1
|
Abdelsamie AS, Hamed MM, Schütz C, Röhrig T, Kany AM, Schmelz S, Blankenfeldt W, Hirsch AKH, Hartmann RW, Empting M. Discovery and optimization of thiazole-based quorum sensing inhibitors as potent blockers of Pseudomonas aeruginosa pathogenicity. Eur J Med Chem 2024; 276:116685. [PMID: 39042991 DOI: 10.1016/j.ejmech.2024.116685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024]
Abstract
Pseudomonas aeruginosa causes life-threatening infections especially in hospitalized patients and shows an increasing resistance to established antibiotics. A process known as quorum sensing (QS) enables the pathogen to collectively adapt to various environmental conditions. Disrupting this cell-to-cell communication machinery by small-molecular entities leads to a blockade of bacterial pathogenicity. We aim to devise QS inhibitors acting on the PA-specific PQS QS system via the signal-molecule receptor and transcriptional regulator PqsR (MvfR). In this manuscript, we describe the further optimization of PqsR inverse agonists by broadening the structural space of a previously described triazole-bearing lead compound and arriving at highly potent thiazole derivatives with activities against P. aeruginosa virulence factor pyocyanin in the nanomolar range. All new derivatives were profiled regarding biological activity as well as in vitro ADMET parameters. Additionally, we assessed safety-pharmacology characteristics of the two most promising compounds both bearing a 3-chloro-4-isopropoxyphenyl motive. Demonstrating an overall favorable profile, our new PqsR inverse agonists represent a valuable addition as optimized lead compounds, enabling preclinical development of P. aeruginosa-specific pathoblockers.
Collapse
Affiliation(s)
- Ahmed S Abdelsamie
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany; Department of Chemistry of Natural and Microbial Products, Institute of Pharmaceutical and Drug Industries Research, National Research Centre, El-Buhouth St., Dokki, P.O. Box 12622, Cairo, Egypt
| | - Mostafa M Hamed
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Christian Schütz
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Teresa Röhrig
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Andreas M Kany
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Stefan Schmelz
- Department of Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Wulf Blankenfeldt
- Department of Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Anna K H Hirsch
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Rolf W Hartmann
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Martin Empting
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany.
| |
Collapse
|
2
|
Mudgil U, Khullar L, Chadha J, Prerna, Harjai K. Beyond antibiotics: Emerging antivirulence strategies to combat Pseudomonas aeruginosa in cystic fibrosis. Microb Pathog 2024; 193:106730. [PMID: 38851361 DOI: 10.1016/j.micpath.2024.106730] [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: 03/02/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that poses a significant threat to individuals suffering from cystic fibrosis (CF). The pathogen is highly prevalent in CF individuals and is responsible for chronic infection, resulting in severe tissue damage and poor patient outcome. Prolonged antibiotic administration has led to the emergence of multidrug resistance in P. aeruginosa. In this direction, antivirulence strategies achieving targeted inhibition of bacterial virulence pathways, including quorum sensing, efflux pumps, lectins, and iron chelators, have been explored against CF isolates of P. aeruginosa. Hence, this review article presents a bird's eye view on the pulmonary infections involving P. aeruginosa in CF patients by laying emphasis on factors contributing to bacterial colonization, persistence, and disease progression along with the current line of therapeutics against P. aeruginosa in CF. We further collate scientific literature and discusses various antivirulence strategies that have been tested against P. aeruginosa isolates from CF patients.
Collapse
Affiliation(s)
- Umang Mudgil
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Lavanya Khullar
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Prerna
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
3
|
Pompilio A, Kaya E, Lupetti V, Catelli E, Bianchi M, Maisetta G, Esin S, Di Bonaventura G, Batoni G. Cell-free supernatants from Lactobacillus strains exert antibacterial, antibiofilm, and antivirulence activity against Pseudomonas aeruginosa from cystic fibrosis patients. Microbes Infect 2024; 26:105301. [PMID: 38237656 DOI: 10.1016/j.micinf.2024.105301] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
Chronic lung infections caused by Pseudomonas aeruginosa play a significant role in the mortality and morbidity of cystic fibrosis (CF) patients. The widespread bacterial resistance to conventional antimicrobials demands identifying new strategies to complement or replace current antibiotic therapies. In this study, we evaluated the antibacterial, antibiofilm, and antivirulence properties of cell-free supernatants (CFS) from several Lactobacillus probiotic strains against P. aeruginosa isolated from the sputum of CF patients. A strong and fast antibacterial activity of CFS from different strains of lactobacilli was observed at acidic pH towards P. aeruginosa, both in planktonic and biofilm mode of growth, in conditions mimicking CF lung. Interestingly, although when adjusted at pH 6.0, CFS lost most of their antibacterial potential, they retained some antivirulence activity towards P. aeruginosa, largely dependent on the dose, exposure time, and the Lactobacillus-P. aeruginosa strain combination. In vivo testing in the invertebrate Galleria mellonella model disclosed the lack of toxicity of acidic CFS and their ability to prevent P. aeruginosa infection. For the first time, the results revealed lactobacilli postbiotic activities in the context of the pulmonary environment, pointing to innovative postbiotics' uses in anti-infective therapy.
Collapse
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Esingül Kaya
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Veronica Lupetti
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Elisa Catelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Marta Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy.
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy.
| |
Collapse
|
4
|
Nafee N, Gaber DM, Abouelfetouh A, Alseqely M, Empting M, Schneider M. Enzyme-Linked Lipid Nanocarriers for Coping Pseudomonal Pulmonary Infection. Would Nanocarriers Complement Biofilm Disruption or Pave Its Road? Int J Nanomedicine 2024; 19:3861-3890. [PMID: 38708178 PMCID: PMC11068056 DOI: 10.2147/ijn.s445955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/28/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Cystic fibrosis (CF) is associated with pulmonary Pseudomonas aeruginosa infections persistent to antibiotics. Methods To eradicate pseudomonal biofilms, solid lipid nanoparticles (SLNs) loaded with quorum-sensing-inhibitor (QSI, disrupting bacterial crosstalk), coated with chitosan (CS, improving internalization) and immobilized with alginate lyase (AL, destroying alginate biofilms) were developed. Results SLNs (140-205 nm) showed prolonged release of QSI with no sign of acute toxicity to A549 and Calu-3 cells. The CS coating improved uptake, whereas immobilized-AL ensured >1.5-fold higher uptake and doubled SLN diffusion across the artificial biofilm sputum model. Respirable microparticles comprising SLNs in carbohydrate matrix elicited aerodynamic diameters MMAD (3.54, 2.48 µm) and fine-particle-fraction FPF (65, 48%) for anionic and cationic SLNs, respectively. The antimicrobial and/or antibiofilm activity of SLNs was explored in Pseudomonas aeruginosa reference mucoid/nonmucoid strains as well as clinical isolates. The full growth inhibition of planktonic bacteria was dependent on SLN type, concentration, growth medium, and strain. OD measurements and live/dead staining proved that anionic SLNs efficiently ceased biofilm formation and eradicated established biofilms, whereas cationic SLNs unexpectedly promoted biofilm progression. AL immobilization increased biofilm vulnerability; instead, CS coating increased biofilm formation confirmed by 3D-time lapse confocal imaging. Incubation of SLNs with mature biofilms of P. aeruginosa isolates increased biofilm density by an average of 1.5-fold. CLSM further confirmed the binding and uptake of the labeled SLNs in P. aeruginosa biofilms. Considerable uptake of CS-coated SLNs in non-mucoid strains could be observed presumably due to interaction of chitosan with LPS glycolipids in the outer cell membrane of P. aeruginosa. Conclusion The biofilm-destructive potential of QSI/SLNs/AL inhalation is promising for site-specific biofilm-targeted interventional CF therapy. Nevertheless, the intrinsic/extrinsic fundamentals of nanocarrier-biofilm interactions require further investigation.
Collapse
Affiliation(s)
- Noha Nafee
- Department of Pharmaceutics, College of Pharmacy, Kuwait University, Safat, 13110, Kuwait
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Dina M Gaber
- Department of Pharmaceutics, Division of Pharmaceutical Sciences, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria, 1029, Egypt
| | - Alaa Abouelfetouh
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alamein International University, Alamein, 5060335, Egypt
| | - Mustafa Alseqely
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Martin Empting
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Department of Antiviral and Antivirulence Drugs (AVID), Saarland University, Saarbrücken, 66123, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, 66123, Germany
| |
Collapse
|
5
|
Shi Y, Li J, Wolf CA, Liu S, Sharma SS, Wolber G, Bureik M, Clark BR. Expected and Unexpected Products from the Biochemical Oxidation of Bacterial Alkylquinolones with CYP4F11. JOURNAL OF NATURAL PRODUCTS 2023; 86:2502-2513. [PMID: 37939299 DOI: 10.1021/acs.jnatprod.3c00689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
2-Alkylquinolones are a class of microbial natural products primarily produced in the Pseudomonas and Burkholderia genera that play a key role in modulating quorum sensing. Bacterial alkylquinolones were synthesized and then subjected to oxidative biotransformation using human cytochrome P450 enzyme CYP4F11, heterologously expressed in the fission yeast Schizosaccharomyces pombe. This yielded a range of hydroxylated and carboxylic acid derivatives which had undergone ω-oxidation of the 2-alkyl chain, the structures of which were determined by analysis of NMR and MS data. Oxidation efficiency depended on chain length, with a chain length of eight or nine carbon atoms proving optimal for high yields. Homology modeling suggested that Glu233 was relevant for binding, due to the formation of a hydrogen bond from the quinolone nitrogen to Glu233, and in this position only the longer alkyl chains could come close enough to the heme moiety for effective oxidation. In addition to the direct oxidation products, a number of esters were also isolated, which was attributed to the action of endogenous yeast enzymes on the newly formed ω-hydroxy-alkylquinolones. ω-Oxidation of the alkyl chain significantly reduced the antimicrobial and antibiofilm activity of the quinolones.
Collapse
Affiliation(s)
- Yue Shi
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, People's Republic of China
| | - Jianye Li
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemical Engineering and Materials, Handan University, Handan 056005, Hebei Province, People's Republic of China
| | - Clemens Alexander Wolf
- Molecular Design Lab, Freie Universität Berlin, Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry, Königin-Luise-Straße 2 + 4, 14195 Berlin, Germany
| | - Sijie Liu
- Molecular Design Lab, Freie Universität Berlin, Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry, Königin-Luise-Straße 2 + 4, 14195 Berlin, Germany
| | - Sangeeta S Sharma
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, People's Republic of China
| | - Gerhard Wolber
- Molecular Design Lab, Freie Universität Berlin, Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry, Königin-Luise-Straße 2 + 4, 14195 Berlin, Germany
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, People's Republic of China
| | - Benjamin R Clark
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, People's Republic of China
| |
Collapse
|
6
|
Mollova-Sapundzhieva Y, Angelov P, Georgiev D, Yanev P. Synthetic approach to 2-alkyl-4-quinolones and 2-alkyl-4-quinolone-3-carboxamides based on common β-keto amide precursors. Beilstein J Org Chem 2023; 19:1804-1810. [PMID: 38033452 PMCID: PMC10682542 DOI: 10.3762/bjoc.19.132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023] Open
Abstract
β-Keto amides were used as convenient precursors to both 2-alkyl-4-quinolones and 2-alkyl-4-quinolone-3-carboxamides. The utility of this approach is demonstrated with the synthesis of fourteen novel and four known quinolone derivatives, including natural products of microbial origin such as HHQ and its C5-congener. Two compounds with high activity against S. aureus have been identified among the newly obtained quinolones, with MICs ≤ 3.12 and ≤ 6.25 µg/mL, respectively.
Collapse
Affiliation(s)
- Yordanka Mollova-Sapundzhieva
- Department of Organic Chemistry, University of Plovdiv Paisii Hilendarski, 24 Tsar Asen Str., 4000 Plovdiv, Bulgaria
| | - Plamen Angelov
- Department of Organic Chemistry, University of Plovdiv Paisii Hilendarski, 24 Tsar Asen Str., 4000 Plovdiv, Bulgaria
| | - Danail Georgiev
- Department of Biochemistry and Microbiology, University of Plovdiv Paisii Hilendarski, 24 Tsar Asen Str., 4000 Plovdiv, Bulgaria
| | - Pavel Yanev
- Department of Organic Chemistry, University of Plovdiv Paisii Hilendarski, 24 Tsar Asen Str., 4000 Plovdiv, Bulgaria
| |
Collapse
|
7
|
Hamed MM, Abdelsamie AS, Rox K, Schütz C, Kany AM, Röhrig T, Schmelz S, Blankenfeldt W, Arce‐Rodriguez A, Borrero‐de Acuña JM, Jahn D, Rademacher J, Ringshausen FC, Cramer N, Tümmler B, Hirsch AKH, Hartmann RW, Empting M. Towards Translation of PqsR Inverse Agonists: From In Vitro Efficacy Optimization to In Vivo Proof-of-Principle. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204443. [PMID: 36596691 PMCID: PMC9929129 DOI: 10.1002/advs.202204443] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/28/2022] [Indexed: 06/17/2023]
Abstract
Pseudomonas aeruginosa (PA) is an opportunistic human pathogen, which is involved in a wide range of dangerous infections. It develops alarming resistances toward antibiotic treatment. Therefore, alternative strategies, which suppress pathogenicity or synergize with antibiotic treatments are in great need to combat these infections more effectively. One promising approach is to disarm the bacteria by interfering with their quorum sensing (QS) system, which regulates the release of various virulence factors as well as biofilm formation. Herein, this work reports the rational design, optimization, and in-depth profiling of a new class of Pseudomonas quinolone signaling receptor (PqsR) inverse agonists. The resulting frontrunner compound features a pyrimidine-based scaffold, high in vitro and in vivo efficacy, favorable pharmacokinetics as well as clean safety pharmacology characteristics, which provide the basis for potential pulmonary as well as systemic routes of administration. An X-ray crystal structure in complex with PqsR facilitated further structure-guided lead optimization. The compound demonstrates potent pyocyanin suppression, synergizes with aminoglycoside antibiotic tobramycin against PA biofilms, and is active against a panel of clinical isolates from bronchiectasis patients. Importantly, this in vitro effect translated into in vivo efficacy in a neutropenic thigh infection model in mice providing a proof-of-principle for adjunctive treatment scenarios.
Collapse
Affiliation(s)
- Mostafa M. Hamed
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Ahmed S. Abdelsamie
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of Chemistry of Natural and Microbial ProductsInstitute of Pharmaceutical and Drug Industries ResearchNational Research CentreEl‐Buhouth St.DokkiCairo12622Egypt
| | - Katharina Rox
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of Chemical Biology (CBIO)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
| | - Christian Schütz
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Andreas M. Kany
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Teresa Röhrig
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Stefan Schmelz
- Department of Structure and Function of Proteins (SFPR)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
| | - Wulf Blankenfeldt
- Department of Structure and Function of Proteins (SFPR)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
- Institute for BiochemistryBiotechnology and BioinformaticsTechnische Universität BraunschweigBraunschweigGermany
| | | | - José Manuel Borrero‐de Acuña
- Institute of MicrobiologyTechnische Universität Braunschweig38106BraunschweigGermany
- Braunschweig Integrated Centre of Systems Biology (BRICS)Technische Universität Braunschweig38106BraunschweigGermany
- Departamento de MicrobiologíaFacultad de BiologíaUniversidad de SevillaAv. de la Reina Mercedesno. 6SevillaCP 41012Spain
| | - Dieter Jahn
- Institute of MicrobiologyTechnische Universität Braunschweig38106BraunschweigGermany
- Braunschweig Integrated Centre of Systems Biology (BRICS)Technische Universität Braunschweig38106BraunschweigGermany
| | - Jessica Rademacher
- Department for Respiratory MedicineMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
| | - Felix C. Ringshausen
- Department for Respiratory MedicineMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
- European Reference Network on Rare and Complex Respiratory Diseases (ERN‐ LUNG)FrankfurtGermany
| | - Nina Cramer
- Department for Pediatric PneumologyAllergology and NeonatologyMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Burkhard Tümmler
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
- Department for Pediatric PneumologyAllergology and NeonatologyMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Anna K. H. Hirsch
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| | - Rolf W. Hartmann
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| | - Martin Empting
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| |
Collapse
|
8
|
Novel quinazolinone disulfide analogues as pqs quorum sensing inhibitors against Pseudomonas aeruginosa. Bioorg Chem 2023; 130:106226. [DOI: 10.1016/j.bioorg.2022.106226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/09/2022] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
|
9
|
Dutta L, Ramasastry SSV. Phosphine-Mediated Redox Cyclization of 1-(2-Nitroaryl)prop-2-ynones to 3-Hydroxyquinolin-4-ones: Formal Intramolecular Oxyamination of α,β-Ynones. Org Lett 2022; 24:7665-7670. [PMID: 36226855 DOI: 10.1021/acs.orglett.2c03232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
3-Hydroxyquinoline-4(1H)-ones (3HQs) are privileged structural motifs. The current methods for their synthesis necessitate strongly acidic or basic conditions, which hamper the generality and practicality. Here, we describe phosphine-mediated redox transformation of easily accessible 1-(2-nitroaryl)prop-2-ynones to 3HQs. Besides establishing a new entry to the synthesis of 3HQs under neutral conditions, this method is the first formal intramolecular oxyamination of α,β-ynones. The synthetic utility of this method is demonstrated in the total synthesis of japonine, its analogs, and rare quinoline derivatives.
Collapse
Affiliation(s)
- Lona Dutta
- Organic Synthesis and Catalysis Lab, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Sector 81, Sahibzada Ajit Singh Nagar, Manauli PO, Punjab 140306, India
| | - S S V Ramasastry
- Organic Synthesis and Catalysis Lab, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Sector 81, Sahibzada Ajit Singh Nagar, Manauli PO, Punjab 140306, India
| |
Collapse
|
10
|
Bu F, Liu M, Xie Z, Chen X, Li G, Wang X. Targeted Anti-Biofilm Therapy: Dissecting Targets in the Biofilm Life Cycle. Pharmaceuticals (Basel) 2022; 15:1253. [PMID: 36297365 PMCID: PMC9611117 DOI: 10.3390/ph15101253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 06/13/2024] Open
Abstract
Biofilm is a crucial virulence factor for microorganisms that causes chronic infection. After biofilm formation, the bacteria present improve drug tolerance and multifactorial defense mechanisms, which impose significant challenges for the use of antimicrobials. This indicates the urgent need for new targeted technologies and emerging therapeutic strategies. In this review, we focus on the current biofilm-targeting strategies and those under development, including targeting persistent cells, quorum quenching, and phage therapy. We emphasize biofilm-targeting technologies that are supported by blocking the biofilm life cycle, providing a theoretical basis for design of targeting technology that disrupts the biofilm and promotes practical application of antibacterial materials.
Collapse
Affiliation(s)
| | | | | | | | | | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
11
|
Singh VK, Almpani M, Maura D, Kitao T, Ferrari L, Fontana S, Bergamini G, Calcaterra E, Pignaffo C, Negri M, de Oliveira Pereira T, Skinner F, Gkikas M, Andreotti D, Felici A, Déziel E, Lépine F, Rahme LG. Tackling recalcitrant Pseudomonas aeruginosa infections in critical illness via anti-virulence monotherapy. Nat Commun 2022; 13:5103. [PMID: 36042245 PMCID: PMC9428149 DOI: 10.1038/s41467-022-32833-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal barrier derangement allows intestinal bacteria and their products to translocate to the systemic circulation. Pseudomonas aeruginosa (PA) superimposed infection in critically ill patients increases gut permeability and leads to gut-driven sepsis. PA infections are challenging due to multi-drug resistance (MDR), biofilms, and/or antibiotic tolerance. Inhibition of the quorum-sensing transcriptional regulator MvfR(PqsR) is a desirable anti-PA anti-virulence strategy as MvfR controls multiple acute and chronic virulence functions. Here we show that MvfR promotes intestinal permeability and report potent anti-MvfR compounds, the N-Aryl Malonamides (NAMs), resulting from extensive structure-activity-relationship studies and thorough assessment of the inhibition of MvfR-controlled virulence functions. This class of anti-virulence non-native ligand-based agents has a half-maximal inhibitory concentration in the nanomolar range and strong target engagement. Using a NAM lead in monotherapy protects murine intestinal barrier function, abolishes MvfR-regulated small molecules, ameliorates bacterial dissemination, and lowers inflammatory cytokines. This study demonstrates the importance of MvfR in PA-driven intestinal permeability. It underscores the utility of anti-MvfR agents in maintaining gut mucosal integrity, which should be part of any successful strategy to prevent/treat PA infections and associated gut-derived sepsis in critical illness settings. NAMs provide for the development of crucial preventive/therapeutic monotherapy options against untreatable MDR PA infections. Pseudomonas aeruginosa infections are increasingly difficult to treat due to the development of antimicrobial resistance. Here, the authors describe the synthesis, characterisation and efficacy of a quorum sensing inhibitor.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA, 02114, USA.,Shriners Hospitals for Children, Boston, MA, 02114, USA.,Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Marianna Almpani
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA, 02114, USA.,Shriners Hospitals for Children, Boston, MA, 02114, USA.,Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Damien Maura
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA, 02114, USA.,Shriners Hospitals for Children, Boston, MA, 02114, USA.,Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.,Voyager Therapeutics, Cambridge, MA, 02139, USA
| | - Tomoe Kitao
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA, 02114, USA.,Shriners Hospitals for Children, Boston, MA, 02114, USA.,Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.,T. Kitao, Department of Microbiology, Graduate School of Medicine, Gifu University, Gifu, 501-1194, Japan
| | - Livia Ferrari
- Translational Biology Department, Aptuit (Verona) S.rl, an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy
| | - Stefano Fontana
- DMPK Department, Aptuit (Verona) S.rl, an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy
| | - Gabriella Bergamini
- Translational Biology Department, Aptuit (Verona) S.rl, an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy
| | - Elisa Calcaterra
- Translational Biology Department, Aptuit (Verona) S.rl, an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy
| | - Chiara Pignaffo
- DMPK Department, Aptuit (Verona) S.rl, an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy
| | - Michele Negri
- In vitro Chemotherapy Laboratory, Aptuit (Verona) S.r.l., an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy
| | - Thays de Oliveira Pereira
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, Quebec, H7V 1B7, Canada
| | - Frances Skinner
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Manos Gkikas
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Danielle Andreotti
- Global Synthetic Chemistry Department, Aptuit (Verona) S.r.l., an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy
| | - Antonio Felici
- Department of Microbiology Discovery, In Vitro Biology, Aptuit (Verona) S.r.l., an Evotec Company, 37135 Via A. Fleming 4, Verona, Italy.,A Felici, Academic Partnership, Evotec SE, 37135 Via A. Fleming 4, Verona, Italy
| | - Eric Déziel
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, Quebec, H7V 1B7, Canada
| | - Francois Lépine
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, Quebec, H7V 1B7, Canada
| | - Laurence G Rahme
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA, 02114, USA. .,Shriners Hospitals for Children, Boston, MA, 02114, USA. .,Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
12
|
Huang XH, She MT, Zhang YH, Liu YF, Zhong DX, Zhang YH, Zheng JX, Sun N, Wong WL, Lu YJ. Novel quinoline-based derivatives as the PqsR inhibitor against Pseudomonas aeruginosa PAO1. J Appl Microbiol 2022; 133:2167-2181. [PMID: 35490292 DOI: 10.1111/jam.15601] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/27/2022]
Abstract
AIMS The emerging of drug resistant Pseudomonas aeruginosa is a critical challenge and renders an urgent action to discover innovative antimicrobial interventions. One of these interventions is to disrupt the pseudomonas quinolone signal (pqs) quorum sensing (QS) system, which governs multiple virulence traits and biofilm formation. This study aimed to investigate the QS inhibitory activity of a series of new PqsR inhibitors bearing a quinoline scaffold against Ps. aeruginosa. METHODS AND RESULTS The results showed that compound 1 suppressed the expression of QS-related genes and showed the best inhibitory activity to the pqs system of wild-type Ps. aeruginosa PAO1 with an IC50 of 20.22 μmol l-1 . The virulence factors including pyocyanin, total protease, elastase, and rhamnolipid were significantly suppressed in a concentration-dependent manner with the compound. In addition, 1 in combination with tetracycline inhibited synergistically the bacterial growth and suppressed the biofilm formation of PAO1. The molecular docking studies also suggested that 1 could potentially interact with the ligand-binding domain of the Lys-R type transcriptional regulator PqsR as a competitive antagonist. CONCLUSIONS The quinoline-based derivatives were found to interrupt the quorum sensing system via the pqs pathway and thus the production of virulence factors was inhibited and the antimicrobial susceptibility of Ps. aeruginosa was enhanced. SIGNIFICANCE AND IMPACT OF STUDY The study showed that the quinoline-based derivatives could be used as an anti-virulence agent for treating Ps. aeruginosa infections.
Collapse
Affiliation(s)
- Xuan-He Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China
| | - Meng-Ting She
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China
| | - Yi-Hang Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China
| | - Yi-Fu Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China
| | - Dong-Xiao Zhong
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China
| | - Yi-Han Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China
| | - Jun-Xia Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China
| | - Ning Sun
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, SAR, China.,Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P. R. China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, P. R. China.,Engineering Research Academy of High Value Utilization of Green Plants, Meizhou, P. R. China.,Golden Health (Guangdong) Biotechnology Co., Ltd, Foshan, P. R. China
| |
Collapse
|
13
|
Arif SM, Floto RA, Blundell TL. Using Structure-guided Fragment-Based Drug Discovery to Target Pseudomonas aeruginosa Infections in Cystic Fibrosis. Front Mol Biosci 2022; 9:857000. [PMID: 35433835 PMCID: PMC9006449 DOI: 10.3389/fmolb.2022.857000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) is progressive genetic disease that predisposes lungs and other organs to multiple long-lasting microbial infections. Pseudomonas aeruginosa is the most prevalent and deadly pathogen among these microbes. Lung function of CF patients worsens following chronic infections with P. aeruginosa and is associated with increased mortality and morbidity. Emergence of multidrug-resistant, extensively drug-resistant and pandrug-resistant strains of P. aeruginosa due to intrinsic and adaptive antibiotic resistance mechanisms has failed the current anti-pseudomonal antibiotics. Hence new antibacterials are urgently needed to treat P. aeruginosa infections. Structure-guided fragment-based drug discovery (FBDD) is a powerful approach in the field of drug development that has succeeded in delivering six FDA approved drugs over the past 20 years targeting a variety of biological molecules. However, FBDD has not been widely used in the development of anti-pseudomonal molecules. In this review, we first give a brief overview of our structure-guided FBDD pipeline and then give a detailed account of FBDD campaigns to combat P. aeruginosa infections by developing small molecules having either bactericidal or anti-virulence properties. We conclude with a brief overview of the FBDD efforts in our lab at the University of Cambridge towards targeting P. aeruginosa infections.
Collapse
Affiliation(s)
| | - R. Andres Floto
- Molecular Immunity Unit, Department of Medicine University of Cambridge, MRC-Laboratory of Molecular Biology, Cambridge, United Kingdom
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, United Kingdom
| | - Tom L. Blundell
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Tom L. Blundell,
| |
Collapse
|
14
|
Investigation of Direct and Retro Chromone-2-Carboxamides Based Analogs of Pseudomonas aeruginosa Quorum Sensing Signal as New Anti-Biofilm Agents. Pharmaceuticals (Basel) 2022; 15:ph15040417. [PMID: 35455414 PMCID: PMC9026348 DOI: 10.3390/ph15040417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/19/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022] Open
Abstract
Biofilm formation is considered a major cause of therapeutic failure because bacteria in biofilms have higher protection against antimicrobials. Thus, biofilm-related infections are extremely challenging to treat and pose major concerns for public health, along with huge economic impacts. Pseudomonas aeruginosa, in particular, is a “critical priority” pathogen, responsible for severe infections, especially in cystic fibrosis patients because of its capacity to form resistant biofilms. Therefore, new therapeutic approaches are needed to complete the pipeline of molecules offering new targets and modes of action. Biofilm formation is mainly controlled by Quorum Sensing (QS), a communication system based on signaling molecules. In the present study, we employed a molecular docking approach (Autodock Vina) to assess two series of chromones-based compounds as possible ligands for PqsR, a LuxR-type receptor. Most compounds showed good predicted affinities for PqsR, higher than the PQS native ligand. Encouraged by these docking results, we synthesized a library of 34 direct and 25 retro chromone carboxamides using two optimized routes from 2-chromone carboxylic acid as starting material for both series. We evaluated the synthesized carboxamides for their ability to inhibit the biofilm formation of P. aeruginosa in vitro. Overall, results showed several chromone 2-carboxamides of the retro series are potent inhibitors of the formation of P. aeruginosa biofilms (16/25 compound with % inhibition ≥ 50% at 50 μM), without cytotoxicity on Vero cells (IC50 > 1.0 mM). The 2,4-dinitro-N-(4-oxo-4H-chromen-2-yl) benzamide (6n) was the most promising antibiofilm compound, with potential for hit to lead optimization.
Collapse
|
15
|
Lamin A, Kaksonen AH, Cole IS, Chen XB. Quorum sensing inhibitors applications: a new prospect for mitigation of microbiologically influenced corrosion. Bioelectrochemistry 2022; 145:108050. [DOI: 10.1016/j.bioelechem.2022.108050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/30/2021] [Accepted: 01/02/2022] [Indexed: 12/21/2022]
|
16
|
Duplantier M, Lohou E, Sonnet P. Quorum Sensing Inhibitors to Quench P. aeruginosa Pathogenicity. Pharmaceuticals (Basel) 2021; 14:1262. [PMID: 34959667 PMCID: PMC8707152 DOI: 10.3390/ph14121262] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022] Open
Abstract
The emergence and the dissemination of multidrug-resistant bacteria constitute a major public health issue. Among incriminated Gram-negative bacteria, Pseudomonas aeruginosa has been designated by the WHO as a critical priority threat. During the infection process, this pathogen secretes various virulence factors in order to adhere and colonize host tissues. Furthermore, P. aeruginosa has the capacity to establish biofilms that reinforce its virulence and intrinsic drug resistance. The regulation of biofilm and virulence factor production of this micro-organism is controlled by a specific bacterial communication system named Quorum Sensing (QS). The development of anti-virulence agents targeting QS that could attenuate P. aeruginosa pathogenicity without affecting its growth seems to be a promising new therapeutic strategy. This could prevent the selective pressure put on bacteria by the conventional antibiotics that cause their death and promote resistant strain survival. This review describes the QS-controlled pathogenicity of P. aeruginosa and its different specific QS molecular pathways, as well as the recent advances in the development of innovative QS-quenching anti-virulence agents to fight anti-bioresistance.
Collapse
Affiliation(s)
| | | | - Pascal Sonnet
- AGIR, UR4294, UFR of Pharmacy, Jules Verne University of Picardie, 80037 Amiens, France; (M.D.); (E.L.)
| |
Collapse
|
17
|
Schütz C, Hodzic A, Hamed M, Abdelsamie AS, Kany AM, Bauer M, Röhrig T, Schmelz S, Scrima A, Blankenfeldt W, Empting M. Divergent synthesis and biological evaluation of 2-(trifluoromethyl)pyridines as virulence-attenuating inverse agonists targeting PqsR. Eur J Med Chem 2021; 226:113797. [PMID: 34520957 DOI: 10.1016/j.ejmech.2021.113797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/10/2021] [Accepted: 08/22/2021] [Indexed: 11/19/2022]
Abstract
A short and divergent route towards new derivatives of 2-(trifluoromethyl)pyridines as potent inverse agonists of the bacterial target PqsR against Pseudomonas aeruginosa (PA) infections is described. This Gram-negative pathogen causes severe nosocomial infections and common antibiotic treatment options are rendered ineffective due to resistance issues. Based on an earlier identified optimized hit, we conducted derivatization and rigidification attempts employing two central building blocks. The western part of the molecule is built up via a 2-(trifluoromethyl)pyridine head group equipped with a terminal alkyne. The eastern section is then introduced through aryliode motifs exploiting Sonogashira as well as Suzuki-type chemistry. Subsequent modification provided quick access to an array of compounds, allowed for deep SAR insights, and enabled to optimize the hit scaffold into a lead structure of nanomolar potency combined with favorable in vitro ADME/T features.
Collapse
Affiliation(s)
- Christian Schütz
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Amir Hodzic
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Mostafa Hamed
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Ahmed S Abdelsamie
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany; Chemistry of Natural and Microbial Products Department, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Andreas M Kany
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Maximilian Bauer
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Teresa Röhrig
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Stefan Schmelz
- Department of Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Andrea Scrima
- Department of Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Wulf Blankenfeldt
- Department of Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany; Biotechnology and Bioinformatics, Institute for Biochemistry, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Martin Empting
- Department of Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany.
| |
Collapse
|
18
|
Ramos AF, Woods DF, Shanahan R, Cano R, McGlacken GP, Serra C, O'Gara F, Reen FJ. A structure-function analysis of interspecies antagonism by the 2-heptyl-4-alkyl-quinolone signal molecule from Pseudomonas aeruginosa. MICROBIOLOGY-SGM 2020; 166:169-179. [PMID: 31860435 DOI: 10.1099/mic.0.000876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, the alkyl-quinolone molecular framework has already provided a rich source of bioactivity for the development of novel anti-infective compounds. Based on the quorum-sensing signalling molecules 4-hydroxy-2-heptylquinoline (HHQ) and 3,4-dihydroxy-2-heptylquinoline (PQS) from the nosocomial pathogen Pseudomonas aeruginosa, modifications have been developed with markedly enhanced anti-biofilm bioactivity towards important fungal and bacterial pathogens, including Candida albicans and Aspergillus fumigatus. Here we show that antibacterial activity of HHQ against Vibrionaceae is species-specific and it requires an exquisite level of structural fidelity within the alkyl-quinolone molecular framework. Antibacterial activity was demonstrated against the serious human pathogens Vibrio vulnificus and Vibrio cholerae as well as a panel of bioluminescent squid symbiont Allivibrio fischeri isolates. In contrast, Vibrio parahaemolyticus growth and biofilm formation was unaffected in the presence of HHQ and all the structural variants tested. In general, modification to almost all of the molecule except the alkyl-chain end, led to loss of activity. This suggests that the bacteriostatic activity of HHQ requires the concerted action of the entire framework components. The only exception to this pattern was deuteration of HHQ at the C3 position. HHQ modified with a terminal alkene at the quinolone alkyl chain retained bacteriostatic activity and was also found to activate PqsR signalling comparable to the native agonist. The data from this integrated analysis provides novel insights into the structural flexibility underpinning the signalling activity of the complex alkyl-quinolone molecular communication system.
Collapse
Affiliation(s)
- Ana F Ramos
- CIIMAR, -Centro Interdisciplinar de Investigação Marinha e Ambiental University of Porto, Porto Matosinhos, Portugal.,Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - David F Woods
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland
| | - Rachel Shanahan
- School of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Rafael Cano
- School of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Gerard P McGlacken
- SSPC, Synthesis and Solid State Pharmaceutical Centre, Ireland.,School of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Claudia Serra
- CIIMAR, -Centro Interdisciplinar de Investigação Marinha e Ambiental University of Porto, Porto Matosinhos, Portugal
| | - Fergal O'Gara
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland.,Telethon Kids Institute, Perth Children's Hospital, PerthWA 6009, Australia.,SSPC, Synthesis and Solid State Pharmaceutical Centre, Ireland.,School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, PerthWA, Australia
| | - F Jerry Reen
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
Xie C, Yang D, Wang X, Ma C. A Cascade Reaction of Michael Addition and Truce-Smiles Rearrangement to Synthesize Trisubstituted 4-Quinolone Derivatives. J Org Chem 2020; 85:14937-14944. [PMID: 33146531 DOI: 10.1021/acs.joc.0c01662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A novel transition-metal-free cascade reaction to synthesize 4-quinolone derivatives has been demonstrated. Michael addition and Truce-Smiles rearrangement are included in this protocol, providing a broad scope of 4-quinolones in moderate-to-excellent yields. This work serves as an example of the use of sulfonamides through Truce-Smiles rearrangement to build heterocyclic compounds under mild conditions.
Collapse
Affiliation(s)
- Caixia Xie
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, P. R. China.,School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, P. R. China
| | - Di Yang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, P. R. China
| | - Xinfeng Wang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, P. R. China
| | - Chen Ma
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, P. R. China
| |
Collapse
|
20
|
Ho D, Murgia X, De Rossi C, Christmann R, Hüfner de Mello Martins AG, Koch M, Andreas A, Herrmann J, Müller R, Empting M, Hartmann RW, Desmaele D, Loretz B, Couvreur P, Lehr C. Squalenyl Hydrogen Sulfate Nanoparticles for Simultaneous Delivery of Tobramycin and an Alkylquinolone Quorum Sensing Inhibitor Enable the Eradication of
P. aeruginosa
Biofilm Infections. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202001407] [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]
Affiliation(s)
- Duy‐Khiet Ho
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- Department of Pharmacy Saarland University 66123 Saarbrücken Germany
- Current address: Department of Bioengineering School of Medicine University of Washington Seattle WA 98195 USA
| | - Xabier Murgia
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- Department of Pharmacy Saarland University 66123 Saarbrücken Germany
- Current address: Kusudama Therapeutics Parque Científico y Tecnológico de Gipuzkoa 20014 Donostia-San Sebastián Spain
| | - Chiara De Rossi
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
| | - Rebekka Christmann
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- Department of Pharmacy Saarland University 66123 Saarbrücken Germany
| | | | - Marcus Koch
- INM—Leibniz Institute for New Materials 66123 Saarbrücken Germany
| | - Anastasia Andreas
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Partner Site Hannover-Braunschweig 66123 Saarbrücken Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Partner Site Hannover-Braunschweig 66123 Saarbrücken Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Partner Site Hannover-Braunschweig 66123 Saarbrücken Germany
- Department of Pharmacy Saarland University 66123 Saarbrücken Germany
| | - Martin Empting
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Partner Site Hannover-Braunschweig 66123 Saarbrücken Germany
- Department of Pharmacy Saarland University 66123 Saarbrücken Germany
| | - Rolf W. Hartmann
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- German Centre for Infection Research (DZIF) Partner Site Hannover-Braunschweig 66123 Saarbrücken Germany
- Department of Pharmacy Saarland University 66123 Saarbrücken Germany
| | - Didier Desmaele
- Faculté de Pharmacie Institut Galien Paris Sud Université Paris-Saclay, UMR CNRS 8612 92296 Châtenay-Malabry France
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
| | - Patrick Couvreur
- Faculté de Pharmacie Institut Galien Paris Sud Université Paris-Saclay, UMR CNRS 8612 92296 Châtenay-Malabry France
| | - Claus‐Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland Helmholtz Center for Infection Research 66123 Saarbrücken Germany
- Department of Pharmacy Saarland University 66123 Saarbrücken Germany
| |
Collapse
|
21
|
Ho DK, Murgia X, De Rossi C, Christmann R, Hüfner de Mello Martins AG, Koch M, Andreas A, Herrmann J, Müller R, Empting M, Hartmann RW, Desmaele D, Loretz B, Couvreur P, Lehr CM. Squalenyl Hydrogen Sulfate Nanoparticles for Simultaneous Delivery of Tobramycin and an Alkylquinolone Quorum Sensing Inhibitor Enable the Eradication of P. aeruginosa Biofilm Infections. Angew Chem Int Ed Engl 2020; 59:10292-10296. [PMID: 32243047 PMCID: PMC7317969 DOI: 10.1002/anie.202001407] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/16/2020] [Indexed: 12/02/2022]
Abstract
Elimination of pulmonary Pseudomonas aeruginosa (PA) infections is challenging to accomplish with antibiotic therapies, mainly due to resistance mechanisms. Quorum sensing inhibitors (QSIs) interfering with biofilm formation can thus complement antibiotics. For simultaneous and improved delivery of both active agents to the infection sites, self‐assembling nanoparticles of a newly synthesized squalenyl hydrogen sulfate (SqNPs) were prepared. These nanocarriers allowed for remarkably high loading capacities of hydrophilic antibiotic tobramycin (Tob) and a novel lipophilic QSI at 30 % and circa 10 %, respectively. The drug‐loaded SqNPs showed improved biofilm penetration and enhanced efficacy in relevant biological barriers (mucin/human tracheal mucus, biofilm), leading to complete eradication of PA biofilms at circa 16‐fold lower Tob concentration than Tob alone. This study offers a viable therapy optimization and invigorates the research and development of QSIs for clinical use.
Collapse
Affiliation(s)
- Duy-Khiet Ho
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.,Current address: Department of Bioengineering, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Xabier Murgia
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.,Current address: Kusudama Therapeutics, Parque Científico y Tecnológico de Gipuzkoa, 20014, Donostia-San Sebastián, Spain
| | - Chiara De Rossi
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany
| | - Rebekka Christmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | | | - Marcus Koch
- INM-Leibniz Institute for New Materials, 66123, Saarbrücken, Germany
| | - Anastasia Andreas
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Martin Empting
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Rolf W Hartmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Didier Desmaele
- Faculté de Pharmacie, Institut Galien Paris Sud, Université Paris-Saclay, UMR CNRS 8612, 92296, Châtenay-Malabry, France
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany
| | - Patrick Couvreur
- Faculté de Pharmacie, Institut Galien Paris Sud, Université Paris-Saclay, UMR CNRS 8612, 92296, Châtenay-Malabry, France
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
22
|
Soukarieh F, Liu R, Romero M, Roberston SN, Richardson W, Lucanto S, Oton EV, Qudus NR, Mashabi A, Grossman S, Ali S, Sou T, Kukavica-Ibrulj I, Levesque RC, Bergström CAS, Halliday N, Mistry SN, Emsley J, Heeb S, Williams P, Cámara M, Stocks MJ. Hit Identification of New Potent PqsR Antagonists as Inhibitors of Quorum Sensing in Planktonic and Biofilm Grown Pseudomonas aeruginosa. Front Chem 2020; 8:204. [PMID: 32432073 PMCID: PMC7213079 DOI: 10.3389/fchem.2020.00204] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/04/2020] [Indexed: 12/11/2022] Open
Abstract
Current treatments for Pseudomonas aeruginosa infections are becoming less effective because of the increasing rates of multi-antibiotic resistance. Pharmacological targeting of virulence through inhibition of quorum sensing (QS) dependent virulence gene regulation has considerable therapeutic potential. In P. aeruginosa, the pqs QS system regulates the production of multiple virulence factors as well as biofilm maturation and is a promising approach for developing antimicrobial adjuvants for combatting drug resistance. In this work, we report the hit optimisation for a series of potent novel inhibitors of PqsR, a key regulator of the pqs system, bearing a 2-((5-methyl-5H-[1,2,4]triazino[5,6-b]indol-3-yl)thio) acetamide scaffold. The initial hit compound 7 (PAO1-L IC50 0.98 ± 0.02 μM, PA14 inactive at 10 μM) was obtained through a virtual screening campaign performed on the PqsR ligand binding domain using the University of Nottingham Managed Chemical Compound Collection. Hit optimisation gave compounds with enhanced potency against strains PAO1-L and PA14, evaluated using P. aeruginosa pqs-based QS bioreporter assays. Compound 40 (PAO1-L IC50 0.25 ± 0.12 μM, PA14 IC50 0.34 ± 0.03 μM) is one of the most potent PqsR antagonists reported showing significant inhibition of P. aeruginosa pyocyanin production and pqs system signaling in both planktonic cultures and biofilms. The co-crystal structure of 40 with the PqsR ligand binding domain revealed the specific binding interactions occurring between inhibitor and this key regulatory protein.
Collapse
Affiliation(s)
- Fadi Soukarieh
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom.,The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom
| | - Ruiling Liu
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Manuel Romero
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom.,The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom
| | - Shaun N Roberston
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom.,The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom
| | - William Richardson
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Simone Lucanto
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom.,The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom
| | - Eduard Vico Oton
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Naim Ruhul Qudus
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Alaa Mashabi
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Scott Grossman
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Sadiqur Ali
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Tomás Sou
- Drug Delivery Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden.,Pharmacometrics Group, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Irena Kukavica-Ibrulj
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Quebec City, QC, Canada
| | - Roger C Levesque
- Institut de Biologie Intégrative et des Systèmes, Université Laval, Quebec City, QC, Canada
| | - Christel A S Bergström
- Drug Delivery Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden.,The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Nigel Halliday
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Shailesh N Mistry
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Jonas Emsley
- The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom.,School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Stephan Heeb
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Paul Williams
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom.,The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom
| | - Miguel Cámara
- School of Life Sciences, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom.,The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom
| | - Michael J Stocks
- The National Biofilms Innovation Centre, University of Nottingham, Nottingham, United Kingdom.,School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
23
|
Díaz MA, González SN, Alberto MR, Arena ME. Human probiotic bacteria attenuate Pseudomonas aeruginosa biofilm and virulence by quorum-sensing inhibition. BIOFOULING 2020; 36:597-609. [PMID: 32573275 DOI: 10.1080/08927014.2020.1783253] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 06/11/2023]
Abstract
This work investigated chloroform extracts from culture supernatants of two human probiotic bacteria, Lactobacillus casei CRL 431 and Lactobacillus acidophilus CRL 730 for the production of virulence factors and quorum sensing (QS) interference against three Pseudomonas aeruginosa strains. Both extracts inhibited biofilm biomass (up to 50%), biofilm metabolic activity (up to 39%), the production of the enzyme elastase (up to 63%) and pyocyanin (up to 77%), and decreased QS, without presenting any antibacterial acgivity. In addition, the chloroform extracts of both strains disrupted preformed biofilms of the three strains of P. aeruginosa analyzed (up to 40%). GC-MS analysis revealed that the major compounds detected in the bioactive extracts were four diketopiperazines. This study suggests that the metabolites of L. casei and L. acidophilus could be a promising alternative to combat the pathogenicity of P. aeruginosa.
Collapse
Affiliation(s)
- Myriam Anabel Díaz
- Instituto de Biotecnología Farmacéutica y Alimentaria (INBIOFAL), CONICET-UNT, Tucumán, Argentina
| | - Silvia Nelina González
- Instituto de Biotecnología Farmacéutica y Alimentaria (INBIOFAL), CONICET-UNT, Tucumán, Argentina
- Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán (UNT), Tucumán, Argentina
| | - María Rosa Alberto
- Instituto de Biotecnología Farmacéutica y Alimentaria (INBIOFAL), CONICET-UNT, Tucumán, Argentina
- Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán (UNT), Tucumán, Argentina
| | - Mario Eduardo Arena
- Instituto de Biotecnología Farmacéutica y Alimentaria (INBIOFAL), CONICET-UNT, Tucumán, Argentina
- Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán (UNT), Tucumán, Argentina
| |
Collapse
|
24
|
Ma Y, Wang C, Li Y, Li J, Wan Q, Chen J, Tay FR, Niu L. Considerations and Caveats in Combating ESKAPE Pathogens against Nosocomial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901872. [PMID: 31921562 PMCID: PMC6947519 DOI: 10.1002/advs.201901872] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Indexed: 05/19/2023]
Abstract
ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) are among the most common opportunistic pathogens in nosocomial infections. ESKAPE pathogens distinguish themselves from normal ones by developing a high level of antibiotic resistance that involves multiple mechanisms. Contemporary therapeutic strategies which are potential options in combating ESKAPE bacteria need further investigation. Herein, a broad overview of the antimicrobial research on ESKAPE pathogens over the past five years is provided with prospective clinical applications.
Collapse
Affiliation(s)
- Yu‐Xuan Ma
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Chen‐Yu Wang
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Yuan‐Yuan Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Jing Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Ji‐Hua Chen
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Franklin R. Tay
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| | - Li‐Na Niu
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| |
Collapse
|
25
|
Zender M, Witzgall F, Kiefer A, Kirsch B, Maurer CK, Kany AM, Xu N, Schmelz S, Börger C, Blankenfeldt W, Empting M. Flexible Fragment Growing Boosts Potency of Quorum-Sensing Inhibitors against Pseudomonas aeruginosa Virulence. ChemMedChem 2019; 15:188-194. [PMID: 31709767 PMCID: PMC7004148 DOI: 10.1002/cmdc.201900621] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 12/24/2022]
Abstract
Hit-to-lead optimization is a critical phase in drug discovery. Herein, we report on the fragment-based discovery and optimization of 2-aminopyridine derivatives as a novel lead-like structure for the treatment of the dangerous opportunistic pathogen Pseudomonas aeruginosa. We pursue an innovative treatment strategy by interfering with the Pseudomonas quinolone signal (PQS) quorum sensing (QS) system leading to an abolishment of bacterial pathogenicity. Our compounds act on the PQS receptor (PqsR), a key transcription factor controlling the expression of various pathogenicity determinants. In this target-driven approach, we made use of biophysical screening via surface plasmon resonance (SPR) followed by isothermal titration calorimetry (ITC)-enabled enthalpic efficiency (EE) evaluation. Hit optimization then involved growth vector identification and exploitation. Astonishingly, the latter was successfully achieved by introducing flexible linkers rather than rigid motifs leading to a boost in activity on the target receptor and anti-virulence potency.
Collapse
Affiliation(s)
- Michael Zender
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Florian Witzgall
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Alexander Kiefer
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Benjamin Kirsch
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Christine K Maurer
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Andreas M Kany
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
| | - Ningna Xu
- Lehrstuhl für Biochemie, Universität Bayreuth, Universitätsstr. 30, 95447, Bayreuth, Germany
| | - Stefan Schmelz
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Carsten Börger
- PharmBioTec GmbH, Science Park 1, 66123, Saarbrücken, Germany
| | - Wulf Blankenfeldt
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124, Braunschweig, Germany.,Biotechnology and Bioinformatics, Institute for Biochemistry, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Martin Empting
- Drug Design and Optimization (DDOP), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| |
Collapse
|
26
|
Design, synthesis, and evaluation of compounds capable of reducing Pseudomonas aeruginosa virulence. Eur J Med Chem 2019; 185:111800. [PMID: 31706639 DOI: 10.1016/j.ejmech.2019.111800] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 12/26/2022]
Abstract
Anti-virulence approaches in the treatment of Pseudomonas aeruginosa (PA)-induced infections have shown clinical potential in multiple in vitro and in vivo studies. However, development of these compounds is limited by several factors, including the lack of molecules capable of penetrating the membrane of gram-negative organisms. Here, we report the identification of novel structurally diverse compounds that inhibit PqsR and LasR-based signaling and diminish virulence factor production and biofilm growth in two clinically relevant strains of P. aeruginosa. It is the first report where potential anti-virulent agents were evaluated for inhibition of several virulence factors of PA. Finally, co-treatment with these inhibitors significantly reduced the production of virulence factors induced by the presence of sub-inhibitory levels of ciprofloxacin. Further, we have analyzed the drug-likeness profile of designed compounds using quantitative estimates of drug-likeness (QED) and confirmed their potential as hit molecules for further development.
Collapse
|
27
|
Mellini M, Di Muzio E, D’Angelo F, Baldelli V, Ferrillo S, Visca P, Leoni L, Polticelli F, Rampioni G. In silico Selection and Experimental Validation of FDA-Approved Drugs as Anti-quorum Sensing Agents. Front Microbiol 2019; 10:2355. [PMID: 31649658 PMCID: PMC6796623 DOI: 10.3389/fmicb.2019.02355] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
The emergence of antibiotic resistant bacterial pathogens is increasing at an unprecedented pace, calling for the development of new therapeutic options. Small molecules interfering with virulence processes rather than growth hold promise as an alternative to conventional antibiotics. Anti-virulence agents are expected to decrease bacterial virulence and to pose reduced selective pressure for the emergence of resistance. In the opportunistic pathogen Pseudomonas aeruginosa the expression of key virulence traits is controlled by quorum sensing (QS), an intercellular communication process that coordinates gene expression at the population level. Hence, QS inhibitors represent promising anti-virulence agents against P. aeruginosa. Virtual screenings allow fast and cost-effective selection of target ligands among vast libraries of molecules, thus accelerating the time and limiting the cost of conventional drug-discovery processes, while the drug-repurposing approach is based on the identification of off-target activity of FDA-approved drugs, likely endowed with low cytotoxicity and favorable pharmacological properties. This study aims at combining the advantages of virtual screening and drug-repurposing approaches to identify new QS inhibitors targeting the pqs QS system of P. aeruginosa. An in silico library of 1,467 FDA-approved drugs has been screened by molecular docking, and 5 hits showing the highest predicted binding affinity for the pqs QS receptor PqsR (also known as MvfR) have been selected. In vitro experiments have been performed by engineering ad hoc biosensor strains, which were used to verify the ability of hit compounds to decrease PqsR activity in P. aeruginosa. Phenotypic analyses confirmed the impact of the most promising hit, the antipsychotic drug pimozide, on the expression of P. aeruginosa PqsR-controlled virulence traits. Overall, this study highlights the potential of virtual screening campaigns of FDA-approved drugs to rapidly select new inhibitors of important bacterial functions.
Collapse
Affiliation(s)
- Marta Mellini
- Department of Science, University Roma Tre, Rome, Italy
| | | | | | | | | | - Paolo Visca
- Department of Science, University Roma Tre, Rome, Italy
| | - Livia Leoni
- Department of Science, University Roma Tre, Rome, Italy
| | - Fabio Polticelli
- Department of Science, University Roma Tre, Rome, Italy
- National Institute of Nuclear Physics, Roma Tre Section, Rome, Italy
| | | |
Collapse
|
28
|
Reen FJ, McGlacken GP, O'Gara F. The expanding horizon of alkyl quinolone signalling and communication in polycellular interactomes. FEMS Microbiol Lett 2019; 365:4953739. [PMID: 29718276 DOI: 10.1093/femsle/fny076] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/25/2018] [Indexed: 02/07/2023] Open
Abstract
Population dynamics within natural ecosystems is underpinned by microbial diversity and the heterogeneity of host-microbe and microbe-microbe interactions. Small molecule signals that intersperse between species have been shown to govern many virulence-related processes in established and emerging pathogens. Understanding the capacity of microbes to decode diverse languages and adapt to the presence of 'non-self' cells will provide an important new direction to the understanding of the 'polycellular' interactome. Alkyl quinolones (AQs) have been described in the ESKAPE pathogen Pseudomonas aeruginosa, the primary agent associated with mortality in patients with cystic fibrosis and the third most prevalent nosocomial pathogen worldwide. The role of these molecules in governing the physiology and virulence of P. aeruginosa and other pathogens has received considerable attention, while a role in interspecies and interkingdom communication has recently emerged. Herein we discuss recent advances in our understanding of AQ signalling and communication in the context of microbe-microbe and microbe-host interactions. The integrated knowledge from these systems-based investigations will facilitate the development of new therapeutics based on the AQ framework that serves to disarm the pathogenesis of P. aeruginosa and competing pathogens.
Collapse
Affiliation(s)
- F Jerry Reen
- School of Microbiology, University College Cork, Cork, Ireland
| | - Gerard P McGlacken
- School of Chemistry and Analytical & Biological Chemistry Research Facility (ABCRF), University College Cork, Ireland
| | - Fergal O'Gara
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland
- Human Microbiome Programme, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, USA
| |
Collapse
|
29
|
Quorum Sensing as Antivirulence Target in Cystic Fibrosis Pathogens. Int J Mol Sci 2019; 20:ijms20081838. [PMID: 31013936 PMCID: PMC6515091 DOI: 10.3390/ijms20081838] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder which leads to the secretion of a viscous mucus layer on the respiratory epithelium that facilitates colonization by various bacterial pathogens. The problem of drug resistance has been reported for all the species able to colonize the lung of CF patients, so alternative treatments are urgently needed. In this context, a valid approach is to investigate new natural and synthetic molecules for their ability to counteract alternative pathways, such as virulence regulating quorum sensing (QS). In this review we describe the pathogens most commonly associated with CF lung infections: Staphylococcus aureus, Pseudomonas aeruginosa, species of the Burkholderia cepacia complex and the emerging pathogens Stenotrophomonas maltophilia, Haemophilus influenzae and non-tuberculous Mycobacteria. For each bacterium, the QS system(s) and the molecules targeting the different components of this pathway are described. The amount of investigations published in the last five years clearly indicate the interest and the expectations on antivirulence therapy as an alternative to classical antibiotics.
Collapse
|
30
|
Senerovic L, Opsenica D, Moric I, Aleksic I, Spasić M, Vasiljevic B. Quinolines and Quinolones as Antibacterial, Antifungal, Anti-virulence, Antiviral and Anti-parasitic Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1282:37-69. [PMID: 31515709 DOI: 10.1007/5584_2019_428] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Infective diseases have become health threat of a global proportion due to appearance and spread of microorganisms resistant to majority of therapeutics currently used for their treatment. Therefore, there is a constant need for development of new antimicrobial agents, as well as novel therapeutic strategies. Quinolines and quinolones, isolated from plants, animals, and microorganisms, have demonstrated numerous biological activities such as antimicrobial, insecticidal, anti-inflammatory, antiplatelet, and antitumor. For more than two centuries quinoline/quinolone moiety has been used as a scaffold for drug development and even today it represents an inexhaustible inspiration for design and development of novel semi-synthetic or synthetic agents exhibiting broad spectrum of bioactivities. The structural diversity of synthetized compounds provides high and selective activity attained through different mechanisms of action, as well as low toxicity on human cells. This review describes quinoline and quinolone derivatives with antibacterial, antifungal, anti-virulent, antiviral, and anti-parasitic activities with the focus on the last 10 years literature.
Collapse
Affiliation(s)
- Lidija Senerovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.
| | - Dejan Opsenica
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
- Center of excellence in Environmental Chemistry and Engineering, ICTM - University of Belgrade, Belgrade, Serbia
| | - Ivana Moric
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ivana Aleksic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marta Spasić
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Branka Vasiljevic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
31
|
Schütz C, Empting M. Targeting the Pseudomonas quinolone signal quorum sensing system for the discovery of novel anti-infective pathoblockers. Beilstein J Org Chem 2018; 14:2627-2645. [PMID: 30410625 PMCID: PMC6204780 DOI: 10.3762/bjoc.14.241] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/28/2018] [Indexed: 12/12/2022] Open
Abstract
The Gram-negative opportunistic pathogen Pseudomonas aeruginosa causes severe nosocomial infections. It uses quorum sensing (QS) to regulate and coordinate population-wide group behaviours in the infection process like concerted secretion of virulence factors. One very important signalling network is the Pseudomonas quinolone signal (PQS) QS. With the aim to devise novel and innovative anti-infectives, inhibitors have been designed to address the various potential drug targets present within pqs QS. These range from enzymes within the biosynthesis cascade of the signal molecules PqsABCDE to the receptor of these autoinducers PqsR (MvfR). This review shortly introduces P. aeruginosa and its pathogenicity traits regulated by the pqs system and highlights the published drug discovery efforts providing insights into the compound binding modes if available. Furthermore, suitability of the individual targets for pathoblocker design is discussed.
Collapse
Affiliation(s)
- Christian Schütz
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization (DDOP), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Martin Empting
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization (DDOP), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Saarbrücken, Germany
| |
Collapse
|
32
|
Lakemeyer M, Zhao W, Mandl FA, Hammann P, Sieber SA. Thinking Outside the Box-Novel Antibacterials To Tackle the Resistance Crisis. Angew Chem Int Ed Engl 2018; 57:14440-14475. [PMID: 29939462 DOI: 10.1002/anie.201804971] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Indexed: 12/13/2022]
Abstract
The public view on antibiotics as reliable medicines changed when reports about "resistant superbugs" appeared in the news. While reasons for this resistance development are easily spotted, solutions for re-establishing effective antibiotics are still in their infancy. This Review encompasses several aspects of the antibiotic development pipeline from very early strategies to mature drugs. An interdisciplinary overview is given of methods suitable for mining novel antibiotics and strategies discussed to unravel their modes of action. Select examples of antibiotics recently identified by using these platforms not only illustrate the efficiency of these measures, but also highlight promising clinical candidates with therapeutic potential. Furthermore, the concept of molecules that disarm pathogens by addressing gatekeepers of virulence will be covered. The Review concludes with an evaluation of antibacterials currently in clinical development. Overall, this Review aims to connect select innovative antimicrobial approaches to stimulate interdisciplinary partnerships between chemists from academia and industry.
Collapse
Affiliation(s)
- Markus Lakemeyer
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| | - Weining Zhao
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| | - Franziska A Mandl
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| | - Peter Hammann
- R&D Therapeutic Area Infectious Diseases, Sanofi-Aventis (Deutschland) GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Stephan A Sieber
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| |
Collapse
|
33
|
Calvert MB, Jumde VR, Titz A. Pathoblockers or antivirulence drugs as a new option for the treatment of bacterial infections. Beilstein J Org Chem 2018; 14:2607-2617. [PMID: 30410623 PMCID: PMC6204809 DOI: 10.3762/bjoc.14.239] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022] Open
Abstract
The rapid development of antimicrobial resistance is threatening mankind to such an extent that the World Health Organization expects more deaths from infections than from cancer in 2050 if current trends continue. To avoid this scenario, new classes of anti-infectives must urgently be developed. Antibiotics with new modes of action are needed, but other concepts are also currently being pursued. Targeting bacterial virulence as a means of blocking pathogenicity is a promising new strategy for disarming pathogens. Furthermore, it is believed that this new approach is less susceptible towards resistance development. In this review, recent examples of anti-infective compounds acting on several types of bacterial targets, e.g., adhesins, toxins and bacterial communication, are described.
Collapse
Affiliation(s)
- Matthew B Calvert
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Varsha R Jumde
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
34
|
Lakemeyer M, Zhao W, Mandl FA, Hammann P, Sieber SA. Über bisherige Denkweisen hinaus - neue Wirkstoffe zur Überwindung der Antibiotika-Krise. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201804971] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Markus Lakemeyer
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Weining Zhao
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Franziska A. Mandl
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Peter Hammann
- R&D Therapeutic Area Infectious Diseases; Sanofi-Aventis (Deutschland) GmbH; Industriepark Höchst 65926 Frankfurt am Main Deutschland
| | - Stephan A. Sieber
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| |
Collapse
|
35
|
Shanahan RM, Hickey A, Reen FJ, O'Gara F, McGlacken GP. Synthesis of Benzofuroquinolines via Phosphine-Free Direct Arylation of 4-Phenoxyquinolines in Air. European J Org Chem 2018. [DOI: 10.1002/ejoc.201800923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Rachel M. Shanahan
- Analytical and Biological Research Facility (ABCRF) & School of Chemistry; University College Cork; Cork Ireland
| | - Aobha Hickey
- Analytical and Biological Research Facility (ABCRF) & School of Chemistry; University College Cork; Cork Ireland
| | - F. Jerry Reen
- School of Microbiology; University College Cork; Cork Ireland
| | - Fergal O'Gara
- BIOMERIT Research Centre; School of Microbiology; University College Cork; Cork Ireland
- School of Biomedical Sciences; Curtin Health Innovation Research Institute; Curtin University; 6102 Perth WA Australia
| | - Gerard P. McGlacken
- Analytical and Biological Research Facility (ABCRF) & School of Chemistry; University College Cork; Cork Ireland
| |
Collapse
|
36
|
Kany AM, Sikandar A, Yahiaoui S, Haupenthal J, Walter I, Empting M, Köhnke J, Hartmann RW. Tackling Pseudomonas aeruginosa Virulence by a Hydroxamic Acid-Based LasB Inhibitor. ACS Chem Biol 2018; 13:2449-2455. [PMID: 30088919 DOI: 10.1021/acschembio.8b00257] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In search of novel antibiotics to combat the challenging spread of resistant pathogens, bacterial proteases represent promising targets for pathoblocker development. A common motif for protease inhibitors is the hydroxamic acid function, yet this group has often been related to unspecific inhibition of various metalloproteases. In this work, the inhibition of LasB, a harmful zinc metalloprotease secreted by Pseudomonas aeruginosa, through a hydroxamate derivative is described. The present inhibitor was developed based on a recently reported, highly selective thiol scaffold. Using X-ray crystallography, the lack of inhibition of a range of human matrix metalloproteases could be attributed to a distinct binding mode sparing the S1' pocket. The inhibitor was shown to restore the effect of the antimicrobial peptide LL-37, decrease the formation of P. aeruginosa biofilm and, for the first time for a LasB inhibitor, reduce the release of extracellular DNA. Hence, it is capable of disrupting several important bacterial resistance mechanisms. These results highlight the potential of protease inhibitors to fight bacterial infections and point out the possibility to achieve selective inhibition even with a strong zinc anchor.
Collapse
Affiliation(s)
- Andreas M. Kany
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Asfandyar Sikandar
- Workgroup Structural Biology of Biosynthetic Enzymes, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Samir Yahiaoui
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Jörg Haupenthal
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Isabell Walter
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Martin Empting
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Jesko Köhnke
- Workgroup Structural Biology of Biosynthetic Enzymes, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
37
|
Rezzoagli C, Wilson D, Weigert M, Wyder S, Kümmerli R. Probing the evolutionary robustness of two repurposed drugs targeting iron uptake in Pseudomonas aeruginosa. Evol Med Public Health 2018; 2018:246-259. [PMID: 30455950 PMCID: PMC6234326 DOI: 10.1093/emph/eoy026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
LAY SUMMARY We probed the evolutionary robustness of two antivirulence drugs, gallium and flucytosine, targeting the iron-scavenging pyoverdine in the opportunistic pathogen Pseudomonas aeruginosa. Using an experimental evolution approach in human serum, we showed that antivirulence treatments are not evolutionarily robust per se, but vary in their propensity to select for resistance. BACKGROUND AND OBJECTIVES Treatments that inhibit the expression or functioning of bacterial virulence factors hold great promise to be both effective and exert weaker selection for resistance than conventional antibiotics. However, the evolutionary robustness argument, based on the idea that antivirulence treatments disarm rather than kill pathogens, is controversial. Here, we probe the evolutionary robustness of two repurposed drugs, gallium and flucytosine, targeting the iron-scavenging pyoverdine of the opportunistic human pathogen Pseudomonas aeruginosa. METHODOLOGY We subjected replicated cultures of bacteria to two concentrations of each drug for 20 consecutive days in human serum as an ex vivo infection model. We screened evolved populations and clones for resistance phenotypes, including the restoration of growth and pyoverdine production, and the evolution of iron uptake by-passing mechanisms. We whole-genome sequenced evolved clones to identify the genetic basis of resistance. RESULTS We found that mutants resistant against antivirulence treatments readily arose, but their selective spreading varied between treatments. Flucytosine resistance quickly spread in all populations due to disruptive mutations in upp, a gene encoding an enzyme required for flucytosine activation. Conversely, resistance against gallium arose only sporadically, and was based on mutations in transcriptional regulators, upregulating pyocyanin production, a redox-active molecule promoting siderophore-independent iron acquisition. The spread of gallium resistance was presumably hampered because pyocyanin-mediated iron delivery benefits resistant and susceptible cells alike. CONCLUSIONS AND IMPLICATIONS Our work highlights that antivirulence treatments are not evolutionarily robust per se. Instead, evolutionary robustness is a relative measure, with specific treatments occupying different positions on a continuous scale.
Collapse
Affiliation(s)
- Chiara Rezzoagli
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - David Wilson
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Michael Weigert
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Stefan Wyder
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Rolf Kümmerli
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
38
|
Graef F, Richter R, Fetz V, Murgia X, De Rossi C, Schneider-Daum N, Allegretta G, Elgaher W, Haupenthal J, Empting M, Beckmann F, Brönstrup M, Hartmann R, Gordon S, Lehr CM. In Vitro Model of the Gram-Negative Bacterial Cell Envelope for Investigation of Anti-Infective Permeation Kinetics. ACS Infect Dis 2018; 4:1188-1196. [PMID: 29750862 DOI: 10.1021/acsinfecdis.7b00165] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The cell envelope of Gram-negative bacteria is a formidable biological barrier, inhibiting the action of antibiotics by impeding their permeation into the intracellular environment. In-depth understanding of permeation through this barrier remains a challenge, despite its critical role in antibiotic activity. We therefore designed a divisible in vitro permeation model of the Gram-negative bacterial cell envelope, mimicking its three essential structural elements, the inner membrane and the periplasmic space as well as the outer membrane, on a Transwell setup. The model was characterized by contemporary imaging techniques and employed to generate reproducible quantitative and time-resolved permeation data for various fluorescent probes and anti-infective molecules of different structure and physicochemical properties. For a set of three fluorescent probes, the permeation through the overall membrane model was found to correlate with in bacterio permeation. Even more interestingly, for a set of six Pseudomonas quorum sensing inhibitors, such permeability data were found to be predictive for their corresponding in bacterio activities. Further exploration of the capabilities of the overall model yielded a correlation between the permeability of porin-independent antibiotics and published in bacterio accumulation data; a promising ability to provide structure-permeability information was also demonstrated. Such a model may therefore constitute a valuable tool for the development of novel anti-infective drugs.
Collapse
Affiliation(s)
- Florian Graef
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Robert Richter
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Verena Fetz
- Department of Chemical Biology, HZI, German Center for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany
| | - Xabier Murgia
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Chiara De Rossi
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Nicole Schneider-Daum
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Giuseppe Allegretta
- Department of Drug Design and Optimization, HIPS, HZI, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Walid Elgaher
- Department of Drug Design and Optimization, HIPS, HZI, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Jörg Haupenthal
- Department of Drug Design and Optimization, HIPS, HZI, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Martin Empting
- Department of Pharmacy, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
- Department of Drug Design and Optimization, HIPS, HZI, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Felix Beckmann
- Institute of Materials Research, Helmholtz-Zentrum Geesthacht, Max-Planck-Straße 1, 21502 Geesthacht, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, HZI, German Center for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Rolf Hartmann
- Department of Pharmacy, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
- Department of Drug Design and Optimization, HIPS, HZI, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| | - Sarah Gordon
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, L3 3AF Liverpool, United Kingdom
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8 1, 66123 Saarbrücken, Germany
| |
Collapse
|
39
|
Soukarieh F, Williams P, Stocks MJ, Cámara M. Pseudomonas aeruginosa Quorum Sensing Systems as Drug Discovery Targets: Current Position and Future Perspectives. J Med Chem 2018; 61:10385-10402. [PMID: 29999316 DOI: 10.1021/acs.jmedchem.8b00540] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antimicrobial resistance (AMR) is a serious threat to public health globally, manifested by the frequent emergence of multidrug resistant pathogens that render current chemotherapy inadequate. Health organizations worldwide have recognized the severity of this crisis and implemented action plans to contain its adverse consequences and prolong the utility of conventional antibiotics. Hence, there is a pressing need for new classes of antibacterial agents with novel modes of action. Quorum sensing (QS), a communication system employed by bacterial populations to coordinate virulence gene expression, is a potential target that has been intensively investigated over the past decade. This Perspective will focus on recent advances in targeting the three main quorum sensing systems ( las, rhl, and pqs) of a major opportunistic human pathogen, Pseudomonas aeruginosa, and will specifically evaluate the medicinal chemistry strategies devised to develop QS inhibitors from a drug discovery perspective.
Collapse
Affiliation(s)
- Fadi Soukarieh
- School of Life Sciences, Centre for Biomolecular Sciences , University of Nottingham , Nottingham , NG7 2RD , U.K
| | - Paul Williams
- School of Life Sciences, Centre for Biomolecular Sciences , University of Nottingham , Nottingham , NG7 2RD , U.K
| | - Michael J Stocks
- School of Pharmacy, Centre for Biomolecular Sciences , University of Nottingham , Nottingham , NG7 2RD , U.K
| | - Miguel Cámara
- School of Life Sciences, Centre for Biomolecular Sciences , University of Nottingham , Nottingham , NG7 2RD , U.K
| |
Collapse
|
40
|
Kany AM, Sikandar A, Haupenthal J, Yahiaoui S, Maurer CK, Proschak E, Köhnke J, Hartmann RW. Binding Mode Characterization and Early in Vivo Evaluation of Fragment-Like Thiols as Inhibitors of the Virulence Factor LasB from Pseudomonas aeruginosa. ACS Infect Dis 2018; 4:988-997. [PMID: 29485268 DOI: 10.1021/acsinfecdis.8b00010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The increasing emergence of antibiotic resistance necessitates the development of anti-infectives with novel modes of action. Targeting bacterial virulence is considered a promising approach to develop novel antibiotics with reduced selection pressure. The extracellular collagenase elastase (LasB) plays a pivotal role in the infection process of Pseudomonas aeruginosa and therefore represents an attractive antivirulence target. Mercaptoacetamide-based thiols have been reported to inhibit LasB as well as collagenases from clostridia and bacillus species. The present work provides an insight into the structure-activity relationship (SAR) of these fragment-like LasB inhibitors, demonstrating an inverse activity profile compared to similar inhibitors of clostridial collagenase H (ColH). An X-ray cocrystal structure is presented, revealing distinct binding of two compounds to the active site of LasB, which unexpectedly maintains an open conformation. We further demonstrate in vivo efficacy in a Galleria mellonella infection model and high selectivity of the LasB inhibitors toward human matrix metalloproteinases (MMPs).
Collapse
Affiliation(s)
- Andreas M. Kany
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123, Saarbrücken, Germany
| | - Asfandyar Sikandar
- Workgroup Structural Biology of Biosynthetic Enzymes, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123, Saarbrücken, Germany
| | - Jörg Haupenthal
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123, Saarbrücken, Germany
| | - Samir Yahiaoui
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123, Saarbrücken, Germany
| | - Christine K. Maurer
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123, Saarbrücken, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt, Germany
| | - Jesko Köhnke
- Workgroup Structural Biology of Biosynthetic Enzymes, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123, Saarbrücken, Germany
| | - Rolf W. Hartmann
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123, Saarbrücken, Germany
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| |
Collapse
|
41
|
Kamal AAM, Petrera L, Eberhard J, Hartmann RW. Structure-functionality relationship and pharmacological profiles of Pseudomonas aeruginosa alkylquinolone quorum sensing modulators. Org Biomol Chem 2018; 15:4620-4630. [PMID: 28513746 DOI: 10.1039/c7ob00263g] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An important paradigm in anti-infective research is the antivirulence concept. Pathoblockers are compounds which disarm bacteria of their arsenal of virulence factors. PqsR is a transcriptional regulator controlling the production of such factors in Pseudomonas aeruginosa, most prominently pyocyanin. In this work, a series of tool compounds based on the structure of the natural ligand 2-heptyl-4-quinolone (HHQ) were used for probing the structure-functionality relationship. Four different profiles are identified namely agonists, antagonists, inverse agonists and biphasic modulators. Molecular docking studies revealed that each class of the PqsR modulators showed distinctive interactions in the PqsR binding domain. It was found that the substituents in position 3 of the quinolone core act as a switch between the different profiles, according to their ability to donate or accept a hydrogen bond, or form a hydrophobic interaction. Finally, it was shown that only inverse agonists were able to strongly inhibit pyocyanin production.
Collapse
Affiliation(s)
- Ahmed A M Kamal
- Helmholtz-Institute for Pharmaceutical Research Saarland, Department of Drug Design and Optimization, Campus E8.1, 66123 Saarbrücken, Germany
| | | | | | | |
Collapse
|
42
|
Rajkumari J, Borkotoky S, Murali A, Suchiang K, Mohanty SK, Busi S. Attenuation of quorum sensing controlled virulence factors and biofilm formation in Pseudomonas aeruginosa by pentacyclic triterpenes, betulin and betulinic acid. Microb Pathog 2018. [PMID: 29526565 DOI: 10.1016/j.micpath.2018.03.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The production of virulence determinants and biofilm formation in numerous pathogens is regulated by the cell-density-dependent phenomenon, Quorum sensing (QS). The QS system in multidrug resistant opportunistic pathogen, P. aeruginosa constitutes of three main regulatory circuits namely Las, Rhl, and Pqs which are closely linked to its pathogenicity and establishment of chronic infections. In spite intensive antibiotic therapy, P. aeruginosa continue to be an important cause of nosocomial infections and also the major cause of mortality in Cystic Fibrosis patients with 80% of the adults suffering from chronic P. aeruginosa infection. Hence, targeting QS circuit offers an effective intervention to the ever increasing problem of drug resistant pathogens. In the present study, the pentacyclic triterpenes i.e. Betulin (BT) and Betulinic acid (BA) exhibited significant attenuation in production of QS-regulated virulence factors and biofilm formation in P. aeruginosa, at the sub-lethal concentration. The test compound remarkably interfered in initial stages of biofilm development by decreasing the exopolysaccharide production and cell surface hydrophobicity. Based on the in vivo studies, the test compounds notably enhanced the survival of Caenorhabditis elegans infected with P. aeruginosa. Furthermore, molecular docking analysis revealed that BT and BA can act as a strong competitive inhibitor for QS receptors, LasR and RhlR. The findings suggest that BT and BA can serve as potential anti-infectives in the controlling chronic infection of P. aeruginosa.
Collapse
Affiliation(s)
- Jobina Rajkumari
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Subhomoi Borkotoky
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Ayaluru Murali
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Kitlangki Suchiang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Saswat Kumar Mohanty
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, India.
| |
Collapse
|
43
|
Soukarieh F, Vico Oton E, Dubern JF, Gomes J, Halliday N, de Pilar Crespo M, Ramírez-Prada J, Insuasty B, Abonia R, Quiroga J, Heeb S, Williams P, Stocks MJ, Cámara M. In Silico and in Vitro-Guided Identification of Inhibitors of Alkylquinolone-Dependent Quorum Sensing in Pseudomonas aeruginosa. Molecules 2018; 23:E257. [PMID: 29382099 PMCID: PMC6017655 DOI: 10.3390/molecules23020257] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/20/2018] [Accepted: 01/20/2018] [Indexed: 01/21/2023] Open
Abstract
Pseudomonas aeruginosa is a major opportunistic pathogen in cystic fibrosis, wound and nosocomial infections, posing a serious burden to public health, due to its antibiotic resistance. The P. aeruginosa Pseudomonas Quinolone System (pqs) quorum sensing system, driven by the activation of the transcriptional regulator, PqsR (MvfR) by alkylquinolone (AQ) signal molecules, is a key player in the regulation of virulence and a potential target for the development of novel antibacterial agents. In this study, we performed in silico docking analysis, coupled with screening using a P. aeruginosa mCTX::PpqsA-lux chromosomal promoter fusion, to identify a series of new PqsR antagonists. The hit compounds inhibited pyocyanin and alkylquinolone signal molecule production in P. aeruginosa PAO1-L and PA14 strains. The inhibitor Ia, which showed the highest activity in PA14, reduced biofilm formation in PAO1-L and PA14, increasing their sensitivity to tobramycin. Furthermore, the hepatic and plasma stabilities for these compounds were determined in both rat and human in vitro microsomal assays, to gain a further understanding of their therapeutic potential. This work has uncovered a new class of P. aeruginosa PqsR antagonists with potential for hit to lead optimisation in the search for quorum sensing inhibitors for future anti-infective drug discovery programs.
Collapse
Affiliation(s)
- Fadi Soukarieh
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| | - Eduard Vico Oton
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| | - Jean-Frédéric Dubern
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| | - Janice Gomes
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| | - Nigel Halliday
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| | - Maria de Pilar Crespo
- Department of Microbiology, Universidad del Valle and Departamento of Biomedical Sciences, Universidad Santiago de Cali, Cali AA 760035, Colombia;
| | - Jonathan Ramírez-Prada
- Department of Chemistry, Universidad del Valle, Cali AA 25360, Colombia; (J.R.-P.); (B.I.); (R.A.); (J.Q.)
| | - Braulio Insuasty
- Department of Chemistry, Universidad del Valle, Cali AA 25360, Colombia; (J.R.-P.); (B.I.); (R.A.); (J.Q.)
| | - Rodrigo Abonia
- Department of Chemistry, Universidad del Valle, Cali AA 25360, Colombia; (J.R.-P.); (B.I.); (R.A.); (J.Q.)
| | - Jairo Quiroga
- Department of Chemistry, Universidad del Valle, Cali AA 25360, Colombia; (J.R.-P.); (B.I.); (R.A.); (J.Q.)
| | - Stephan Heeb
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| | - Paul Williams
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| | - Michael J. Stocks
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Miguel Cámara
- School of Life Sciences, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (F.S.); (E.V.O.); (J.-F.D.); (J.G.); (N.H.); (S.H.); (P.W.)
| |
Collapse
|
44
|
Horák R, Kvapil L, Motyka K, Slaninová L, Grepl M, Kořistek K, Urbášek M, Hradil P, Soural M. Synthesis of 2-alkenyl-3-hydroxyquinolin-4(1 H )-ones as promising antimicrobial and fluorescent agents. Tetrahedron 2018. [DOI: 10.1016/j.tet.2017.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
45
|
Schönauer E, Kany AM, Haupenthal J, Hüsecken K, Hoppe IJ, Voos K, Yahiaoui S, Elsässer B, Ducho C, Brandstetter H, Hartmann RW. Discovery of a Potent Inhibitor Class with High Selectivity toward Clostridial Collagenases. J Am Chem Soc 2017; 139:12696-12703. [PMID: 28820255 PMCID: PMC5607459 DOI: 10.1021/jacs.7b06935] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
Secreted virulence
factors like bacterial collagenases are conceptually
attractive targets for fighting microbial infections. However, previous
attempts to develop potent compounds against these metalloproteases
failed to achieve selectivity against human matrix metalloproteinases
(MMPs). Using a surface plasmon resonance-based screening complemented
with enzyme inhibition assays, we discovered an N-aryl mercaptoacetamide-based inhibitor scaffold that showed
sub-micromolar affinities toward collagenase H (ColH) from the human
pathogen Clostridium histolyticum. Moreover, these
inhibitors also efficiently blocked the homologous bacterial collagenases,
ColG from C. histolyticum, ColT from C. tetani, and ColQ1 from the Bacillus cereus strain Q1,
while showing negligible activity toward human MMPs-1, -2, -3, -7,
-8, and -14. The most active compound displayed a more than 1000-fold
selectivity over human MMPs. This selectivity can be rationalized
by the crystal structure of ColH with this compound, revealing a distinct
non-primed binding mode to the active site. The non-primed binding
mode presented here paves the way for the development of selective
broad-spectrum bacterial collagenase inhibitors with potential therapeutic
application in humans.
Collapse
Affiliation(s)
- Esther Schönauer
- Division of Structural Biology, Department of Molecular Biology, University of Salzburg , Billrothstrasse 11, 5020 Salzburg, Austria
| | - Andreas M Kany
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) , Campus E8.1, 66123 Saarbrücken, Germany
| | - Jörg Haupenthal
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) , Campus E8.1, 66123 Saarbrücken, Germany
| | - Kristina Hüsecken
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) , Campus E8.1, 66123 Saarbrücken, Germany
| | - Isabel J Hoppe
- Division of Structural Biology, Department of Molecular Biology, University of Salzburg , Billrothstrasse 11, 5020 Salzburg, Austria
| | - Katrin Voos
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University , Campus C2.3, 66123 Saarbrücken, Germany
| | - Samir Yahiaoui
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) , Campus E8.1, 66123 Saarbrücken, Germany
| | - Brigitta Elsässer
- Division of Structural Biology, Department of Molecular Biology, University of Salzburg , Billrothstrasse 11, 5020 Salzburg, Austria
| | - Christian Ducho
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University , Campus C2.3, 66123 Saarbrücken, Germany
| | - Hans Brandstetter
- Division of Structural Biology, Department of Molecular Biology, University of Salzburg , Billrothstrasse 11, 5020 Salzburg, Austria
| | - Rolf W Hartmann
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) , Campus E8.1, 66123 Saarbrücken, Germany.,Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University , Campus C2.3, 66123 Saarbrücken, Germany
| |
Collapse
|
46
|
Allegretta G, Maurer CK, Eberhard J, Maura D, Hartmann RW, Rahme L, Empting M. In-depth Profiling of MvfR-Regulated Small Molecules in Pseudomonas aeruginosa after Quorum Sensing Inhibitor Treatment. Front Microbiol 2017; 8:924. [PMID: 28596760 PMCID: PMC5442231 DOI: 10.3389/fmicb.2017.00924] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 05/08/2017] [Indexed: 01/02/2023] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative bacterium, which causes opportunistic infections in immuno-compromised individuals. Due to its multiple resistances toward antibiotics, the development of new drugs is required. Interfering with Quorum Sensing (QS), a cell-to-cell communication system, has shown to be highly efficient in reducing P. aeruginosa pathogenicity. One of its QS systems employs Pseudomonas Quinolone Signal (PQS) and 4-hydroxy-2-heptylquinoline (HHQ) as signal molecules. Both activate the transcriptional regulator MvfR (Multiple Virulence Factor Regulator), also called PqsR, driving the production of QS molecules as well as toxins and biofilm formation. The aim of this work was to elucidate the effects of QS inhibitors (QSIs), such as MvfR antagonists and PqsBC inhibitors, on the biosynthesis of the MvfR-regulated small molecules 2′-aminoacetophenone (2-AA), dihydroxyquinoline (DHQ), HHQ, PQS, and 4-hydroxy-2-heptylquinoline-N-oxide (HQNO). The employed synthetic MvfR antagonist fully inhibited pqs small molecule formation showing expected sigmoidal dose-response curves for 2-AA, HQNO, HHQ and PQS. Surprisingly, DHQ levels were enhanced at lower antagonist concentrations followed by a full suppression at higher QSI amounts. This particular bi-phasic profile hinted at the accumulation of a biosynthetic intermediate resulting in the observed overproduction of the shunt product DHQ. Additionally, investigations on PqsBC inhibitors showed a reduction of MvfR natural ligands, while increased 2-AA, DHQ and HQNO levels compared to the untreated cells were detected. Moreover, PqsBC inhibitors did not show any significant effect in PA14 pqsC mutant demonstrating their target selectivity. As 2-AA is important for antibacterial tolerance, the QSIs were evaluated in their capability to attenuate persistence. Indeed, persister cells were reduced along with 2-AA inhibition resulting from MvfR antagonism, but not from PqsBC inhibition. In conclusion, antagonizing MvfR using a dosage capable of fully suppressing this QS system will lead to a favorable therapeutic outcome as DHQ overproduction is avoided and bacterial persistence is reduced.
Collapse
Affiliation(s)
- Giuseppe Allegretta
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrücken, Germany
| | - Christine K Maurer
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrücken, Germany
| | - Jens Eberhard
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrücken, Germany
| | - Damien Maura
- Department of Surgery and Department of Microbiology and Immunobiology, Harvard Medical School, BostonMA, United States.,Department of Surgery, Center for Surgery, Innovation and Bioengineering, Massachusetts General Hospital, BostonMA, United States.,Shriners Hospitals for Children, BostonMA, United States
| | - Rolf W Hartmann
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrücken, Germany.,Pharmaceutical and Medicinal Chemistry, Saarland UniversitySaarbrücken, Germany
| | - Laurence Rahme
- Department of Surgery and Department of Microbiology and Immunobiology, Harvard Medical School, BostonMA, United States.,Department of Surgery, Center for Surgery, Innovation and Bioengineering, Massachusetts General Hospital, BostonMA, United States.,Shriners Hospitals for Children, BostonMA, United States
| | - Martin Empting
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrücken, Germany
| |
Collapse
|
47
|
Maura D, Drees SL, Bandyopadhaya A, Kitao T, Negri M, Starkey M, Lesic B, Milot S, Déziel E, Zahler R, Pucci M, Felici A, Fetzner S, Lépine F, Rahme LG. Polypharmacology Approaches against the Pseudomonas aeruginosa MvfR Regulon and Their Application in Blocking Virulence and Antibiotic Tolerance. ACS Chem Biol 2017; 12:1435-1443. [PMID: 28379691 DOI: 10.1021/acschembio.6b01139] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pseudomonas aeruginosa is an important nosocomial pathogen that is frequently recalcitrant to available antibiotics, underlining the urgent need for alternative therapeutic options against this pathogen. Targeting virulence functions is a promising alternative strategy as it is expected to generate less-selective resistance to treatment compared to antibiotics. Capitalizing on our nonligand-based benzamide-benzimidazole (BB) core structure compounds reported to efficiently block the activity of the P. aeruginosa multiple virulence factor regulator MvfR, here we report the first class of inhibitors shown to interfere with PqsBC enzyme activity, responsible for the synthesis of the MvfR activating ligands HHQ and PQS, and the first to target simultaneously MvfR and PqsBC activity. The use of these compounds reveals that inhibiting PqsBC is sufficient to block P. aeruginosa's acute virulence functions, as the synthesis of MvfR ligands is inhibited. Our results show that MvfR remains the best target of this QS pathway, as we show that antagonists of this target block both acute and persistence-related functions. The structural properties of the compounds reported in this study provide several insights that are instrumental for the design of improved MvfR regulon inhibitors against both acute and persistent P. aeruginosa infections. Moreover, the data presented offer the possibility of a polypharmacology approach of simultaneous silencing two targets in the same pathway. Such a combined antivirulence strategy holds promise in increasing therapeutic efficacy and providing alternatives in the event of a single target's resistance development.
Collapse
Affiliation(s)
- Damien Maura
- Shriners Hospitals for Children Boston, Boston, Massachusetts 02114, United States
| | - Steffen L. Drees
- Institute
for Molecular Microbiology and Biotechnology, University of Münster, 48149 Münster, Germany
| | | | - Tomoe Kitao
- Shriners Hospitals for Children Boston, Boston, Massachusetts 02114, United States
| | | | - Melissa Starkey
- Shriners Hospitals for Children Boston, Boston, Massachusetts 02114, United States
| | - Biliana Lesic
- Shriners Hospitals for Children Boston, Boston, Massachusetts 02114, United States
| | - Sylvain Milot
- INRS Institut Armand Frappier, Laval, Quebec H7V 1B7, Canada
| | - Eric Déziel
- INRS Institut Armand Frappier, Laval, Quebec H7V 1B7, Canada
| | - Robert Zahler
- Spero Therapeutics, Cambridge, Massachusetts 02139, United States
| | - Mike Pucci
- Spero Therapeutics, Cambridge, Massachusetts 02139, United States
| | | | - Susanne Fetzner
- Institute
for Molecular Microbiology and Biotechnology, University of Münster, 48149 Münster, Germany
| | - François Lépine
- INRS Institut Armand Frappier, Laval, Quebec H7V 1B7, Canada
| | - Laurence G. Rahme
- Shriners Hospitals for Children Boston, Boston, Massachusetts 02114, United States
| |
Collapse
|
48
|
Aleksić I, Šegan S, Andrić F, Zlatović M, Moric I, Opsenica DM, Senerovic L. Long-Chain 4-Aminoquinolines as Quorum Sensing Inhibitors in Serratia marcescens and Pseudomonas aeruginosa. ACS Chem Biol 2017; 12:1425-1434. [PMID: 28350449 DOI: 10.1021/acschembio.6b01149] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Antibiotic resistance has become a serious global threat to public health; therefore, improved strategies and structurally novel antimicrobials are urgently needed to combat infectious diseases. Here we report a new type of highly potent 4-aminoquinoline derivatives as quorum sensing inhibitors in Serratia marcescens and Pseudomonas aeruginosa, exhibiting weak bactericidal activities (minimum inhibitory concentration (MIC) > 400 μM). Through detailed structure-activity study, we have identified 7-Cl and 7-CF3 substituted N-dodecylamino-4-aminoquinolines (5 and 10) as biofilm formation inhibitors with 50% biofilm inhibition at 69 μM and 63 μM in S. marcescens and P. aeruginosa, respectively. These two compounds, 5 and 10, are the first quinoline derivatives with anti-biofilm formation activity reported in S. marcescens. Quantitative structure-activity relationship (QSAR) analysis identified structural descriptors such as Wiener indices, hyper-distance-path index (HDPI), mean topological charge (MTC), topological charge index (TCI), and log D(o/w)exp as the most influential in biofilm inhibition in this bacterial species. Derivative 10 is one of the most potent quinoline type inhibitors of pyocyanin production described so far (IC50 = 2.5 μM). While we have demonstrated that 5 and 10 act as Pseudomonas quinolone system (PQS) antagonists, the mechanism of inhibition of S. marcescens biofilm formation with these compounds remains open since signaling similar to P. aeruginosa PQS system has not yet been described in Serratia and activity of these compounds on acylhomoserine lactone (AHL) signaling has not been detected. Our data show that 7-Cl and 7-CF3 substituted N-dodecylamino-4-aminoquinolines present the promising scaffolds for developing antivirulence and anti-biofilm formation agents against multidrug-resistant bacterial species.
Collapse
Affiliation(s)
- Ivana Aleksić
- Institute
of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia
| | - Sandra Šegan
- Institute
of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, P.O. Box 473, 11000 Belgrade, Serbia
| | - Filip Andrić
- Institute
of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, P.O. Box 473, 11000 Belgrade, Serbia
| | - Mario Zlatović
- Faculty
of Chemistry, University of Belgrade, Studentski trg 12-16, P.O. Box 51, 11158 Belgrade, Serbia
| | - Ivana Moric
- Institute
of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia
| | - Dejan M. Opsenica
- Institute
of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, P.O. Box 473, 11000 Belgrade, Serbia
| | - Lidija Senerovic
- Institute
of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia
| |
Collapse
|
49
|
Abstract
Bacteria use quorum sensing to orchestrate gene expression programmes that underlie collective behaviours. Quorum sensing relies on the production, release, detection and group-level response to extracellular signalling molecules, which are called autoinducers. Recent work has discovered new autoinducers in Gram-negative bacteria, shown how these molecules are recognized by cognate receptors, revealed new regulatory components that are embedded in canonical signalling circuits and identified novel regulatory network designs. In this Review we examine how, together, these features of quorum sensing signal-response systems combine to control collective behaviours in Gram-negative bacteria and we discuss the implications for host-microbial associations and antibacterial therapy.
Collapse
|
50
|
Zhou J, Bi S, Chen H, Chen T, Yang R, Li M, Fu Y, Jia AQ. Anti-Biofilm and Antivirulence Activities of Metabolites from Plectosphaerella cucumerina against Pseudomonas aeruginosa. Front Microbiol 2017; 8:769. [PMID: 28515715 PMCID: PMC5413567 DOI: 10.3389/fmicb.2017.00769] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/13/2017] [Indexed: 12/25/2022] Open
Abstract
This study reported the efficacy of the metabolites of Plectosphaerella cucumerina, one phyllosphere fungus from Orychophragmus violaceus, against Pseudomonas aeruginosa quorum sensing (QS) and QS-regulated biofilms. The minimum inhibitory concentration (MIC) of the ethyl acetate (EtOAc) extract from P. cucumerina against P. aeruginosa PAO1 was 1.25 mg mL−1. At sub-MIC concentrations, P. cucumerina extract (0.25–1 mg mL−1) not only inhibited biofilm formation but also disrupted preformed biofilms of P. aeruginosa PAO1 without affecting its growth. Fluorescence and scanning electron microscope (SEM) showed architectural disruption of the biofilms when treated with P. cucumerina metabolites. Further investigation demonstrated that metabolites in P. cucumerina attenuated the QS-dependent virulence factors. LC-MS/MS spectra coupled with experimentally standard samples suggested that patulin and emodin might act as the principal components possessing anti-biofilm and antivirulence activities. This is the first report of (1) the isolation of P. cucumerina from the phyllosphere of O. violaceus and (2) anti-biofilm, antivirulence, and biofilm disruption activities of this fungus. Thus, this study provides fascinating new pathways for screening antipathogenic agents.
Collapse
Affiliation(s)
- Jinwei Zhou
- School of Environmental and Biological Engineering, Nanjing University of Science and TechnologyNanjing, China
| | - Shiyuan Bi
- School of Environmental and Biological Engineering, Nanjing University of Science and TechnologyNanjing, China
| | - Hongjuan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing UniversityNanjing, China
| | - Tongtong Chen
- School of Environmental and Biological Engineering, Nanjing University of Science and TechnologyNanjing, China
| | - Rui Yang
- School of Environmental and Biological Engineering, Nanjing University of Science and TechnologyNanjing, China
| | - Minghui Li
- School of Environmental and Biological Engineering, Nanjing University of Science and TechnologyNanjing, China
| | - Yonghong Fu
- School of Environmental and Biological Engineering, Nanjing University of Science and TechnologyNanjing, China
| | - Ai-Qun Jia
- School of Environmental and Biological Engineering, Nanjing University of Science and TechnologyNanjing, China.,State Key Laboratory of Marine Resource Utilization in South China Sea, Key Laboratory of Tropical Biological Resources of Ministry Education, Hainan UniversityHaikou, China
| |
Collapse
|