possesses high double-stranded DNA-binding affinity. Besides DNA binding, AsfvPCNA can also modestly enhance the ligation activity of the AsfvLIG protein, which is essential for the replication and repair of ASFV genome. The unique structural features make AsfvPCNA a potential target for drug development, which will help combat the deadly virus.
Collapse
Affiliation(s)
- Zhiwei Shao
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jie Yang
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanqing Gao
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yixi Zhang
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xin Zhao
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Qiyuan Shao
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weizhen Zhang
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Chulei Cao
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Hehua Liu
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jianhua Gan
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Dumitrascu F, Caira MR, Avram S, Buiu C, Udrea AM, Vlad IM, Zarafu I, Ioniță P, Nuță DC, Popa M, Chifiriuc MC, Limban C. Repurposing anti-inflammatory drugs for fighting planktonic and biofilm growth. New carbazole derivatives based on the NSAID carprofen: synthesis, in silico and in vitro bioevaluation. Front Cell Infect Microbiol 2023; 13:1181516. [PMID: 37680749 PMCID: PMC10482414 DOI: 10.3389/fcimb.2023.1181516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/14/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction One of the promising leads for the rapid discovery of alternative antimicrobial agents is to repurpose other drugs, such as nonsteroidal anti-inflammatory agents (NSAIDs) for fighting bacterial infections and antimicrobial resistance. Methods A series of new carbazole derivatives based on the readily available anti-inflammatory drug carprofen has been obtained by nitration, halogenation and N-alkylation of carprofen and its esters. The structures of these carbazole compounds were assigned by NMR and IR spectroscopy. Regioselective electrophilic substitution by nitration and halogenation at the carbazole ring was assigned from H NMR spectra. The single crystal X-ray structures of two representative derivatives obtained by dibromination of carprofen, were also determined. The total antioxidant capacity (TAC) was measured using the DPPH method. The antimicrobial activity assay was performed using quantitative methods, allowing establishment of the minimal inhibitory/bactericidal/biofilm eradication concentrations (MIC/MBC/MBEC) on Gram-positive (Staphylococcus aureus, Enterococcus faecalis) and Gram-negative (Escherichia coli, Pseudomonas aeruginosa) strains. Computational assays have been performed to assess the drug- and lead-likeness, pharmacokinetics (ADME-Tox) and pharmacogenomics profiles. Results and discussion The crystal X-ray structures of 3,8-dibromocarprofen and its methyl ester have revealed significant differences in their supramolecular assemblies. The most active antioxidant compound was 1i, bearing one chlorine and two bromine atoms, as well as the CO2Me group. Among the tested derivatives, 1h bearing one chlorine and two bromine atoms has exhibited the widest antibacterial spectrum and the most intensive inhibitory activity, especially against the Gram-positive strains, in planktonic and biofilm growth state. The compounds 1a (bearing one chlorine, one NO2 and one CO2Me group) and 1i (bearing one chlorine, two bromine atoms and a CO2Me group) exhibited the best antibiofilm activity in the case of the P. aeruginosa strain. Moreover, these compounds comply with the drug-likeness rules, have good oral bioavailability and are not carcinogenic or mutagenic. The results demonstrate that these new carbazole derivatives have a molecular profile which deserves to be explored further for the development of novel antibacterial and antibiofilm agents.
Collapse
Affiliation(s)
- Florea Dumitrascu
- ”C. D. Nenitzescu” Institute of Organic and Supramolecular Chemistry, Center for Organic Chemistry, Bucharest, Romania
| | - Mino R. Caira
- Department of Chemistry, University of Cape Town, Cape Town, South Africa
| | - Speranta Avram
- Department of Anatomy, Animal Physiology, and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Catalin Buiu
- Department of Automatic Control and Systems Engineering, Politehnica University of Bucharest, Bucharest, Romania
| | - Ana Maria Udrea
- Laser Department, National Institute for Laser, Plasma and Radiation Physics, Magurele, Romania
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, Bucharest, Romania
| | - Ilinca Margareta Vlad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Irina Zarafu
- Department of Organic Chemistry, Biochemistry and Catalysis, Faculty of Chemistry, University of Bucharest, Bucharest, Romania
| | - Petre Ioniță
- Department of Organic Chemistry, Biochemistry and Catalysis, Faculty of Chemistry, University of Bucharest, Bucharest, Romania
| | - Diana Camelia Nuță
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Marcela Popa
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, Bucharest, Romania
- Department of Botany and Microbiology, University of Bucharest, Bucharest, Romania
- Biological Sciences Section, Romanian Academy, Bucharest, Romania
| | - Carmen Limban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
12
|
Gil D, Hugard S, Borodinov N, Ovchinnikova OS, Muratoglu OK, Bedair H, Oral E. Dual-analgesic loaded UHMWPE exhibits synergistic antibacterial effects against Staphylococci. J Biomed Mater Res B Appl Biomater 2023; 111:912-922. [PMID: 36462210 DOI: 10.1002/jbm.b.35201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/15/2022] [Accepted: 11/06/2022] [Indexed: 12/07/2022]
Abstract
Total joint arthroplasty is one of the most common surgeries in the United States, with almost a million procedures performed annually. Periprosthetic joint infections (PJI) remain the most devastating complications associated with total joint replacement. Effective antibacterial prophylaxis after primary arthroplasty could substantially reduce incidence rate of PJI. In the present study we propose to provide post-arthroplasty prophylaxis via dual-analgesic loaded ultra-high molecular weight polyethylene (UHMWPE). Our approach is based on previous studies that showed pronounced antibacterial activity of analgesic- and NSAID-loaded UHMWPE against Staphylococci. Here, we prepared bupivacaine/tolfenamic acid-loaded UHMWPE and assessed its antibacterial activity against Staphylococcus aureus and Staphylococcus epidermidis. Dual-drug loaded UHMWPE yielded an additional 1-2 log reduction of bacteria, when compared with single-drug loaded UHMWPE. Analysis of the drug elution kinetics suggested that the observed increase in antibacterial activity is due to the increased tolfenamic acid elution from dual-drug loaded UHMWPE. We showed that the increased fractal dimension of the drug domains in UHMWPE could be associated with increased drug elution, leading to higher antibacterial activity. Dual-analgesic loaded UHMWPE proposed here can be used as part of multi-modal antibacterial prophylaxis and promises substantial reduction in post-arthroplasty mortality and morbidity.
Collapse
Affiliation(s)
- Dmitry Gil
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Shannon Hugard
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nikolay Borodinov
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Olga S Ovchinnikova
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Orhun K Muratoglu
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Hany Bedair
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Hamed MM, Sayed M, Abdel-Mohsen SA, Saddik AA, Ibrahim OA, El-Dean AMK, Tolba MS. Synthesis, biological evaluation, and molecular docking studies of novel diclofenac derivatives as antibacterial agents. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
14
|
Donoso F, Cryan JF, Olavarría-Ramírez L, Nolan YM, Clarke G. Inflammation, Lifestyle Factors, and the Microbiome-Gut-Brain Axis: Relevance to Depression and Antidepressant Action. Clin Pharmacol Ther 2023; 113:246-259. [PMID: 35278334 PMCID: PMC10084001 DOI: 10.1002/cpt.2581] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/08/2022] [Indexed: 01/27/2023]
Abstract
Depression is considered a major public health concern, where existing pharmacological treatments are not equally effective across all patients. The pathogenesis of depression involves the interaction of complex biological components, such as the immune system and the microbiota-gut-brain axis. Adjunctive lifestyle-oriented approaches for depression, including physical exercise and special diets are promising therapeutic options when combined with traditional antidepressants. However, the mechanisms of action of these strategies are incompletely understood. Accumulating evidence suggests that physical exercise and specific dietary regimens can modulate both the immune system and gut microbiota composition. Here, we review the current information about the strategies to alleviate depression and their crosstalk with both inflammatory mechanisms and the gut microbiome. We further discuss the role of the microbiota-gut-brain axis as a possible mediator for the adjunctive therapies for depression through inflammatory mechanisms. Finally, we review existing and future adjunctive strategies to manipulate the gut microbiota with potential use for depression, including physical exercise, dietary interventions, prebiotics/probiotics, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Francisco Donoso
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | | | - Yvonne M Nolan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland
| |
Collapse
|
15
|
Shoaib M, Aqib AI, Muzammil I, Majeed N, Bhutta ZA, Kulyar MFEA, Fatima M, Zaheer CNF, Muneer A, Murtaza M, Kashif M, Shafqat F, Pu W. MRSA compendium of epidemiology, transmission, pathophysiology, treatment, and prevention within one health framework. Front Microbiol 2023; 13:1067284. [PMID: 36704547 PMCID: PMC9871788 DOI: 10.3389/fmicb.2022.1067284] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus aureus is recognized as commensal as well as opportunistic pathogen of humans and animals. Methicillin resistant strain of S. aureus (MRSA) has emerged as a major pathogen in hospitals, community and veterinary settings that compromises the public health and livestock production. MRSA basically emerged from MSSA after acquiring SCCmec element through gene transfer containing mecA gene responsible for encoding PBP-2α. This protein renders the MRSA resistant to most of the β-lactam antibiotics. Due to the continuous increasing prevalence and transmission of MRSA in hospitals, community and veterinary settings posing a major threat to public health. Furthermore, high pathogenicity of MRSA due to a number of virulence factors produced by S. aureus along with antibiotic resistance help to breach the immunity of host and responsible for causing severe infections in humans and animals. The clinical manifestations of MRSA consist of skin and soft tissues infection to bacteremia, septicemia, toxic shock, and scalded skin syndrome. Moreover, due to the increasing resistance of MRSA to number of antibiotics, there is need to approach alternatives ways to overcome economic as well as human losses. This review is going to discuss various aspects of MRSA starting from emergence, transmission, epidemiology, pathophysiology, disease patterns in hosts, novel treatment, and control strategies.
Collapse
Affiliation(s)
- Muhammad Shoaib
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of the Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Amjad Islam Aqib
- Department of Medicine, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Iqra Muzammil
- Department of Medicine, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Noreen Majeed
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | | | - Mahreen Fatima
- Faculty of Biosciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | | | - Afshan Muneer
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Maheen Murtaza
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Muhammad Kashif
- Department of Microbiology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Furqan Shafqat
- Department of Microbiology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Wanxia Pu
- Key Laboratory of New Animal Drug Project, Gansu Province/Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs/Lanzhou Institute of Husbandry and Pharmaceutical Sciences of the Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
16
|
Zádori ZS, Király K, Al-Khrasani M, Gyires K. Interactions between NSAIDs, opioids and the gut microbiota - Future perspectives in the management of inflammation and pain. Pharmacol Ther 2023; 241:108327. [PMID: 36473615 DOI: 10.1016/j.pharmthera.2022.108327] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The composition of intestinal microbiota is influenced by a number of factors, including medications, which may have a substantial impact on host physiology. Nonsteroidal anti-inflammatory drugs (NSAIDs) and opioid analgesics are among those widely used medications that have been shown to alter microbiota composition in both animals and humans. Although much effort has been devoted to identify microbiota signatures associated with these medications, much less is known about the underlying mechanisms. Mucosal inflammation, changes in intestinal motility, luminal pH and bile acid metabolism, or direct drug-induced inhibitory effect on bacterial growth are all potential contributors to NSAID- and opioid-induced dysbiosis, however, only a few studies have addressed directly these issues. In addition, there is a notable overlap between the microbiota signatures of these drugs and certain diseases in which they are used, such as spondyloarthritis (SpA), rheumatoid arthritis (RA) and neuropathic pain associated with type 2 diabetes (T2D). The aims of the present review are threefold. First, we aim to provide a comprehensive up-to-date summary on the bacterial alterations caused by NSAIDs and opioids. Second, we critically review the available data on the possible underlying mechanisms of dysbiosis. Third, we review the current knowledge on gut dysbiosis associated with SpA, RA and neuropathic pain in T2D, and highlight the similarities between them and those caused by NSAIDs and opioids. We posit that drug-induced dysbiosis may contribute to the persistence of these diseases, and may potentially limit the therapeutic effect of these medications by long-term use. In this context, we will review the available literature data on the effect of probiotic supplementation and fecal microbiota transplantation on the therapeutic efficacy of NSAIDs and opioids in these diseases.
Collapse
Affiliation(s)
- Zoltán S Zádori
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
| | - Kornél Király
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Klára Gyires
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
17
|
Hishiki A, Okazaki S, Hara K, Hashimoto H. Crystal structure of the sliding DNA clamp from the Gram-positive anaerobic bacterium Clostridioides difficile. J Biochem 2022; 173:13-20. [PMID: 36166824 DOI: 10.1093/jb/mvac079] [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: 07/05/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 01/07/2023] Open
Abstract
The sliding DNA clamp is a ring-shaped protein that encircles DNA within its central channel. It binds to multiple proteins, such as DNA polymerases and DNA repair enzymes, and stimulates their enzymatic activities, thereby playing a crucial role in cell survival and proliferation. Accordingly, the bacterial clamp DnaN is considered to be a promising target for bacterial infection therapy. In this regard, 3D structures of DnaN from pathogenic bacteria are essential for the development of chemical compounds with antimicrobial activity. Here, the crystal structure of DnaN from a Gram-positive bacterium Clostridioides difficile, a human pathogen causing infectious diarrhoea, has been determined at 2.13 Å resolution. A comparison of the structures of DnaN from other bacteria indicates that the structural features of DnaN in terms of overall organization are essentially conserved within Gram-positive and Gram-negative bacteria. However, DnaN from C. difficile has structural differences in the potential binding pocket for partner proteins, implying a non-conventional interaction with its binding partners. Our findings will provide insight into the development of new therapies for C. difficile infection.
Collapse
Affiliation(s)
- Asami Hishiki
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Sumire Okazaki
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Kodai Hara
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| | - Hiroshi Hashimoto
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Shizuoka 422-8002, Japan
| |
Collapse
|
18
|
Zhu B, Xiong W, Tan X, Wu W, Jiang H. Cu2O-Catalyzed Ullmann-type C N cross-coupling reaction of carbazole and aryl chlorides. Tetrahedron Lett 2022. [DOI: 10.1016/j.tetlet.2022.154140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
19
|
Drug repurposing strategy: An emerging approach to identify potential therapeutics for treatment of bovine mastitis. Microb Pathog 2022; 171:105691. [PMID: 35995254 DOI: 10.1016/j.micpath.2022.105691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022]
Abstract
The current study was designed to characterize methicillin-resistant Staphylococcus aureus (MRSA) isolated from bovine milk, along with its response to antibiotics, and ultimately reverse its mechanism of resistance by modulation with non-antibiotics. The synergistic combination of antibiotics with NSAIDs were tested in-vivo by giving MRSA challenge to rabbits. The current study reported an overall 23.79% prevalence of MRSA. The BLAST alignment of current study sequences revealed 99% similarity with mecA gene of MRSA from NCBI database. The current study isolates were more similar to each other and also with reference sequences as compared to other mecA gene sequences from Turkey, India, and Russia. Antibiogram of MRSA isolates showed a highly resistant response to cefoxitin, amoxicillin, and gentamicin. Amoxicillin, gentamicin, tylosin, vancomycin, and ciprofloxacin elicited a significant response (p < 0.05) in combination with non-antibiotics against tested MRSA isolates. The highest zone of inhibition (ZOI) increase was noted for vancomycin in combination with flunixin meglumine (145.45%) and meloxicam (139.36%); gentamicin with flunixin meglumine (85.71%) and ciprofloxacin with ivermectin (71.13%). Synergistic behavior was observed in the combination of gentamicin with ketoprofen; sulfamethoxazole and oxytetracycline with meloxicam. Hematological analysis showed significant differences (p < 0.05) among lymphocyte count and bilirubin. On histopathological examination of skin tissue, hyperplasia of epithelium, sloughed off epidermis, hyperkeratosis, infiltration of inflammatory cells, and hemorrhages were observed. The highest cure rate was observed in case of gentamicin in combination with ketoprofen as compared to other treatment groups. The current study concluded antibiotics in combination with non-antibiotics as potential therapeutic agents for resistance modulation against MRSA. This study will help to devise treatment and control strategies against bovine mastitis. Although the prospect of using NSAIDs to manage infections caused by MRSA appears to be a promising direction, further studies should be conducted to test these medications using suitable in-vivo models in controlled clinical trials to justify their repurposing as a treatment for MRSA infections.
Collapse
|
20
|
Tolba MS, Hamed MM, Sayed M, Kamal El-Dean AM, Abdel-Mohsen SA, Ibrahim OA, Elgaher WA, Hirsch AKH, Saddik AA. Design, Synthesis, Antimicrobial Activity, and Molecular Docking of Some New Diclofenac Derivatives. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2102661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Mahmoud S. Tolba
- Chemistry Department, Faculty of Science, New Valley University, El-Kharja, Egypt
| | - Mahmoud M. Hamed
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Mostafa Sayed
- Chemistry Department, Faculty of Science, New Valley University, El-Kharja, Egypt
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | | | | | - Omneya A. Ibrahim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Walid A.M. Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrucken, Germany
- Department of Pharmacy, Saarland University, Saarbrucken, Germany
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrucken, Germany
- Department of Pharmacy, Saarland University, Saarbrucken, Germany
| | - Abdelreheem Abdelfatah Saddik
- Materials Science and Engineering Laboratory, Department of Chemistry, Faculty of Science, Assiut University, Assiut, Egypt
| |
Collapse
|
21
|
Reddy DS, Sinha A, Kumar A, Saini VK. Drug re-engineering and repurposing: A significant and rapid approach to tuberculosis drug discovery. Arch Pharm (Weinheim) 2022; 355:e2200214. [PMID: 35841594 DOI: 10.1002/ardp.202200214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 01/11/2023]
Abstract
The prevalence of tuberculosis (TB) remains the leading cause of death from a single infectious agent, ranking it above all other contagious diseases. The problem to tackle this disease seems to become even worse due to the outbreak of SARS-CoV-2. Further, the complications related to drug-resistant TB, prolonged treatment regimens, and synergy between TB and HIV are significant drawbacks. There are several drugs to treat TB, but there is still no rapid and accurate treatment available. Intensive research is, therefore, necessary to discover newer molecular analogs that can probably eliminate this disease within a short span. An increase in efficacy can be achieved through re-engineering old TB-drug families and repurposing known drugs. These two approaches have led to the production of newer classes of compounds with novel mechanisms to treat multidrug-resistant strains. With respect to this context, we discuss structural aspects of developing new anti-TB drugs as well as examine advances in TB drug discovery. It was found that the fluoroquinolone, oxazolidinone, and nitroimidazole classes of compounds have greater potential to be further explored for TB drug development. Most of the TB drug candidates in the clinical phase are modified versions of these classes of compounds. Therefore, here we anticipate that modification or repurposing of these classes of compounds has a higher probability to reach the clinical phase of drug development. The information provided will pave the way for researchers to design and identify newer molecular analogs for TB drug development and also broaden the scope of exploring future-generation potent, yet safer anti-TB drugs.
Collapse
Affiliation(s)
- Dinesh S Reddy
- Centre for Nano and Material Sciences, Jain University, Bangalore, India
| | - Anamika Sinha
- Centre for Nano and Material Sciences, Jain University, Bangalore, India
| | - Amit Kumar
- Centre for Nano and Material Sciences, Jain University, Bangalore, India
| | - Vipin K Saini
- Materials and Environmental Chemistry Research Laboratory, School of Environment & Natural Resources, Doon University, Dehradun, India
| |
Collapse
|
22
|
Gümüşgöz Çelik G, Gonca S, Şahin B, Özdemir S, Atilla D, Gürek AG. Novel axially symmetric and unsymmetric silicon(IV) phthalocyanines having anti-inflammatory groups: synthesis, characterization and their biological properties. Dalton Trans 2022; 51:7517-7529. [PMID: 35506506 DOI: 10.1039/d2dt00652a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
New asymmetric Si(IV)Pc (1), monomeloxicammonotriethyleneglycolmonomethylether (phthalocyaninano)silicone, axially ligated with meloxicam as a non-steroidal anti-inflammatory drug (NSAID), or triethylene glycol monomethyl ether and symmetric Si(IV)Pc (2), diclofenac(phthalocyaninano)silicone, axially ligated with two diclofenac as NSAID, were synthesized and characterized as antioxidant and antimicrobial agents together with polyoxo-SiPc (3), ditriethyleneglycolmonomethylether(phthalocyaninano)silicone, and SiPc(OH)2 (4), dihydroxy(phthalocyaninano)silicone. The photophysical and photochemical properties of these compounds were investigated. Then, antioxidant assays, including 2,2-diphenyl-1-picrylhydrazyl (DPPH) and ferrous ion chelating activities, were performed for these Si(IV) phthalocyanine derivatives (1, 2, 3 and 4). The highest DPPH scavenging activity of 73.48% was achieved with compound 2 and the highest ferrous chelating ability of 66.42% was obtained with compound 3. The results of the antioxidant assays indicated that Pc derivatives 1, 2 and 3 have remarkable superoxide radical scavenging activities, and moderate 2,2-diphenyl-1-picrylhydrazyl activities and metal chelating activities. The antimicrobial effects of the Si(IV) phthalocyanine compounds were studied against six pathogenic bacteria and two pathogenic microfungi. The results for the antimicrobial activity of these compounds indicated that Enterococcus faecalis (ATCC 29212) was the most sensitive microorganism and Pseudomonas aeruginosa (ATCC 27853) and Legionella pneumophila subsp. pneumophila (ATCC 33152) were the most resistant microorganisms against the tested compounds. The DNA cleavage ability and microbial cell viability of these compounds were studied. The studied compounds demonstrated excellent DNA nuclease activity and exhibited 100% cell viability inhibition at 500 mg L-1. Also, the antimicrobial photodynamic therapy of the compounds was tested against Escherichia coli (ATCC 25922) and significant photodynamic antimicrobial activity was observed. In addition, the effect of phthalocyanines on biofilm inhibition produced by Staphylococcus aureus (ATCC 25923) was also tested and 3 showed excellent biofilm inhibition of 82.14%.
Collapse
Affiliation(s)
- Gizem Gümüşgöz Çelik
- Department of Chemistry, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey.
| | - Serpil Gonca
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Mersin, Turkey, TR-33343 Yenisehir, Mersin, Turkey
| | - Belgin Şahin
- Department of Chemistry, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey.
| | - Sadin Özdemir
- Food Processing Programme, Technical Science Vocational School, Mersin University, TR-33343 Yenisehir, Mersin, Turkey.
| | - Devrim Atilla
- Department of Chemistry, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey.
| | - Ayşe Gül Gürek
- Department of Chemistry, Gebze Technical University, 41400 Gebze, Kocaeli, Turkey.
| |
Collapse
|
23
|
Immunobiology of tubercle bacilli and prospects of immunomodulatory drugs to tackle tuberculosis (TB) and other non-tubercular mycobacterial infections. Immunobiology 2022; 227:152224. [PMID: 35533535 PMCID: PMC9068598 DOI: 10.1016/j.imbio.2022.152224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/25/2022] [Accepted: 05/03/2022] [Indexed: 01/17/2023]
Abstract
The COVID-19 pandemic has set back progress made on antimicrobial resistance (AMR). Without urgent re-focus, we risk slowing down drug discovery and providing treatment for drug resistant Mycobacterium tuberculosis. Unique in its immune evasion, dormancy and resuscitation, the causal pathogens of tuberculosis (TB) have demonstrated resistance to antibiotics with efflux pumps and the ability to form biofilms. Repurposing drugs is a prospective avenue for finding new anti-TB drugs. There are many advantages to discovering novel targets of an existing drug, as the pharmacokinetic and pharmacodynamic properties have already been established, they are cost-efficient and can be commercially accelerated for the new development. One such group of drugs are non-steroidal anti-inflammatory drugs (NSAIDs) that are originally known for their ability to supress the host proinflammatory responses. In addition to their anti-inflammatory properties, some NSAIDs have been discovered to have antimicrobial modes of action. Of particular interest is Carprofen, identified to inhibit the efflux mechanism and disrupt biofilm formation in mycobacteria. Due to the complexities of host-pathogens interactions in the lung microbiome, inflammatory responses must carefully be controlled alongside the in vivo actions of the prospective anti-infectives. This critical review explores the potential dual role of a selection of NSAIDs, as an anti-inflammatory and anti-tubercular adjunct to reverse the tide of antimicrobial resistance in existing treatments.
Collapse
|
24
|
In Silico and Experimental Investigation of the Biological Potential of Some Recently Developed Carprofen Derivatives. Molecules 2022; 27:molecules27092722. [PMID: 35566083 PMCID: PMC9101252 DOI: 10.3390/molecules27092722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
The efficient regioselective bromination and iodination of the nonsteroidal anti-inflammatory drug (NSAID) carprofen were achieved by using bromine and iodine monochloride in glacial acetic acid. The novel halogenated carprofen derivatives were functionalized at the carboxylic group by esterification. The regioselectivity of the halogenation reaction was evidenced by NMR spectroscopy and confirmed by X-ray analysis. The compounds were screened for their in vitro antibacterial activity against planktonic cells and also for their anti-biofilm effect, using Gram-positive bacteria (Staphylococcus aureus ATCC 29213, Enterococcus faecalis ATCC 29212) and Gram-negative bacteria (Escherichia coli ATCC 25922 and Pseudomonas aeruginosa ATCC 27853). The cytotoxic activity of the novel compounds was tested against HeLa cells. The pharmacokinetic and pharmacodynamic profiles of carprofen derivatives, as well as their toxicity, were established by in silico analyses.
Collapse
|
25
|
Santos ACF, Monteiro LPG, Gomes ACC, Martel F, Santos TM, Ferreira BJML. NSAID-Based Coordination Compounds for Biomedical Applications: Recent Advances and Developments. Int J Mol Sci 2022; 23:2855. [PMID: 35269997 PMCID: PMC8911414 DOI: 10.3390/ijms23052855] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/19/2022] Open
Abstract
After the serendipitous discovery of cisplatin, a platinum-based drug with chemotherapeutic effects, an incredible amount of research in the area of coordination chemistry has been produced. Other transition metal compounds were studied, and several new relevant metallodrugs have been synthetized in the past few years. This review is focused on coordination compounds with first-row transition metals, namely, copper, cobalt, nickel or manganese, or with zinc, which have potential or effective pharmacological properties. It is known that metal complexes, once bound to organic drugs, can enhance the drugs' biological activities, such as anticancer, antimicrobial or anti-inflammatory ones. NSAIDs are a class of compounds with anti-inflammatory properties used to treat pain or fever. NSAIDs' properties can be strongly improved when included in complexes using their compositional N and O donor atoms, which facilitate their coordination to metal ions. This review focuses on the research on this topic and on the promising or effective results that complexes of first-row transition metals and NSAIDs can exhibit.
Collapse
Affiliation(s)
- Ariana C. F. Santos
- Department of Chemistry & CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (A.C.F.S.); (L.P.G.M.); (A.C.C.G.); (T.M.S.)
| | - Luís P. G. Monteiro
- Department of Chemistry & CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (A.C.F.S.); (L.P.G.M.); (A.C.C.G.); (T.M.S.)
| | - Adriana C. C. Gomes
- Department of Chemistry & CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (A.C.F.S.); (L.P.G.M.); (A.C.C.G.); (T.M.S.)
| | - Fátima Martel
- Instituto de Investigação e Inovação em Saúde (i3S), R. Alfredo Allen 208, 4200-135 Porto, Portugal
- Department of Biomedicine–Unit of Biochemistry, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal;
| | - Teresa M. Santos
- Department of Chemistry & CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (A.C.F.S.); (L.P.G.M.); (A.C.C.G.); (T.M.S.)
| | - Bárbara J. M. Leite Ferreira
- Department of Chemistry & CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (A.C.F.S.); (L.P.G.M.); (A.C.C.G.); (T.M.S.)
| |
Collapse
|
26
|
Wang N, Luo J, Deng F, Huang Y, Zhou H. Antibiotic Combination Therapy: A Strategy to Overcome Bacterial Resistance to Aminoglycoside Antibiotics. Front Pharmacol 2022; 13:839808. [PMID: 35281905 PMCID: PMC8905495 DOI: 10.3389/fphar.2022.839808] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
After the first aminoglycoside antibiotic streptomycin being applied in clinical practice in the mid-1940s, aminoglycoside antibiotics (AGAs) are widely used to treat clinical bacterial infections and bacterial resistance to AGAs is increasing. The bacterial resistance to AGAs is owed to aminoglycoside modifying enzyme modification, active efflux pump gene overexpression and 16S rRNA ribosomal subunit methylation, leading to modification of AGAs' structures and decreased concentration of drugs within bacteria. As AGAs's side effects and bacterial resistance, the development of AGAs is time-consuming and difficult. Because bacterial resistance may occur in a short time after application in clinical practice, it was found that the antibacterial effect of the combination was not only better than that of AGAs alone but also reduce the dosage of antibiotics, thereby reducing the occurrence of side effects. This article reviews the clinical use of AGAs, the antibacterial mechanisms, the molecular mechanisms of bacterial resistance, and especially focuses a recent development of the combination of AGAs with other drugs to exert a synergistic antibacterial effect to provide a new strategy to overcome bacterial resistance to AGAs.
Collapse
Affiliation(s)
| | | | | | | | - Hong Zhou
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint Laboratory of International Cooperation, Ministry of Education of Characteristic Ethnic Medicine, School of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
27
|
Monsarrat C, Compain G, André C, Engilberge S, Martiel I, Oliéric V, Wolff P, Brillet K, Landolfo M, Silva da Veiga C, Wagner J, Guichard G, Burnouf DY. Iterative Structure-Based Optimization of Short Peptides Targeting the Bacterial Sliding Clamp. J Med Chem 2021; 64:17063-17078. [PMID: 34806883 DOI: 10.1021/acs.jmedchem.1c00918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The bacterial DNA sliding clamp (SC), or replication processivity factor, is a promising target for the development of novel antibiotics. We report a structure-activity relationship study of a new series of peptides interacting within the Escherichia coli SC (EcSC) binding pocket. Various modifications were explored including N-alkylation of the peptide bonds, extension of the N-terminal moiety, and introduction of hydrophobic and constrained residues at the C-terminus. In each category, single modifications were identified that increased affinity to EcSC. A combination of such modifications yielded in several cases to a substantially increased affinity compared to the parent peptides with Kd in the range of 30-80 nM. X-ray structure analysis of 11 peptide/EcSC co-crystals revealed new interactions at the peptide-protein interface (i.e., stacking interactions, hydrogen bonds, and hydrophobic contacts) that can account for the improved binding. Several compounds among the best binders were also found to be more effective in inhibiting SC-dependent DNA synthesis.
Collapse
Affiliation(s)
- Clément Monsarrat
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Institut Européen de Chimie et Biologie, 2 rue Robert Escarpit, F-33607 Pessac, France
| | - Guillaume Compain
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Institut Européen de Chimie et Biologie, 2 rue Robert Escarpit, F-33607 Pessac, France
| | - Christophe André
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Institut Européen de Chimie et Biologie, 2 rue Robert Escarpit, F-33607 Pessac, France
| | - Sylvain Engilberge
- Swiss Light Source (SLS), Paul Scherrer Institute (PSI), Forschungstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Isabelle Martiel
- Swiss Light Source (SLS), Paul Scherrer Institute (PSI), Forschungstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Vincent Oliéric
- Swiss Light Source (SLS), Paul Scherrer Institute (PSI), Forschungstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Philippe Wolff
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Institut de Biologie Moléculaire et Cellulaire du CNRS, 2 rue Conrad Roentgen, F-67000 Strasbourg, France
| | - Karl Brillet
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Institut de Biologie Moléculaire et Cellulaire du CNRS, 2 rue Conrad Roentgen, F-67000 Strasbourg, France
| | - Marie Landolfo
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Institut de Biologie Moléculaire et Cellulaire du CNRS, 2 rue Conrad Roentgen, F-67000 Strasbourg, France
| | - Cyrielle Silva da Veiga
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Institut de Biologie Moléculaire et Cellulaire du CNRS, 2 rue Conrad Roentgen, F-67000 Strasbourg, France
| | - Jérôme Wagner
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS/Université de Strasbourg, ESBS, 300 Boulevard Sébastien Brant, 67412 Illkirch, France
| | - Gilles Guichard
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Institut Européen de Chimie et Biologie, 2 rue Robert Escarpit, F-33607 Pessac, France
| | - Dominique Y Burnouf
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Institut de Biologie Moléculaire et Cellulaire du CNRS, 2 rue Conrad Roentgen, F-67000 Strasbourg, France
| |
Collapse
|
28
|
Lata K, Vishwakarma J, Kumar S, Khanam T, Ramachandran R. Mycobacterium tuberculosis Endonuclease VIII 2 (Nei2) forms a prereplicative BER complex with DnaN: Identification, characterization, and disruption of complex formation. Mol Microbiol 2021; 117:320-333. [PMID: 34820919 DOI: 10.1111/mmi.14848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022]
Abstract
Mycobacterium tuberculosis Nei2 (Rv3297) is a BER glycosylase that removes oxidized base lesions from ssDNA and replication fork-mimicking substrates. We show that Endonuclease VIII 2 (Nei2) forms a BER complex with the β-clamp (DnaN, Rv0002) with a KD of 170 nM. The Nei2-β-clamp interactions enhance Nei2's activities up to several folds. SEC analysis shows that one molecule of Nei2 binds to a single β-clamp dimer. Nei2 interacts with subsites I and II of the β-clamp via a noncanonical 223 QGCRRCGTLIAY239 Clamp Interacting Protein (CIP) motif in the C-terminal zinc-finger domain, which was previously shown by us to be dispensable for intrinsic Nei2 activity. The 12-mer peptide alone exhibited a KD of 10.28 nM, suggesting that the motif is a key mediator of Nei2-β-clamp interactions. Finally, we identified inhibitors of Nei2-β-clamp interactions using rational methods, in vitro disruption, and SPR assays after querying a database of natural products. We found that Tubulosine, Fumitremorgin C, Toyocamycin, and Aleuritic acid exhibit IC50 values of 94.47, 83.49, 109.7, and 71.49 µM, respectively. They act by disrupting Nei2-β-clamp interactions and do not affect intrinsic Nei2 activity. Among other things, the present study gives insights into the role of Nei2 in bacterial prereplicative BER.
Collapse
Affiliation(s)
- Kiran Lata
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Jyoti Vishwakarma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanjay Kumar
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Taran Khanam
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravishankar Ramachandran
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
29
|
Kim HW, Woo HJ, Yang JY, Kim JB, Kim SH. Hesperetin Inhibits Expression of Virulence Factors and Growth of Helicobacter pylori. Int J Mol Sci 2021; 22:ijms221810035. [PMID: 34576198 PMCID: PMC8472136 DOI: 10.3390/ijms221810035] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a bacterium known to infect the human stomach. It can cause various gastrointestinal diseases including gastritis and gastric cancer. Hesperetin is a major flavanone component contained in citrus fruits. It has been reported to possess antibacterial, antioxidant, and anticancer effects. However, the antibacterial mechanism of hesperetin against H. pylori has not been reported yet. Therefore, the objective of this study was to determine the inhibitory effects of hesperetin on H. pylori growth and its inhibitory mechanisms. The results of this study showed that hesperetin inhibits the growth of H. pylori reference strains and clinical isolates. Hesperetin inhibits the expression of genes in replication (dnaE, dnaN, dnaQ, and holB) and transcription (rpoA, rpoB, rpoD, and rpoN) machineries of H. pylori. Hesperetin also inhibits the expression of genes related to H. pylori motility (flhA, flaA, and flgE) and adhesion (sabA, alpA, alpB, hpaA, and hopZ). It also inhibits the expression of urease. Hespereti n downregulates major virulence factors such as cytotoxin-associated antigen A (CagA) and vacuolating cytotoxin A (VacA) and decreases the translocation of CagA and VacA proteins into gastric adenocarcinoma (AGS) cells. These results might be due to decreased expression of the type IV secretion system (T4SS) and type V secretion system (T5SS) involved in translocation of CagA and VacA, respectively. The results of this study indicate that hesperetin has antibacterial effects against H. pylori. Thus, hesperetin might be an effective natural product for the eradication of H. pylori.
Collapse
Affiliation(s)
- Hyun Woo Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea; (H.W.K.); (J.-B.K.)
| | - Hyun Jun Woo
- Department of Clinical Laboratory Science, Semyung University, Jecheon 27136, Korea;
| | - Ji Yeong Yang
- Division of Crop Foundation, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea;
| | - Jong-Bae Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea; (H.W.K.); (J.-B.K.)
| | - Sa-Hyun Kim
- Department of Clinical Laboratory Science, Semyung University, Jecheon 27136, Korea;
- Correspondence:
| |
Collapse
|
30
|
Sundaresh B, Xu S, Noonan B, Mansour MK, Leong JM, van Opijnen T. Host-informed therapies for the treatment of pneumococcal pneumonia. Trends Mol Med 2021; 27:971-989. [PMID: 34376327 DOI: 10.1016/j.molmed.2021.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022]
Abstract
Over the past two decades, traditional antimicrobial strategies have lost efficacy due to a rapid rise in antibiotic resistance and limited success in developing new antibiotics. Rather than relying on therapeutics solely targeting the bacterial pathogen, therapies are emerging that simultaneously focus on host responses. Here, we describe the most promising 'host-informed therapies' (HITs) in two categories: those that aid patients with fully functional immune systems, and those that aid patients with perturbed immune processes. Using Streptococcus pneumoniae, the leading cause of bacterial pneumonia, as a case study, we show HITs as an attractive option for supplementing infection management. However, to broaden their applicability and design new strategies, targeted research and clinical trials will be essential.
Collapse
Affiliation(s)
| | - Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA; Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Brian Noonan
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Tufts Medical Center, Boston, MA, USA
| | - Michael K Mansour
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA; Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Tufts Medical Center, Boston, MA, USA.
| | - Tim van Opijnen
- Department of Biology, Boston College, Chestnut Hill, MA, USA; Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Tufts Medical Center, Boston, MA, USA.
| |
Collapse
|
31
|
Öztürk İ, Eraç Y, Ballar Kırmızibayrak P, Ermertcan Ş. Nonsteroidal antiinflammatory drugs alter antibiotic susceptibility and expression of virulence-related genes and protein A of Staphylococcus aureus. Turk J Med Sci 2021; 51:835-847. [PMID: 33078603 PMCID: PMC8203164 DOI: 10.3906/sag-2003-60] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 10/20/2020] [Indexed: 11/26/2022] Open
Abstract
Background/aim Nonsteroidal antiinflammatory drugs (NSAIDs) including diclofenac, naproxen, ibuprofen, acetylsalicylic acid, and acetaminophen have been shown to have antimicrobial effects on various microorganisms. The aim of this study was to investigate the antibacterial effects of NSAIDs on
Staphylococcus aureus
. Materials and methods Susceptibilities of
S. aureus
strains to NSAIDs with or without antimicrobials (moxifloxacin, vancomycin, ciprofloxacin, clindamycin, and gentamicin) were determined using the microdilution method and disk diffusion test. Expression levels of genes in the presence of drugs were investigated by real-time quantitative RT-PCR (qRT-PCR), and immunoblotting analysis was performed for staphylococcal protein A (SpA). Results Our results showed that all NSAIDs were active against
S. aureus
strains with MIC values ranging from 195 µg/mL to 6250 µg/mL. NSAIDs increased the antibiotic susceptibility of the strains, and diclofenac was found to be more effective than the other drugs. Drugs showed different effects on expression levels of virulence factor and/or regulatory genes. Immunoblotting analysis of SpA protein was mostly in accordance with qRT-PCR results. Conclusion The regulatory/virulence factor genes and proteins of
S. aureus
investigated in this study may be reasonable targets for these drugs, and we suggest that the data may contribute to the field of infection control and antimicrobial resistance.
Collapse
Affiliation(s)
- İsmail Öztürk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, İzmir Katip Çelebi University, İzmir, Turkey
| | - Yasemin Eraç
- Department of Pharmacology, Faculty of Pharmacy, Ege University, İzmir, Turkey
| | | | - Şafak Ermertcan
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Ege University, İzmir, Turkey
| |
Collapse
|
32
|
Grecka K, Szweda P. Synergistic Effects of Propolis Combined with 2-Phenoxyethanol and Antipyretics on the Growth of Staphylococcus aureus. Pharmaceutics 2021; 13:pharmaceutics13020215. [PMID: 33557393 PMCID: PMC7916011 DOI: 10.3390/pharmaceutics13020215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 11/16/2022] Open
Abstract
The present investigation aimed to assess the combinational effect of commonly used antipyretics and antiseptics with ethanolic extracts of propolis (EEPs) on the growth inhibition of Staphylococcus aureus. The broth microdilution checkerboard assay revealed synergistic interactions between all investigated antipyretics, namely acetylsalicylic acid, ibuprofen, and acetaminophen, with EEPs samples. The values of the fractional inhibitory concentration (ΣFIC) index for all these combinations were <0.5. While, in the case of considered antiseptics, namely chlorhexidine, octenidine dihydrochloride, and 2-phenoxyethanol, the positive interaction was confirmed only for the last one (values of ΣFIC in the range 0.0625-0.25). Combinations of two other agents with all four samples of EEPs resulted in an important antagonistic effect (values of ΣFIC ≥ 4.5). Propolis is mostly dedicated to the treatment of skin/wound infections; thus, these findings are of particular practical importance. The outcomes of the study also support the hypothesis that the propolis's antimicrobial effect is due to the combined (synergistic) action of several ingredients rather than the presence of one component of high antibacterial activity. The composition of 13 ingredients of EEPs (at a concentration below the MIC (minimum inhibitory concentration) of the most active agent) exhibited considerably high anti-staphylococcal efficiency with MIC = 128 µg/mL.
Collapse
Affiliation(s)
| | - Piotr Szweda
- Correspondence: (K.G.); (P.S.); Tel.: +48-58-347-11-44 (P.S.)
| |
Collapse
|
33
|
Non-steroidal anti-inflammatory drugs, plant extracts, and characterized microparticles to modulate antimicrobial resistance of epidemic mecA positive S. aureus of dairy origin. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-020-01628-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
34
|
Mancini F, Unver MY, Elgaher WAM, Jumde VR, Alhayek A, Lukat P, Herrmann J, Witte MD, Köck M, Blankenfeldt W, Müller R, Hirsch AKH. Protein-Templated Hit Identification through an Ugi Four-Component Reaction*. Chemistry 2020; 26:14585-14593. [PMID: 32428268 PMCID: PMC7756422 DOI: 10.1002/chem.202002250] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Indexed: 12/21/2022]
Abstract
Kinetic target-guided synthesis represents an efficient hit-identification strategy, in which the protein assembles its own inhibitors from a pool of complementary building blocks via an irreversible reaction. Herein, we pioneered an in situ Ugi reaction for the identification of novel inhibitors of a model enzyme and binders for an important drug target, namely, the aspartic protease endothiapepsin and the bacterial β-sliding clamp DnaN, respectively. Highly sensitive mass-spectrometry methods enabled monitoring of the protein-templated reaction of four complementary reaction partners, which occurred in a background-free manner for endothiapepsin or with a clear amplification of two binders in the presence of DnaN. The Ugi products we identified show low micromolar activity on endothiapepsin or moderate affinity for the β-sliding clamp. We succeeded in expanding the portfolio of chemical reactions and biological targets and demonstrated the efficiency and sensitivity of this approach, which can find application on any drug target.
Collapse
Affiliation(s)
- Federica Mancini
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - M. Yagiz Unver
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 79747AGGroningenThe Netherlands
| | - Walid A. M. Elgaher
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
| | - Varsha R. Jumde
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
| | - Alaa Alhayek
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Peer Lukat
- Department of Structure and Function of ProteinsHZI38124BraunschweigGermany
| | - Jennifer Herrmann
- Department of Microbial Natural ProductsHIPS–HZI66123SaarbrückenGermany
| | - Martin D. Witte
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 79747AGGroningenThe Netherlands
| | - Matthias Köck
- Department of Microbial Natural ProductsHIPS–HZI66123SaarbrückenGermany
| | - Wulf Blankenfeldt
- Department of Structure and Function of ProteinsHZI38124BraunschweigGermany
- Institute for Biochemistry, Biotechnology and BioinformaticsTechnische Universität BraunschweigSpielmannstr. 738106BraunschweigGermany
| | - Rolf Müller
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
- Department of Microbial Natural ProductsHIPS–HZI66123SaarbrückenGermany
| | - Anna K. H. Hirsch
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 79747AGGroningenThe Netherlands
| |
Collapse
|
35
|
Novel Antibiotics Targeting Bacterial Replicative DNA Polymerases. Antibiotics (Basel) 2020; 9:antibiotics9110776. [PMID: 33158178 PMCID: PMC7694242 DOI: 10.3390/antibiotics9110776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022] Open
Abstract
Multidrug resistance is a worldwide problem that is an increasing threat to global health. Therefore, the development of new antibiotics that inhibit novel targets is of great urgency. Some of the most successful antibiotics inhibit RNA transcription, RNA translation, and DNA replication. Transcription and translation are inhibited by directly targeting the RNA polymerase or ribosome, respectively. DNA replication, in contrast, is inhibited indirectly through targeting of DNA gyrases, and there are currently no antibiotics that inhibit DNA replication by directly targeting the replisome. This contrasts with antiviral therapies where the viral replicases are extensively targeted. In the last two decades there has been a steady increase in the number of compounds that target the bacterial replisome. In particular a variety of inhibitors of the bacterial replicative polymerases PolC and DnaE have been described, with one of the DNA polymerase inhibitors entering clinical trials for the first time. In this review we will discuss past and current work on inhibition of DNA replication, and the potential of bacterial DNA polymerase inhibitors in particular as attractive targets for a new generation of antibiotics.
Collapse
|
36
|
Zhang S, Tang H, Wang Y, Nie B, Yang H, Yuan W, Qu X, Yue B. Antibacterial and antibiofilm effects of flufenamic acid against methicillin-resistant Staphylococcus aureus. Pharmacol Res 2020; 160:105067. [PMID: 32650057 DOI: 10.1016/j.phrs.2020.105067] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are one of the most serious surgery complications, and their prevention is of utmost importance. Flufenamic acid is a non-steroid anti-inflammatory drug approved for clinical use to relieve inflammation and pain in rheumatoid arthritis patients. In this study, we explored the antibacterial efficacy of flufenamic acid and the mechanisms underlying this effect. By using minimal inhibitory concentration (MIC), time-kill, resistance induction assays, and the antibiotic synergy test, we demonstrated that flufenamic acid inhibited the growth of methicillin-resistant staphylococci and did not induce resistance when it was used at the MIC. Furthermore, flufenamic acid acted synergistically with the beta-lactam antibiotic oxacillin and did not show significant toxicity toward mammalian cells. The biofilm inhibition assay revealed that flufenamic acid could prevent biofilm formation on medical implants and destroy the ultrastructure of the bacterial cell wall. RNA sequencing and quantitative RT-PCR indicated that flufenamic acid inhibited the expression of genes associated with peptidoglycan biosynthesis, beta-lactam resistance, quorum sensing, and biofilm formation. Furthermore, flufenamic acid efficiently ameliorated a local infection caused by MRSA in mice. In conclusion, flufenamic acid may be a potent therapeutic compound against MRSA infections and a promising candidate for antimicrobial coating of implants and surgical devices.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin'en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongtao Yang
- Department of Plastic & Reconstructive Surgery, The Ohio State University, Columbus, OH 43210, United States
| | - Weien Yuan
- Ministry of Education Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Bing Yue
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
37
|
Shinde RR, Gaikwad D, Farooqui M. Synthesis and antimicrobial activity of 2‐(4‐(benzo[d]thiazol‐5‐ylsulfonyl)piperazine‐1‐yl)‐N‐substituted acetamide derivatives. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Ravindra R. Shinde
- Post Graduate and Research Centre Maulana Azad College of Arts, Science and Commerce Aurangabad Aurangabad India
| | | | - Mazahar Farooqui
- Post Graduate and Research Centre Maulana Azad College of Arts, Science and Commerce Aurangabad Aurangabad India
| |
Collapse
|
38
|
Leão C, Borges A, Simões M. NSAIDs as a Drug Repurposing Strategy for Biofilm Control. Antibiotics (Basel) 2020; 9:antibiotics9090591. [PMID: 32927675 PMCID: PMC7558876 DOI: 10.3390/antibiotics9090591] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022] Open
Abstract
Persistent infections, usually associated with biofilm-producing bacteria, are challenging for both medical and scientific communities. The potential interest in drug repurposing for biofilm control is growing due to both disinvestment in antibiotic R&D and reduced efficacy of the available panel of antibiotics. In the present study, the antibacterial and antibiofilm activities of four non-steroidal anti-inflammatory drugs (NSAIDs), piroxicam (PXC), diclofenac sodium (DCF), acetylsalicylic acid (ASA) and naproxen sodium (NPX) were evaluated against Escherichia coli and Staphylococcus aureus. The minimum inhibitory/bactericidal concentrations (MICs and MBCs) and the dose–response curves from exposure to the selected NSAIDs were determined. MICs were found for PXC (800 μg/mL) and ASA (1750 μg/mL) against E. coli, and for DCF (2000 μg/mL) and ASA (2000 μg/mL) against S. aureus. No MBCs were found (>2000 μg/mL). The potential of NSAIDs to eradicate preformed biofilms was characterized in terms of biofilm mass, metabolic activity and cell culturability. Additionally, the NSAIDs were tested in combination with kanamycin (KAN) and tetracycline (TET). ASA, DCF and PXC promoted significant reductions in metabolic activity and culturability. However, only PXC promoted biofilm mass removal. Additive interactions were obtained for most of the combinations between NSAIDs and KAN or TET. In general, NSAIDs appear to be a promising strategy to control biofilms as they demonstrated to be more effective than conventional antibiotics.
Collapse
Affiliation(s)
- Cláudia Leão
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (C.L.); (A.B.)
| | - Anabela Borges
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (C.L.); (A.B.)
- DEQ—Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (C.L.); (A.B.)
- DEQ—Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- Correspondence: ; Tel.: +351-225-081-654; Fax: +351-225-081-449
| |
Collapse
|
39
|
Yan F, Gao F. A systematic strategy for the investigation of vaccines and drugs targeting bacteria. Comput Struct Biotechnol J 2020; 18:1525-1538. [PMID: 32637049 PMCID: PMC7327267 DOI: 10.1016/j.csbj.2020.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Infectious and epidemic diseases induced by bacteria have historically caused great distress to people, and have even resulted in a large number of deaths worldwide. At present, many researchers are working on the discovery of viable drug and vaccine targets for bacteria through multiple methods, including the analyses of comparative subtractive genome, core genome, replication-related proteins, transcriptomics and riboswitches, which plays a significant part in the treatment of infectious and pandemic diseases. The 3D structures of the desired target proteins, drugs and epitopes can be predicted and modeled through target analysis. Meanwhile, molecular dynamics (MD) analysis of the constructed drug/epitope-protein complexes is an important standard for testing the suitability of these screened drugs and vaccines. Currently, target discovery, target analysis and MD analysis are integrated into a systematic set of drug and vaccine analysis strategy for bacteria. We hope that this comprehensive strategy will help in the design of high-performance vaccines and drugs.
Collapse
Affiliation(s)
- Fangfang Yan
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Feng Gao
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| |
Collapse
|
40
|
Kuźmycz O, Stączek P. Prospects of NSAIDs administration as double-edged agents against endometrial cancer and pathological species of the uterine microbiome. Cancer Biol Ther 2020; 21:486-494. [PMID: 32174282 PMCID: PMC7515452 DOI: 10.1080/15384047.2020.1736483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Many types of cancers, including endometrial cancer, were found to have cyclooxygenase-2 (COX-2) overexpression. Because this enzyme belongs to the group of pro-inflammatory enzymes, so-called NSAIDs (non-steroidal anti–inflammatory drugs) directly inhibit its activity. An increasing number of reports on COX-2 involvement in cancer, as well as on the role of microbiota in abnormal metabolism and signaling of cells, forces the development of new NSAID types. Besides, NSAIDs can affect some bacteria, which are vaginal/endometrial microbiome members. The overgrowth of those species was found to be a major cause of some uterus diseases. Those infections can lead to chronic inflammatory response and suppress anti-tumorigenic cell pathways. The purpose of this review is to highlight the COX-2 enzyme role in endometrial cancer, the potential effect of the endometrial microbiome on COX-2 enzyme overexpression, and the prospects of NSAIDs use in terms of this type of cancer.
Collapse
Affiliation(s)
- Olga Kuźmycz
- Department of Microbial Genetics, Institute of Microbiology, Biotechnology, and Immunology, Faculty of Biology, University of Łódź, Łódź, Poland
| | - Paweł Stączek
- Department of Microbial Genetics, Institute of Microbiology, Biotechnology, and Immunology, Faculty of Biology, University of Łódź, Łódź, Poland
| |
Collapse
|
41
|
Gil D, Daffinee K, Friedman R, Bhushan B, Muratoglu OK, LaPlante K, Oral E. Synergistic antibacterial effects of analgesics and antibiotics against Staphylococcus aureus. Diagn Microbiol Infect Dis 2019; 96:114967. [PMID: 32057521 DOI: 10.1016/j.diagmicrobio.2019.114967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/28/2022]
Abstract
The local use of analgesics and antibiotics is common during the treatment of periprosthetic joint infection (PJI). The effect of nonantimicrobial drugs on antibacterial activity is underappreciated in clinical practice. This study focuses on the novel assessment of the combined antibacterial effects of commonly used analgesics and antibiotics against methicillin-sensitive Staphylococcus aureus (MSSA)-pathogen associated with most PJIs. We identified that bupivacaine/lidocaine and ketorolac/gentamicin combinations yielded fractional inhibitory concentration indices below 0.4, indicative of synergistic antibacterial effect. Time-kill curves were used for in-depth characterization of the synergy, and the obtained results demonstrated pronounced synergistic effects of bupivacaine/lidocaine and ketorolac/gentamicin combinations against MSSA.
Collapse
Affiliation(s)
- Dmitry Gil
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA; Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, MA
| | - Kathryn Daffinee
- Rhode Island Infectious Diseases Research Program, Veterans Affairs Medical Center, Providence, RI
| | - Renee Friedman
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA
| | - Bhavya Bhushan
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA
| | - Orhun K Muratoglu
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA; Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, MA
| | - Kerry LaPlante
- Rhode Island Infectious Diseases Research Program, Veterans Affairs Medical Center, Providence, RI; Department of Pharmacy Practice, University of Rhode Island, Kingston, RI; Warren Alpert Medical School, Brown University, Providence, RI
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA; Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, MA.
| |
Collapse
|
42
|
Patel H, Makker J, Vakde T, Shaikh D, Badipatla K, Dunne J, Mantri N, Nayudu SK, Glandt M, Balar B, Chilimuri S. Nonsteroidal Anti-Inflammatory Drugs Impact on the Outcomes of Hospitalized Patients with Clostridium difficile Infection. Clin Exp Gastroenterol 2019; 12:449-456. [PMID: 31849510 PMCID: PMC6911331 DOI: 10.2147/ceg.s223886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Mouse model experiments have demonstrated an increased Clostridium difficile infection (CDI) severity with Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) use. We aim to evaluate the impact of NSAIDs in humans after a diagnosis of CDI on primary outcomes defined as I) all-cause mortality and II) toxic mega-colon attributable to CDI. Patients and methods All hospitalized patients with a diagnosis of CDI were divided into two groups; those with NSAIDs administered up to 10 days after onset of CDI versus no NSAIDs use. The primary outcomes were analyzed between the groups, while controlling for severity of CDI. A logistic regression analysis was performed to identify the predictors of worse outcomes. Results NSAIDs were administered in 14% (n=80) of the 568 hospitalized visits for an average of 2.5 days after the CDI diagnosis. All-cause mortality was high in patients who did not receive NSAIDs as compared to those who did receive NSAIDs (16.6% vs 12.5%, p 0.354). Patients who were prescribed NSAIDs were more likely to have toxic mega-colon as compared to those who were not prescribed NSAIDs (2.5% vs 0.6%, p 0.094). Results were not statistically significant, even after controlling for CDI severity. Logistic regression analysis did not identify NSAIDs administration as a significant factor for all-cause mortality in CDI patients. Conclusion This retrospective study results, contrary to mouse model, did not show association between NSAID use and CDI related mortality and toxic mega-colon. Shorter duration of NSAIDs use, younger people in study group, and timely CDI treatment may have resulted in contrasting results.
Collapse
Affiliation(s)
- Harish Patel
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Gastroenterology, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Jasbir Makker
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Gastroenterology, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Trupti Vakde
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Pulmonary and Critical Care Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Danial Shaikh
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Gastroenterology, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Kanthi Badipatla
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Gastroenterology, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - James Dunne
- Support Service and Operation, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Nikhitha Mantri
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Suresh Kumar Nayudu
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Gastroenterology, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Mariela Glandt
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Bhavna Balar
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Gastroenterology, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| | - Sridhar Chilimuri
- Department of Medicine, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA.,Division of Gastroenterology, Bronx Care Health System, Affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, USA
| |
Collapse
|
43
|
Magnowska Z, Jana B, Brochmann RP, Hesketh A, Lametsch R, De Gobba C, Guardabassi L. Carprofen-induced depletion of proton motive force reverses TetK-mediated doxycycline resistance in methicillin-resistant Staphylococcus pseudintermedius. Sci Rep 2019; 9:17834. [PMID: 31780689 PMCID: PMC6882848 DOI: 10.1038/s41598-019-54091-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/31/2019] [Indexed: 11/09/2022] Open
Abstract
We previously showed that doxycycline (DOX) and carprofen (CPF), a veterinary non-steroidal anti-inflammatory drug, have synergistic antimicrobial activity against methicillin-resistant Staphylococcus pseudintermedius (MRSP) carrying the tetracycline resistance determinant TetK. To elucidate the molecular mechanism of this synergy, we investigated the effects of the two drugs, individually and in combination, using a comprehensive approach including RNA sequencing, two-dimensional differential in-gel electrophoresis, macromolecule biosynthesis assays and fluorescence spectroscopy. Exposure of TetK-positive MRSP to CPF alone resulted in upregulation of pathways that generate ATP and NADH, and promote the proton gradient. We showed that CPF is a proton carrier that dissipates the electrochemical potential of the membrane. In the presence of both CPF and DOX, the energy compensation strategy was attenuated by downregulation of all the processes involved, such as citric acid cycle, oxidative phosphorylation and ATP-providing arginine deiminase pathway. Furthermore, protein biosynthesis inhibition increased from 20% under DOX exposure alone to 75% upon simultaneous exposure to CPF. We conclude that synergistic interaction of the drugs restores DOX susceptibility in MRSP by compromising proton-motive-force-dependent TetK-mediated efflux of the antibiotic. MRSP is unable to counterbalance CPF-mediated PMF depletion by cellular metabolic adaptations, resulting in intracellular accumulation of DOX and inhibition of protein biosynthesis.
Collapse
Affiliation(s)
- Zofia Magnowska
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
| | - Bimal Jana
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Rikke Prejh Brochmann
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Andrew Hesketh
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom.,School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Rene Lametsch
- Department of Food Science, Faculty of Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Cristian De Gobba
- Department of Food Science, Faculty of Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Luca Guardabassi
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark. .,Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, United Kingdom.
| |
Collapse
|
44
|
Biological activity of two novel zinc(II) complexes with NSAID mefenamic acid. CHEMICAL PAPERS 2019. [DOI: 10.1007/s11696-019-01003-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
45
|
Carro L. Recent Progress in the Development of Small-Molecule FtsZ Inhibitors as Chemical Tools for the Development of Novel Antibiotics. Antibiotics (Basel) 2019; 8:E217. [PMID: 31717975 PMCID: PMC6963470 DOI: 10.3390/antibiotics8040217] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/26/2019] [Accepted: 11/05/2019] [Indexed: 01/20/2023] Open
Abstract
Antibiotics are potent pharmacological weapons against bacterial pathogens, nevertheless their efficacy is becoming compromised due to the worldwide emergence and spread of multidrug-resistant bacteria or "superbugs". Antibiotic resistance is rising to such dangerous levels that the treatment of bacterial infections is becoming a clinical challenge. Therefore, urgent action is needed to develop new generations of antibiotics that will help tackle this increasing and serious public health problem. Due to its essential role in bacterial cell division, the tubulin-like protein FtsZ has emerged as a promising target for the development of novel antibiotics with new mechanisms of action. This review highlights the medicinal chemistry efforts towards the identification of small-molecule FtsZ inhibitors with antibacterial activity in the last three years.
Collapse
Affiliation(s)
- Laura Carro
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain;
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| |
Collapse
|
46
|
Watching DNA Replication Inhibitors in Action: Exploiting Time-Lapse Microfluidic Microscopy as a Tool for Target-Drug Interaction Studies in Mycobacterium. Antimicrob Agents Chemother 2019; 63:AAC.00739-19. [PMID: 31383667 PMCID: PMC6761567 DOI: 10.1128/aac.00739-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/26/2019] [Indexed: 01/08/2023] Open
Abstract
Spreading resistance to antibiotics and the emergence of multidrug-resistant strains have become frequent in many bacterial species, including mycobacteria, which are the causative agents of severe diseases and which have profound impacts on global health. Here, we used a system of microfluidics, fluorescence microscopy, and target-tagged fluorescent reporter strains of Mycobacterium smegmatis to perform real-time monitoring of replisome and chromosome dynamics following the addition of replication-altering drugs (novobiocin, nalidixic acid, and griselimycin) at the single-cell level. We found that novobiocin stalled replication forks and caused relaxation of the nucleoid and that nalidixic acid triggered rapid replisome collapse and compaction of the nucleoid, while griselimycin caused replisome instability, with the subsequent overinitiation of chromosome replication and overrelaxation of the nucleoid. In addition to study target-drug interactions, our system also enabled us to observe how the tested antibiotics affected the physiology of mycobacterial cells (i.e., growth, chromosome segregation, etc.).
Collapse
|
47
|
Altaf M, Ijaz M, Ghaffar A, Rehman A, Avais M. Antibiotic susceptibility profile and synergistic effect of non-steroidal anti-inflammatory drugs on antibacterial activity of resistant antibiotics (Oxytetracycline and Gentamicin) against methicillin resistant Staphylococcus aureus (MRSA). Microb Pathog 2019; 137:103755. [PMID: 31542423 DOI: 10.1016/j.micpath.2019.103755] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 01/01/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) may exhibit antibacterial activity and have synergistic effects with antibiotics. One way to re-sensitize MRSA to resistant antibiotics is by combining with approved non-antibiotics. The study was intended to explore susceptibility of MRSA to various antibiotics and non-antibiotics (NSAIDs) by micro dilution broth method. MRSA isolates were confirmed by PCR (mecA gene) and in-vitro antibiotic susceptibility was determined by disc diffusion tests. Combinations of NSAIDs with resistant antibiotics were also evaluated in in-vivo trial in mice. In-vitro antibiotic susceptibility profile presented 100% resistance to Cefoxitin, 37.5% to Gentamicin and Amikacin, 25% to Oxytetracycline, 12.5% to Tylosin, Fusidic acid and Vancomycin, while Ciprofloxacin, Levofloxacin, Moxifloxacin, Trimethoprim + Sulfamethoxazole and Linezolid were found sensitive. Synergistic effect was observed when resistant antibiotics (Oxytetracycline/Gentamicin) were combined with sensitive antibiotics (Ciprofloxacin/Linezolid). Combination of Non-antibiotics (NSAIDs) with resistant antibiotics revealed that Meloxicam showed partial synergism with both Oxytetracycline and Gentamicin, while Flunixin Meglumine presented synergistic effect with Oxytetracycline and partial synergism with Gentamicin. Diclofenac Sodium revealed additive effect with Oxytetracycline while in case of Gentamicin indifferent effect was observed. In-vivo results showed that combinations of Oxytetracycline with Meloxicam/Flunixin Meglumine and Gentamicin with Meloxicam/Flunixin Meglumine were effective. The study concluded that the resistance against milk borne zoonotic MRSA infections can be successfully addressed by combining resistant antibiotics with NSAIDs. Flunixin Meglumine can be used in combination with oxytetracycline against MRSA infection.
Collapse
Affiliation(s)
- Muhammad Altaf
- Department of Clinical Medicine and Surgery, University of Veterinary and Animal Sciences, Lahore, 54600, Pakistan
| | - Muhammad Ijaz
- Department of Clinical Medicine and Surgery, University of Veterinary and Animal Sciences, Lahore, 54600, Pakistan.
| | - Awais Ghaffar
- Department of Clinical Medicine and Surgery, University of Veterinary and Animal Sciences, Lahore, 54600, Pakistan
| | - Abdul Rehman
- Department of Epidemiology and Public Health, University of Veterinary and Animal Sciences, Lahore, 54600, Pakistan
| | - Muhammad Avais
- Department of Clinical Medicine and Surgery, University of Veterinary and Animal Sciences, Lahore, 54600, Pakistan
| |
Collapse
|
48
|
Vijayashree Priyadharsini J. In silico validation of the non-antibiotic drugs acetaminophen and ibuprofen as antibacterial agents against red complex pathogens. J Periodontol 2019; 90:1441-1448. [PMID: 31257588 DOI: 10.1002/jper.18-0673] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Acetaminophen (APAP) and ibuprofen (IB) are drugs commonly used to alleviate pain due to their anti-inflammatory, anti-pyretic, and analgesic effect. The aim of the present study is to unravel the molecular mechanisms underlying the antimicrobial potential of these two drugs against red complex pathogens, namely, Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia, by using in silico tools, since they are potentially associated with inflammatory conditions related to periodontal infections. METHODS The STITCH v5.0 pipeline was primarily used for identifying drug-protein interactions; VirulentPred and VICMPred were used for elucidating the virulence property and functional class of the proteins. The subcellular localization of virulent proteins was assessed using PSORTb v3.0 and the epitopes were identified using BepiPred v1.0 Linear Epitope Prediction tool. RESULTS APAP and IB were found to interact with proteins involved in cellular process, metabolism, and virulence. The virulent proteins targeted by the drugs were located in the cytoplasm, which would further add to the effectiveness of the drugs to serve as antimicrobial agents. Finally, epitope prediction revealed multiple epitopes in the virulent proteins which can be specifically focused on. CONCLUSIONS APAP and IB were found to target vital proteins involved in the cellular process, metabolism, and virulence of red complex pathogens. An in-depth knowledge on the interaction of these drugs and their antibacterial activity would add to the plethora of merits gained by these drugs in clinical settings. Further in vitro studies on a wide range of pathogens are warranted to substantiate the true interactions between the drugs and the protein repertoire of pathogens.
Collapse
Affiliation(s)
- Jayaseelan Vijayashree Priyadharsini
- Biomedical Research Unit and Laboratory Animal Centre-Dental Research Cell [BRULAC-DRC], Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
49
|
Yimer EM, Mohammed OA, Mohammedseid SI. Pharmacological Exploitation of Non-Steroidal Anti-inflammatory Drugs as Potential Sources of Novel Antibacterial Agents. ANTI-INFECTIVE AGENTS 2019; 17:81-92. [DOI: 10.2174/2211352516666181008114542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/29/2018] [Accepted: 09/29/2018] [Indexed: 01/04/2025]
Abstract
The ever-increasing threat of bacterial infections and antimicrobial resistance calls for a global effort to deal with this problem. This fast and universal dissemination of antimicrobialresistant bacterial strains resulted in the diminution of therapeutic alternatives for various infectious diseases. Besides, the unaffordability of efficacious antimicrobials coupled with the occurrence of unpleasant adverse effects calls for the exploration of alternative agents with possible antibacterial effect. All these challenges of microbes have posed new drives to the scientific communities. Researchers are now assessing the possible alternative antimicrobial agents for fighting bacterial infections and antimicrobial resistance. Therefore, this review emphasizes on the role of nonsteroidal anti-inflammatory agents as potential sources of novel antibacterial agents on which preliminary studies and randomized controlled trials had been performed. The review also deals with the possible antibacterial mechanism of actions and the likely effects of non-steroidal antiinflammatory drugs when combined along with conventional antibacterial agents.
Collapse
Affiliation(s)
- Ebrahim M. Yimer
- Department of Pharmacology and Toxicology, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Ousman A. Mohammed
- Department of Pharmacology and Toxicology, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Seid I. Mohammedseid
- Department of Pharmacology and Toxicology, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
50
|
Laudy AE. Non-antibiotics, Efflux Pumps and Drug Resistance of Gram-negative Rods. Pol J Microbiol 2019; 67:129-135. [PMID: 30015451 PMCID: PMC7256865 DOI: 10.21307/pjm-2018-017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2018] [Indexed: 01/27/2023] Open
Abstract
Non-antibiotic medicinal products consist of drugs with diverse activity against bacteria. Many non-antibiotics demonstrate direct anti-bacterial activity against Gram-positive cocci. The activity observed against Gram-negative rods is much lower and non-antibiotics primarily from the following groups: non-steroidal anti-inflammatory drugs, cardiovascular and antidepressant medicinal products demonstrate this activity. It has been shown that the low activity of some non-antibiotics or the absence of activity against Gram-negative rods is related, among other things, to the extrusion of these compounds from bacterial cells by multi-drug resistance efflux pumps. Substrates for the resistance-nodulation-division efflux systems include the following non-antibiotics: salicylate, diclofenac, ibuprofen, mefenamic acid, naproxen, amitriptyline, alendronate sodium, nicergoline, and ticlopidine. In addition, interactions between non-antibiotics and multi-drug resistance efflux pumps have been observed. It has also been revealed that depending on the concentration, salicylate induces expression of multi-drug resistance efflux pumps in Escherichia coli, Salmonella enterica subsp. enterica serotype Typhimurium, and Burkholderia cenocepacia. However, salicylate does not affect the expression of the resistance-nodulation-division efflux systems in Stenotrophomonas maltophilia and Acinetobacter baumannii. Most importantly, there were no effects of medicinal products containing some non-antibiotic active substances, except salicylate, as substrates of multi-drug resistance efflux pumps, on the induction of Gram-negative rod resistance to quinolones.
Collapse
Affiliation(s)
- Agnieszka Ewa Laudy
- Department of Pharmaceutical Microbiology, Medical University of Warsaw,Warsaw,Poland
| |
Collapse
|