1
|
Zhang P, Zeng P, Lai CKC, Ip M, To KKW, Zuo Z, Xia J, Leung SSY. Synergism of colistin and globular endolysins against multidrug-resistant gram-negative bacteria. Int J Biol Macromol 2024; 278:134670. [PMID: 39151868 DOI: 10.1016/j.ijbiomac.2024.134670] [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: 02/09/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Endolysins (lysins), a novel class of antibacterial agents derived from bacteriophages, efficiently lyse bacteria by degrading the peptidoglycan layer within the bacterial wall. Colistin, a classic peptide antibiotic with the ability to permeabilize the outer membrane, has recently shown great promise in synergizing with lysins against gram-negative bacteria. However, the exact mechanisms responsible for their synergy remain unclear. Here, we first demonstrated the synergistic bacterial killing of various lysin and colistin combinations. With a model lysin, LysAB2, we then confirmed that there is a threshold concentration of colistin causing sufficient permeabilization of the outer membrane for lysin to access the peptidoglycan layer and subsequently exert its lytic ability. The threshold colistin concentrations were found to range 0.2-0.8 μM for the tested bacteria, with the exact value largely depending on the density of lipopolysaccharides on the outer membrane. Beyond the threshold colistin level, LysAB2 could synergize with colistin at a concentration as low as 0.31 μM. Next, we proved for the first time that lysin-induced degradation of the peptidoglycan layer facilitated the disruption of cytoplasmic membrane by colistin, elevated the level of reactive oxygen species in bacterial cells, and boosted the killing effect of colistin. Additionally, the colistin-lysin combination could effectively eliminate established biofilms due to the biofilm dispersal ability of lysin. The in-vivo efficacy was preliminary confirmed in a Galleria mellonella infection model for combination with colistin doses (≥ 1.8 μg/larvae), which could reach beyond the threshold concentration, and a fixed LysAB2 dose (10 μg/larvae). In summary, our study provided the first experimental evidence unravelling the mechanisms behind the synergy of colistin and lysins. All these findings provided important insights in guiding the dosing strategy for applying this combination in future development.
Collapse
Affiliation(s)
- Pengfei Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Ping Zeng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Christopher K C Lai
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong.
| | - Sharon S Y Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
2
|
Schum D, Elsen FAV, Ruddell S, Schorpp K, Junca H, Müsken M, Chen SY, Fiedler MK, Pickl T, Pieper DH, Hadian K, Zacharias M, Sieber SA. Screening Privileged Alkyl Guanidinium Motifs under Host-Mimicking Conditions Reveals a Novel Antibiotic with an Unconventional Mode of Action. JACS AU 2024; 4:3125-3134. [PMID: 39211621 PMCID: PMC11350587 DOI: 10.1021/jacsau.4c00449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 09/04/2024]
Abstract
Screening large molecule libraries against pathogenic bacteria is often challenged by a low hit rate due to limited uptake, underrepresentation of antibiotic structural motifs, and assays that do not resemble the infection conditions. To address these limitations, we present a screen of a focused library of alkyl guanidinium compounds, a structural motif associated with antibiotic activity and enhanced uptake, under host-mimicking infection conditions against a panel of disease-associated bacteria. Several hit molecules were identified with activities against Gram-positive and Gram-negative bacteria, highlighting the fidelity of the general concept. We selected one compound (L15) for in-depth mode of action studies that exhibited bactericidal activity against methicillin-resistant Staphylococcus aureus USA300 with a minimum inhibitory concentration of 1.5 μM. Structure-activity relationship studies confirmed the necessity of the guanidinium motif for antibiotic activity. The mode of action was investigated using affinity-based protein profiling with an L15 probe and identified the signal peptidase IB (SpsB) as the most promising hit. Validation by activity assays, binding site identification, docking, and molecular dynamics simulations demonstrated SpsB activation by L15, a recently described mechanism leading to the dysregulation of protein secretion and cell death. Overall, this study highlights the need for unconventional screening strategies to identify novel antibiotics.
Collapse
Affiliation(s)
- Dominik Schum
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Franziska A. V. Elsen
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Stuart Ruddell
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Kenji Schorpp
- Helmholtz
Zentrum München, Research Unit Signaling and Translation, Ingolstädter Landstraße
1, Neuherberg, Munich 85764, Germany
| | - Howard Junca
- Helmholtz
Centre for Infection Research, Microbial Interactions and Processes, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Helmholtz
Centre for Infection Research, Central Facility for Microscopy, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Shu-Yu Chen
- TUM
School of Natural Sciences, Department of Bioscience, Theoretical Biophysics (T38), Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Michaela K. Fiedler
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Thomas Pickl
- TUM School
of Natural Sciences, Department of Chemistry, Catalysis Research Center (CRC), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 1, Garching 85748, Germany
| | - Dietmar H. Pieper
- Helmholtz
Centre for Infection Research, Microbial Interactions and Processes, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Kamyar Hadian
- Helmholtz
Zentrum München, Research Unit Signaling and Translation, Ingolstädter Landstraße
1, Neuherberg, Munich 85764, Germany
| | - Martin Zacharias
- TUM
School of Natural Sciences, Department of Bioscience, Theoretical Biophysics (T38), Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Stephan A. Sieber
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| |
Collapse
|
3
|
Buchholz KR, Reichelt M, Johnson MC, Robinson SJ, Smith PA, Rutherford ST, Quinn JG. Potent activity of polymyxin B is associated with long-lived super-stoichiometric accumulation mediated by weak-affinity binding to lipid A. Nat Commun 2024; 15:4733. [PMID: 38830951 PMCID: PMC11148078 DOI: 10.1038/s41467-024-49200-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Polymyxins are gram-negative antibiotics that target lipid A, the conserved membrane anchor of lipopolysaccharide in the outer membrane. Despite their clinical importance, the molecular mechanisms underpinning polymyxin activity remain unresolved. Here, we use surface plasmon resonance to kinetically interrogate interactions between polymyxins and lipid A and derive a phenomenological model. Our analyses suggest a lipid A-catalyzed, three-state mechanism for polymyxins: transient binding, membrane insertion, and super-stoichiometric cluster accumulation with a long residence time. Accumulation also occurs for brevicidine, another lipid A-targeting antibacterial molecule. Lipid A modifications that impart polymyxin resistance and a non-bactericidal polymyxin derivative exhibit binding that does not evolve into long-lived species. We propose that transient binding to lipid A permeabilizes the outer membrane and cluster accumulation enables the bactericidal activity of polymyxins. These findings could establish a blueprint for discovery of lipid A-targeting antibiotics and provide a generalizable approach to study interactions with the gram-negative outer membrane.
Collapse
Affiliation(s)
- Kerry R Buchholz
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA.
| | - Mike Reichelt
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Matthew C Johnson
- Department of Structural Biology, Genentech, Inc., South San Francisco, CA, USA
| | - Sarah J Robinson
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, CA, USA
| | - Peter A Smith
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA
- Revagenix, Inc., San Mateo, CA, USA
| | - Steven T Rutherford
- Department of Infectious Diseases, Genentech, Inc., South San Francisco, CA, USA.
| | - John G Quinn
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
4
|
Varache M, Rizzo S, Sayers EJ, Newbury L, Mason A, Liao CT, Chiron E, Bourdiec N, Jones A, Fraser DJ, Taylor PR, Jones AT, Thomas DW, Ferguson EL. Dextrin conjugation to colistin inhibits its toxicity, cellular uptake and acute kidney injury in vivo. RSC PHARMACEUTICS 2024; 1:68-79. [PMID: 38646595 PMCID: PMC11024668 DOI: 10.1039/d3pm00014a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/04/2024] [Indexed: 04/23/2024]
Abstract
The acute kidney injury (AKI) and dose-limiting nephrotoxicity, which occurs in 20-60% of patients following systemic administration of colistin, represents a challenge in the effective treatment of multi-drug resistant Gram-negative infections. To reduce clinical toxicity of colistin and improve targeting to infected/inflamed tissues, we previously developed dextrin-colistin conjugates, whereby colistin is designed to be released by amylase-triggered degradation of dextrin in infected and inflamed tissues, after passive targeting by the enhanced permeability and retention effect. Whilst it was evident in vitro that polymer conjugation can reduce toxicity and prolong plasma half-life, without significant reduction in antimicrobial activity of colistin, it was unclear how dextrin conjugation would alter cellular uptake and localisation of colistin in renal tubular cells in vivo. We discovered that dextrin conjugation effectively reduced colistin's toxicity towards human kidney proximal tubular epithelial cells (HK-2) in vitro, which was mirrored by significantly less cellular uptake of Oregon Green (OG)-labelled dextrin-colistin conjugate, when compared to colistin. Using live-cell confocal imaging, we revealed localisation of both, free and dextrin-bound colistin in endolysosome compartments of HK-2 and NRK-52E cells. Using a murine AKI model, we demonstrated dextrin-colistin conjugation dramatically diminishes both proximal tubular injury and renal accumulation of colistin. These findings reveal new insight into the mechanism by which dextrin conjugation can overcome colistin's renal toxicity and show the potential of polymer conjugation to improve the side effect profile of nephrotoxic drugs.
Collapse
Affiliation(s)
- Mathieu Varache
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Siân Rizzo
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Edward J Sayers
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University CF10 3NB UK
| | - Lucy Newbury
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University Cardiff CF14 4XN UK
| | - Anna Mason
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University Cardiff CF14 4XN UK
| | - Chia-Te Liao
- Systems Immunity Research Institute, Division of Infection and Immunity, School of Medicine, Cardiff University Cardiff CF14 4XN UK
| | - Emilie Chiron
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Nathan Bourdiec
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| | - Adam Jones
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
- Cellular Pathology Department, University Dental Hospital, Cardiff and Vale University Health Board Cardiff CF14 4XY UK
| | - Donald J Fraser
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University Cardiff CF14 4XN UK
| | - Philip R Taylor
- Systems Immunity Research Institute, Division of Infection and Immunity, School of Medicine, Cardiff University Cardiff CF14 4XN UK
- UK Dementia Research Institute at Cardiff Hadyn Ellis Building Maindy Road Cardiff CF24 4HQ UK
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University CF10 3NB UK
| | - David W Thomas
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
- Systems Immunity Research Institute, Division of Infection and Immunity, School of Medicine, Cardiff University Cardiff CF14 4XN UK
| | - Elaine L Ferguson
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University Heath Park Cardiff CF14 4XY UK
| |
Collapse
|
5
|
Mercer JAM, DeCarlo SJ, Roy Burman SS, Sreekanth V, Nelson AT, Hunkeler M, Chen PJ, Donovan KA, Kokkonda P, Tiwari PK, Shoba VM, Deb A, Choudhary A, Fischer ES, Liu DR. Continuous evolution of compact protein degradation tags regulated by selective molecular glues. Science 2024; 383:eadk4422. [PMID: 38484051 PMCID: PMC11203266 DOI: 10.1126/science.adk4422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/09/2024] [Indexed: 03/19/2024]
Abstract
Conditional protein degradation tags (degrons) are usually >100 amino acids long or are triggered by small molecules with substantial off-target effects, thwarting their use as specific modulators of endogenous protein levels. We developed a phage-assisted continuous evolution platform for molecular glue complexes (MG-PACE) and evolved a 36-amino acid zinc finger (ZF) degron (SD40) that binds the ubiquitin ligase substrate receptor cereblon in complex with PT-179, an orthogonal thalidomide derivative. Endogenous proteins tagged in-frame with SD40 using prime editing are degraded by otherwise inert PT-179. Cryo-electron microscopy structures of SD40 in complex with ligand-bound cereblon revealed mechanistic insights into the molecular basis of SD40's activity and specificity. Our efforts establish a system for continuous evolution of molecular glue complexes and provide ZF tags that overcome shortcomings associated with existing degrons.
Collapse
Affiliation(s)
- Jaron A. M. Mercer
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138
| | - Stephan J. DeCarlo
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138
| | - Shourya S. Roy Burman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Vedagopuram Sreekanth
- Chemical Biology and Therapeutics Science, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Medicine, Harvard Medical School, Boston, MA 02115
- Divisions of Renal Medicine and Engineering, Brigham and Women’s Hospital, Boston, MA 02115
| | - Andrew T. Nelson
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138
| | - Moritz Hunkeler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Peter J. Chen
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138
| | - Katherine A. Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Praveen Kokkonda
- Chemical Biology and Therapeutics Science, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Praveen K. Tiwari
- Chemical Biology and Therapeutics Science, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Medicine, Harvard Medical School, Boston, MA 02115
- Divisions of Renal Medicine and Engineering, Brigham and Women’s Hospital, Boston, MA 02115
| | - Veronika M. Shoba
- Chemical Biology and Therapeutics Science, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Arghya Deb
- Chemical Biology and Therapeutics Science, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Amit Choudhary
- Chemical Biology and Therapeutics Science, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Medicine, Harvard Medical School, Boston, MA 02115
- Divisions of Renal Medicine and Engineering, Brigham and Women’s Hospital, Boston, MA 02115
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138
| |
Collapse
|
6
|
Cai J, Shi J, Chen C, He M, Wang Z, Liu Y. Structural-Activity Relationship-Inspired the Discovery of Saturated Fatty Acids as Novel Colistin Enhancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302182. [PMID: 37552809 PMCID: PMC10582468 DOI: 10.1002/advs.202302182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/19/2023] [Indexed: 08/10/2023]
Abstract
The emergence and prevalence of mobile colistin resistance gene mcr have dramatically compromised the clinical efficacy of colistin, a cyclopeptide antibiotic considered to be the last option for treating different-to-treat infections. The combination strategy provides a productive and cost-effective strategy to expand the lifespan of existing antibiotics. Structural-activity relationship analysis of polymyxins indicates that the fatty acyl chain plays an indispensable role in their antibacterial activity. Herein, it is revealed that three saturated fatty acids (SFAs), especially sodium caprate (SC), substantially potentiate the antibacterial activity of colistin against mcr-positive bacteria. The combination of SFAs and colistin effectively inhibits biofilm formation and eliminates matured biofilms, and is capable of preventing the emergence and spread of mobile colistin resistance. Mechanistically, the addition of SFAs reduces lipopolysaccharide (LPS) modification by simultaneously promoting LPS biosynthesis and inhibiting the activity of MCR enzyme, enhance bacterial membrane damage, and impair the proton motive force-dependent efflux pump, thereby boosting the action of colistin. In three animal models of infection by mcr-positive pathogens, SC combined with colistin exhibit an excellent therapeutic effect. These findings indicate the therapeutic potential of SFAs as novel antibiotic adjuvants for the treatment of infections caused by multidrug-resistant bacteria in combination with colistin.
Collapse
Affiliation(s)
- Jinju Cai
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Jingru Shi
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Chen Chen
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Mengping He
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
| | - Zhiqiang Wang
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhou225009China
- Institute of Comparative MedicineYangzhou UniversityYangzhou225009China
| | - Yuan Liu
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesCollege of Veterinary MedicineYangzhou UniversityYangzhou225009China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safetythe Ministry of Education of ChinaYangzhou UniversityYangzhou225009China
- Institute of Comparative MedicineYangzhou UniversityYangzhou225009China
| |
Collapse
|
7
|
Meir O, Zaknoon F, Mor A. An efflux-susceptible antibiotic-adjuvant with systemic efficacy against mouse infections. Sci Rep 2022; 12:17673. [PMID: 36271103 PMCID: PMC9586926 DOI: 10.1038/s41598-022-21526-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
Scarcity of effective treatments against sepsis is daunting, especially under the contemporary standpoints on antibiotics resistance, entailing the development of alternative treatment strategies. Here, we describe the design and antibiotic adjuvant properties of a new lipopeptide-like pentamer, decanoyl-bis.diaminobutyrate-aminododecanoyl-diaminobutyrate-amide (C10BBc12B), whose sub-maximal tolerated doses combinations with inefficient antibiotics demonstrated systemic efficacies in murine models of peritonitis-sepsis and urinary-tract infections. Attempts to shed light into the mechanism of action using membrane-active fluorescent probes, suggest outer-membrane interactions to dominate the pentamer's adjuvant properties, which were not associated with typical inner-membrane damages or with delayed bacterial growth. Yet, checkerboard titrations with low micromolar concentrations of C10BBc12B exhibited unprecedented capacities in potentiation of hydrophobic antibiotics towards Gram-negative ESKAPE pathogens, with an apparent low propensity for prompting resistance to the antibiotics. Assessment of the pentamer's potentiating activities upon efflux inhibition incites submission of a hitherto unreported, probable action mechanism implicating the pentamer's de-facto capacity to hijack bacterial efflux pumps for boosting its adjuvant activity through repetitive steps including outer-membrane adhesion, translocation and subsequent expulsion.
Collapse
Affiliation(s)
- Ohad Meir
- grid.6451.60000000121102151Faculty of Biotechnology and Food Engineering, Technion – Israel Institute of Technology, 3200003 Haifa, Israel
| | - Fadia Zaknoon
- grid.6451.60000000121102151Faculty of Biotechnology and Food Engineering, Technion – Israel Institute of Technology, 3200003 Haifa, Israel
| | - Amram Mor
- grid.6451.60000000121102151Faculty of Biotechnology and Food Engineering, Technion – Israel Institute of Technology, 3200003 Haifa, Israel
| |
Collapse
|
8
|
Brennan-Krohn T, Grote A, Rodriguez S, Kirby JE, Earl AM. Transcriptomics Reveals How Minocycline-Colistin Synergy Overcomes Antibiotic Resistance in Multidrug-Resistant Klebsiella pneumoniae. Antimicrob Agents Chemother 2022; 66:e0196921. [PMID: 35041511 PMCID: PMC8923212 DOI: 10.1128/aac.01969-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 11/20/2022] Open
Abstract
Multidrug-resistant Gram-negative bacteria are a rapidly growing public health threat, and the development of novel antimicrobials has failed to keep pace with their emergence. Synergistic combinations of individually ineffective drugs present a potential solution, yet little is understood about the mechanisms of most such combinations. Here, we show that the combination of colistin (polymyxin E) and minocycline has a high rate of synergy against colistin-resistant and minocycline-intermediate or -resistant strains of Klebsiella pneumoniae. Furthermore, using transcriptome sequencing (RNA-Seq), we characterized the transcriptional profiles of these strains when treated with the drugs individually and in combination. We found a striking similarity between the transcriptional profiles of bacteria treated with the combination of colistin and minocycline at individually subinhibitory concentrations and those of the same isolates treated with minocycline alone. We observed a similar pattern with the combination of polymyxin B nonapeptide (a polymyxin B analogue that lacks intrinsic antimicrobial activity) and minocycline. We also found that genes involved in polymyxin resistance and peptidoglycan biosynthesis showed significant differential gene expression in the different treatment conditions, suggesting possible mechanisms for the antibacterial activity observed in the combination. These findings suggest that the synergistic activity of this combination against bacteria resistant to each drug alone involves sublethal outer membrane disruption by colistin, which permits increased intracellular accumulation of minocycline.
Collapse
Affiliation(s)
- Thea Brennan-Krohn
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandra Grote
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shade Rodriguez
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - James E. Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ashlee M. Earl
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
Khusnutdinova EF, Sinou V, Babkov DA, Kazakova O, Brunel JM. Development of New Antimicrobial Oleanonic Acid Polyamine Conjugates. Antibiotics (Basel) 2022; 11:antibiotics11010094. [PMID: 35052971 PMCID: PMC8772916 DOI: 10.3390/antibiotics11010094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
A series of oleanolic acid derivatives holding oxo- or 3-N-polyamino-3-deoxy-substituents at C3 as well as carboxamide function at C17 with different long chain polyamines have been synthesized and evaluated for antimicrobial activities. Almost all series presented good to moderate activity against Gram-positive S. aureus, S. faecalis and B. cereus bacteria with minimum inhibitory concentration (MIC) values from 3.125 to 200 µg/mL. Moreover, compounds possess important antimicrobial activities against Gram-negative E. coli, P. aeruginosa, S. enterica, and EA289 bacteria with MICs ranging from 6.25 to 200 µg/mL. The testing of ability to restore antibiotic activity of doxycycline and erythromycin at a 2 µg/mL concentration in a synergistic assay showed that oleanonic acid conjugate with spermine spacered through propargylamide led to a moderate improvement in terms of antimicrobial activities of the different selected combinations against both P. aeruginosa and E. coli. The study of mechanism of action of the lead conjugate 2i presenting a N-methyl norspermidine moiety showed the effect of disruption of the outer bacterial membrane of P. aeruginosa PA01 cells. Computational ADMET profiling renders this compound as a suitable starting point for pharmacokinetic optimization. These results give confidence to the successful outcome of bioconjugation of polyamines and oleanane-type triterpenoids in the development of antimicrobial agents.
Collapse
Affiliation(s)
- Elmira F. Khusnutdinova
- Ufa Institute of Chemistry UFRC RAS, 71 pr. Oktyabrya, 450054 Ufa, Russia;
- Aix Marseille Univ, INSERM, SSA, MCT, 13385 Marseille, France;
- Correspondence: (E.F.K.); (J.M.B.)
| | - Véronique Sinou
- Aix Marseille Univ, INSERM, SSA, MCT, 13385 Marseille, France;
| | - Denis A. Babkov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, Novorossiyskaya st. 39, 400087 Volgograd, Russia;
| | - Oxana Kazakova
- Ufa Institute of Chemistry UFRC RAS, 71 pr. Oktyabrya, 450054 Ufa, Russia;
| | - Jean Michel Brunel
- Aix Marseille Univ, INSERM, SSA, MCT, 13385 Marseille, France;
- Correspondence: (E.F.K.); (J.M.B.)
| |
Collapse
|
10
|
Zeng J, Li Z, Jiang H, Wang X. Progress on photocatalytic semiconductor hybrids for bacterial inactivation. MATERIALS HORIZONS 2021; 8:2964-3008. [PMID: 34609391 DOI: 10.1039/d1mh00773d] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Due to its use of green and renewable energy and negligible bacterial resistance, photocatalytic bacterial inactivation is to be considered a promising sterilization process. Herein, we explore the relevant mechanisms of the photoinduced process on the active sites of semiconductors with an emphasis on the active sites of semiconductors, the photoexcited electron transfer, ROS-induced toxicity and interactions between semiconductors and bacteria. Pristine semiconductors such as metal oxides (TiO2 and ZnO) have been widely reported; however, they suffer some drawbacks such as narrow optical response and high photogenerated carrier recombination. Herein, some typical modification strategies will be discussed including noble metal doping, ion doping, hybrid heterojunctions and dye sensitization. Besides, the biosafety and biocompatibility issues of semiconductor materials are also considered for the evaluation of their potential for further biomedical applications. Furthermore, 2D materials have become promising candidates in recent years due to their wide optical response to NIR light, superior antibacterial activity and favorable biocompatibility. Besides, the current research limitations and challenges are illustrated to introduce the appealing directions and design considerations for the future development of photocatalytic semiconductors for antibacterial applications.
Collapse
Affiliation(s)
- Jiayu Zeng
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Ziming Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
11
|
Maliszewska I, Goldeman W. Pentamidine enhances photosensitization of Acinetobacter baumannii using diode lasers with emission of light at wavelength of ʎ = 405 nm and ʎ = 635 nm. Photodiagnosis Photodyn Ther 2021; 34:102242. [PMID: 33662618 DOI: 10.1016/j.pdpdt.2021.102242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/22/2021] [Accepted: 02/26/2021] [Indexed: 12/01/2022]
Abstract
Antimicrobial photodynamic inactivation is currently one of the most promising trends in the modern bactericidal protocols. Under the conditions defined in our studies, we found that in vitro photosensitization of A. baumannii with 5-ALA as a precursor of protoporphyrin IX (photosensitizer) reduces the concentration of viable cells in planktonic cultures, and this process can be strongly enhanced by pentamidine. Diode lasers with the peak-power wavelength of ʎ = 405 nm (radiation intensity of 26 mW cm-2) and ʎ = 635 nm (radiation intensity of 55 mW cm-2) were used in this study. It was found that a blue laser light (energy fluence of 64 J cm-2; no external photosensitizer) in the presence of pentamidine resulted in a reduction of CFU of 99.992 % compared to 99.97 % killing without pentamidine. When a red laser light was used in the experiments (energy fluence of 136 J cm-2; no external photosensitizer), the mortality rate was 99.98 % in the presence of pentamidine compared to 99.93 % of those killed without the addition of this drug. The lethal effect with 5-ALA was achieved under blue light fluence of 16 J cm-2 (in the presence of pentamidine) and 32 J cm-2 (without pentamidine). In the case of laser light of 635 nm, the lethal effect with 5-ALA was attained with energy fluence of 51 J cm-2 (with pentamidine) and 102 J cm-2 (without pentamidine). The possible roles of pentamidine in enhancing photodynamic inactivation of A. baumannii have been discussed.
Collapse
Affiliation(s)
- Irena Maliszewska
- Department of Organic and Medicinal Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland.
| | - Waldemar Goldeman
- Department of Organic and Medicinal Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| |
Collapse
|
12
|
Comparative genomics of the sequential Pseudomonas aeruginosa isolates obtained from the continuous imipenem stress evolution. Appl Microbiol Biotechnol 2020; 104:10655-10667. [PMID: 33151366 DOI: 10.1007/s00253-020-10994-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/29/2020] [Accepted: 10/31/2020] [Indexed: 12/24/2022]
Abstract
Pseudomonas aeruginosa is a major opportunistic human pathogen that causes nosocomial infections, and the proportion of carbapenem resistance has recently dramatically increased in P. aeruginosa due to the overuse of them. In this study, strains G10 and G20, with minimum inhibitory concentration (MIC) of imipenem of 16 μg/ml and more than 32 μg/ml, were isolated during continuous subculture of cells exposed to stepwise increasing concentrations of imipenem, respectively. The genomes of G10 and G20 were sequenced and compared with parental strain (P. aeruginosa ATCC 27853, G0). There were 59, 59, and 58 genes involved in antibiotic resistance which were predicted in G0, G10, and G20, respectively, while 374, 366, and 363 genes involved in virulence factors were identified among these three strains. Due to the significantly different MICs of imipenem and highly similar profiles of antibiotic resistance and virulence factors related genes among three strains, the specific genetic variations that occurred were identified and compared, including single nucleotide polymorphisms (SNPs), insertions and deletions (InDels), and structural variations (SVs). The increase in the MIC of imipenem was proposed to be linked to mutations involved in polyamine biosynthesis, biofilm formation, OprD, and efflux pump functions. This study aims to clarify the underlying mechanism of imipenem resistance and provide alternative strategies for reducing resistance in P. aeruginosa. KEY POINTS: • Strains with different imipenem MIC were obtained via laboratory selection evolution. • Whole genomes of two strains with different MIC of imipenem were sequenced. • Underlying mechanism of imipenem resistance was clarified via comparative genomics.
Collapse
|
13
|
Hussein M, Hu X, Paulin OK, Crawford S, Tony Zhou Q, Baker M, Schneider-Futschik EK, Zhu Y, Li J, Velkov T. Polymyxin B combinations with FDA-approved non-antibiotic phenothiazine drugs targeting multi-drug resistance of Gram-negative pathogens. Comput Struct Biotechnol J 2020; 18:2247-2258. [PMID: 32952938 PMCID: PMC7481501 DOI: 10.1016/j.csbj.2020.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 12/19/2022] Open
Abstract
The status quo for combating uprising antibacterial resistance is to employ synergistic combinations of antibiotics. Nevertheless, the currently available combination therapies are fast becoming untenable. Combining antibiotics with various FDA-approved non-antibiotic drugs has emerged as a novel strategy against otherwise untreatable multi-drug resistant (MDR) pathogens. The apex of this study was to investigate the mechanisms of antibacterial synergy of the combination of polymyxin B with the phenothiazines against the MDR Gram-negative pathogens Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa. The synergistic antibacterial effects were tested using checkerboard and static time-kill assays. Electron microscopy (EM) and untargeted metabolomics were used to ascertain the mechanism(s) of the antibacterial synergy. The combination of polymyxin B and the phenothiazines showed synergistic antibacterial activity in checkerboard and static time-kill assays at clinically relevant concentrations against both polymyxin-susceptible and polymyxin-resistant isolates. EM revealed that the polymyxin B-prochlorperazine combination resulted in greater damage to the bacterial cell compared to each drug monotherapy. In metabolomics, at 0.5 h, polymyxin B monotherapy and the combination (to a greatest extent) disorganised the bacterial cell envelope as manifested by a major perturbation in bacterial membrane lipids (glycerophospholipids and fatty acids), peptidoglycan and lipopolysaccharide (LPS) biosynthesis. At the late time exposure (4 h), the aforementioned effects (except LPS biosynthesis) perpetuated mainly with the combination therapy, indicating the disorganising bacterial membrane biogenesis is potentially behind the mechanisms of antibacterial synergy. In conclusion, the study highlights the potential usefulness of the combination of polymyxin B with phenothiazines for the treatment of polymyxin-resistant Gram-negative infections (e.g. CNS infections).
Collapse
Affiliation(s)
- Maytham Hussein
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Xiaohan Hu
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Olivia K.A. Paulin
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Simon Crawford
- The Monash Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Victoria 3800, Australia
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907-2091, USA
| | - Mark Baker
- Discipline of Biological Sciences, Priority Research Centre in Reproductive Biology, Faculty of Science and IT, University of Newcastle, University Drive, Callaghan NSW, 2308, Australia
| | - Elena K. Schneider-Futschik
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yan Zhu
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
14
|
Xu C, Chen K, Chan KF, Chan EWC, Guo X, Chow HY, Zhao G, Zeng P, Wang M, Zhu Y, Li X, Wong K, Chen S. Imidazole Type Antifungal Drugs Are Effective Colistin Adjuvants That Resensitize Colistin‐Resistant
Enterobacteriaceae. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000084] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Chen Xu
- State Key Lab of Chemical Biology and Drug Discovery Department of Applied Biology and Chemical Technology The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
- Department of Infectious Diseases and Public Health Jockey Club College of Veterinary Medicine and Life Sciences City University of Hong Kong Kowloon 999077 Hong Kong
| | - Kaichao Chen
- Department of Infectious Diseases and Public Health Jockey Club College of Veterinary Medicine and Life Sciences City University of Hong Kong Kowloon 999077 Hong Kong
| | - Kin Fai Chan
- State Key Lab of Chemical Biology and Drug Discovery Department of Applied Biology and Chemical Technology The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
| | - Edward Wai Chi Chan
- State Key Lab of Chemical Biology and Drug Discovery Department of Applied Biology and Chemical Technology The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
| | - Xuyun Guo
- Department of Applied Physics The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
| | - Hoi Yee Chow
- Department of Chemistry The University of Hong Kong Pokfulam Hong Kong
| | - Guangming Zhao
- Department of Applied Physics The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
| | - Ping Zeng
- State Key Lab of Chemical Biology and Drug Discovery Department of Applied Biology and Chemical Technology The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
| | - Miaomiao Wang
- State Key Lab of Chemical Biology and Drug Discovery Department of Applied Biology and Chemical Technology The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
- Department of Infectious Diseases and Public Health Jockey Club College of Veterinary Medicine and Life Sciences City University of Hong Kong Kowloon 999077 Hong Kong
| | - Ye Zhu
- Department of Applied Physics The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
| | - Xuechen Li
- Department of Chemistry The University of Hong Kong Pokfulam Hong Kong
| | - Kwok‐Yin Wong
- State Key Lab of Chemical Biology and Drug Discovery Department of Applied Biology and Chemical Technology The Hong Kong Polytechnic University Hung Hom Kowloon Hong Kong
| | - Sheng Chen
- Department of Infectious Diseases and Public Health Jockey Club College of Veterinary Medicine and Life Sciences City University of Hong Kong Kowloon 999077 Hong Kong
| |
Collapse
|
15
|
Abstract
Despite efforts to develop new antibiotics, antibacterial resistance still develops too fast for drug discovery to keep pace. Often, resistance against a new drug develops even before it reaches the market. This continued resistance crisis has demonstrated that resistance to antibiotics with single protein targets develops too rapidly to be sustainable. Most successful long-established antibiotics target more than one molecule or possess targets, which are encoded by multiple genes. This realization has motivated a change in antibiotic development toward drug candidates with multiple targets. Some mechanisms of action presuppose multiple targets or at least multiple effects, such as targeting the cytoplasmic membrane or the carrier molecule bactoprenol phosphate and are therefore particularly promising. Moreover, combination therapy approaches are being developed to break antibiotic resistance or to sensitize bacteria to antibiotic action. In this Review, we provide an overview of antibacterial multitarget approaches and the mechanisms behind them.
Collapse
Affiliation(s)
- Declan Alan Gray
- Newcastle University
Biosciences Institute, Newcastle University, NE2 4HH Newcastle
upon Tyne, United Kingdom
| | - Michaela Wenzel
- Division of Chemical
Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| |
Collapse
|
16
|
Lee MW, de Anda J, Kroll C, Bieniossek C, Bradley K, Amrein KE, Wong GCL. How do cyclic antibiotics with activity against Gram-negative bacteria permeate membranes? A machine learning informed experimental study. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183302. [PMID: 32311341 DOI: 10.1016/j.bbamem.2020.183302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/30/2022]
Abstract
All antibiotics have to engage bacterial amphiphilic barriers such as the lipopolysaccharide-rich outer membrane or the phospholipid-based inner membrane in some manner, either by disrupting them outright and/or permeating them and thereby allow the antibiotic to get into bacteria. There is a growing class of cyclic antibiotics, many of which are of bacterial origin, that exhibit activity against Gram-negative bacteria, which constitute an urgent problem in human health. We examine a diverse collection of these cyclic antibiotics, both natural and synthetic, which include bactenecin, polymyxin B, octapeptin, capreomycin, and Kirshenbaum peptoids, in order to identify what they have in common when they interact with bacterial lipid membranes. We find that they virtually all have the ability to induce negative Gaussian curvature (NGC) in bacterial membranes, the type of curvature geometrically required for permeation mechanisms such as pore formation, blebbing, and budding. This is interesting since permeation of membranes is a function usually ascribed to antimicrobial peptides (AMPs) from innate immunity. As prototypical test cases of cyclic antibiotics, we analyzed amino acid sequences of bactenecin, polymyxin B, and capreomycin using our recently developed machine-learning classifier trained on α-helical AMP sequences. Although the original classifier was not trained on cyclic antibiotics, a modified classifier approach correctly predicted that bactenecin and polymyxin B have the ability to induce NGC in membranes, while capreomycin does not. Moreover, the classifier was able to recapitulate empirical structure-activity relationships from alanine scans in polymyxin B surprisingly well. These results suggest that there exists some common ground in the sequence design of hybrid cyclic antibiotics and linear AMPs.
Collapse
Affiliation(s)
- Michelle W Lee
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Jaime de Anda
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Carsten Kroll
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Christoph Bieniossek
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Kenneth Bradley
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Kurt E Amrein
- Roche Pharma Research and Early Development Pharmaceutical Science, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Gerard C L Wong
- Department of Bioengineering, Department of Chemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
17
|
Tyrrell JM, Aboklaish AF, Walsh TR, Vaara T, Vaara M. The polymyxin derivative NAB739 is synergistic with several antibiotics against polymyxin-resistant strains of Escherichia coli, Klebsiella pneumoniae and Acinetobacter baumannii. Peptides 2019; 112:149-153. [PMID: 30586602 DOI: 10.1016/j.peptides.2018.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022]
Abstract
The antibiotic crisis has reinstated polymyxins, once abandoned because of their toxicity. Now, preclinical studies have revealed better tolerated and more effective derivatives of polymyxins such as NAB739. Simultaneously, polymyxin-resistant (PMR) strains such as the mcr-1 strains have received lots of justified publicity, even though they are still very rare. Here we show that NAB739 sensitizes the PMR strains to rifampin, a classic "anti-Gram-positive" antibiotic excluded by the intact outer membrane (OM) permeability barrier, as well as to retapamulin, the surrogate of lefamulin, an antibiotic under development against Gram-positive bacteria. Polymyxin B was used as a comparator. The combination of NAB739 and rifampin was synergistic against ten out of eleven PMR strains of Escherichia coli (Fractional Synergy Indices, FICs, 0.14-0.19) and that of NAB739 and retapamulin against all the tested eleven strains (FICs 0.19-0.25). Against PMR Klebsiella pneumoniae (n = 7), the FICs were 0.13-0.27 for NAB739 + rifampin and 0.14-0.28 for NAB739+retapamulin. Against Acinetobacter baumannii (n = 2), the combination of NAB739 and rifampin had the FIC of 0.09-0.19. Furthermore, NAB739 and meropenem were synergistic (FICs 0.25-0.50) against four out of five PMR strains that were simultaneously resistant to meropenem.
Collapse
Affiliation(s)
- Jonathan M Tyrrell
- Department of Medical Microbiology and Infectious Disease, Cardiff University Medical School, Cardiff, Wales, United Kingdom
| | - Ali F Aboklaish
- Department of Medical Microbiology and Infectious Disease, Cardiff University Medical School, Cardiff, Wales, United Kingdom
| | - Timothy R Walsh
- Department of Medical Microbiology and Infectious Disease, Cardiff University Medical School, Cardiff, Wales, United Kingdom
| | - Timo Vaara
- Northern Antibiotics Ltd, FI-02150, Espoo, Finland
| | - Martti Vaara
- Northern Antibiotics Ltd, FI-02150, Espoo, Finland; Department of Bacteriology and Immunology, Helsinki University Medical School, FI-00014, Helsinki, Finland.
| |
Collapse
|
18
|
Polymyxin Derivatives that Sensitize Gram-Negative Bacteria to Other Antibiotics. Molecules 2019; 24:molecules24020249. [PMID: 30641878 PMCID: PMC6359160 DOI: 10.3390/molecules24020249] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/27/2018] [Accepted: 01/07/2019] [Indexed: 01/11/2023] Open
Abstract
Polymyxins (polymyxin B (PMB) and polymyxin E (colistin)) are cyclic lipodecapeptide antibiotics, highly basic due to five free amino groups, and rapidly bactericidal against Gram-negative bacteria, such as the majority of Enterobacteriaceae as well as Acinetobacter baumannii and Pseudomonas aeruginosa. Their clinical use was abandoned in the 1960s because of nephrotoxicity and because better-tolerated drugs belonging to other antibiotic classes were introduced. Now, due to the global dissemination of extremely-drug resistant Gram-negative bacterial strains, polymyxins have resurged as the last-line drugs against those strains. Novel derivatives that are less toxic and/or more effective at tolerable doses are currently under preclinical development and their properties have recently been described in several extensive reviews. Other derivatives lack any direct bactericidal activity but damage the outermost permeability barrier, the outer membrane, of the target bacteria and make it more permeable to many other antibiotics. This review describes the properties of three thus far best-characterized “permeabilizer” derivatives, i.e., the classic permeabilizer polymyxin B nonapeptide (PMBN), NAB7061, and SPR741/NAB741, a compound that recently successfully passed the clinical phase 1. Also, a few other permeabilizer compounds are brought up.
Collapse
|
19
|
Velkov T, Roberts KD. Discovery of Novel Polymyxin-Like Antibiotics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1145:343-362. [PMID: 31364086 DOI: 10.1007/978-3-030-16373-0_20] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The antimicrobial lipopeptides polymyxin B and colistin (polymyxin E) are used as a 'last-line' therapy for infections caused by multidrug-resistant (MDR) Gram-negative pathogens. However, their effective use as antibiotic drugs in the clinical setting is still plagued by significant toxicity issues, in particular their potential for nephrotoxicity. Furthermore, resistance to the polymyxins has begun to emerge in the clinic, which implies a total lack of antibiotics for the treatment of life-threatening infections caused by the Gram-negative 'superbugs'. This chapter details our current understanding of polymyxin structure-activity relationships as well as recent pre-clinical and clinical drug development efforts aimed at generating new polymyxin antibiotics with improved safety and efficacy.
Collapse
Affiliation(s)
- Tony Velkov
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, Australia.
| | - Kade D Roberts
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
20
|
Spectrum of antibacterial activity and mode of action of a novel tris-stilbene bacteriostatic compound. Sci Rep 2018; 8:6912. [PMID: 29720673 PMCID: PMC5932035 DOI: 10.1038/s41598-018-25080-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/29/2018] [Indexed: 11/22/2022] Open
Abstract
The spectrum of activity and mode of action of a novel antibacterial agent, 135C, was investigated using a range of microbiological and genomic approaches. Compound 135C was active against Gram-positive bacteria with MICs for Staphylococcus aureus ranging from 0.12–0.5 μg/ml. It was largely inactive against Gram-negative bacteria. The compound showed bacteriostatic activity in time-kill studies and did not elicit bacterial cell leakage or cell lysis. Checkerboard assays showed no synergy or antagonism when 135C was combined with a range of other antibacterials. Multi-step serial passage of four S. aureus isolates with increasing concentrations of 135C showed that resistance developed rapidly and was stable after drug-free passages. Minor differences in the fitness of 135C-resistant strains and parent wildtypes were evident by growth curves, but 135C-resistant strains did not show cross-resistance to other antibacterial agents. Genomic comparison of resistant and wildtype parent strains showed changes in genes encoding cell wall teichoic acids. 135C shows promising activity against Gram-positive bacteria but is currently limited by the rapid resistance development. Further studies are required to investigate the effects on cell wall teichoic acids and to determine whether the issue of resistance development can be overcome.
Collapse
|
21
|
Gravel J, Paradis-Bleau C, Schmitzer AR. Adaptation of a bacterial membrane permeabilization assay for quantitative evaluation of benzalkonium chloride as a membrane-disrupting agent. MEDCHEMCOMM 2017; 8:1408-1413. [PMID: 30108851 PMCID: PMC6072009 DOI: 10.1039/c7md00113d] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 04/28/2017] [Indexed: 01/31/2023]
Abstract
We describe the use of the ortho-nitrophenyl-β-galactoside (ONPG) assay developed by Lehrer et al. to which a new mathematical data treatment was applied. In this simplified assay, only one enzymatic assay is needed to provide direct evidence of the kinetics of Escherichia coli membrane permeabilization induced by different concentrations of benzalkonium chloride (BAC). Analysis of the data obtained from the revised ONPG assay with our adapted mathematical formula indicates that BAC induces permeabilization of the bacterial outer and inner membranes in a two-step process. The two effective concentration (EC50) values obtained in this study, combined with the results from an outer membrane permeabilization assay, suggest that the two steps observed in the permeabilization process are related to the two different bacterial membranes. We show that membrane permeabilization occurs very fast upon the addition of bacterial cells to the BAC solutions and demonstrate that sub-lethal concentrations of BAC disturb the integrity of the Gram-negative bacterial membranes. Overall, our work broadens our knowledge on the mode of action of BAC on bacterial cells and emphasizes that BAC, and quaternary ammonium compounds in general, should not be used at sub-lethal concentrations in order to lower the risk of bacterial tolerance and resistance to antibiotics.
Collapse
Affiliation(s)
- Julien Gravel
- Département de Chimie , Université de Montréal , CP 6128, Succ. Centre Ville , Montréal (Québec) , Canada .
| | - Catherine Paradis-Bleau
- Département de Microbiologie, Infectiologie et Immunologie , Université de Montréal , CP 6128, Succ. Centre-ville , Montréal (Québec) , Canada .
| | - Andreea R Schmitzer
- Département de Chimie , Université de Montréal , CP 6128, Succ. Centre Ville , Montréal (Québec) , Canada .
| |
Collapse
|
22
|
Hussein MH, Schneider EK, Elliott AG, Han M, Reyes-Ortega F, Morris F, Blaskovich MAT, Jasim R, Currie B, Mayo M, Baker M, Cooper MA, Li J, Velkov T. From Breast Cancer to Antimicrobial: Combating Extremely Resistant Gram-Negative “Superbugs” Using Novel Combinations of Polymyxin B with Selective Estrogen Receptor Modulators. Microb Drug Resist 2017; 23:640-650. [DOI: 10.1089/mdr.2016.0196] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Maytham H. Hussein
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Elena K. Schneider
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Alysha G. Elliott
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Meiling Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Felisa Reyes-Ortega
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Faye Morris
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Mark A. T. Blaskovich
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Raad Jasim
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Bart Currie
- Menzies School of Health Research, Casuarina, Australia
| | - Mark Mayo
- Menzies School of Health Research, Casuarina, Australia
| | - Mark Baker
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, Australia
| | - Matthew A. Cooper
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jian Li
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Parkville, Australia
| | - Tony Velkov
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| |
Collapse
|
23
|
Antibiotic-non-antibiotic combinations for combating extremely drug-resistant Gram-negative 'superbugs'. Essays Biochem 2017; 61:115-125. [PMID: 28258235 DOI: 10.1042/ebc20160058] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/02/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
Abstract
The emergence of antimicrobial resistance of Gram-negative pathogens has become a worldwide crisis. The status quo for combating resistance is to employ synergistic combinations of antibiotics. Faced with this fast-approaching post-antibiotic era, it is critical that we devise strategies to prolong and maximize the clinical efficacy of existing antibiotics. Unfortunately, reports of extremely drug-resistant (XDR) Gram-negative pathogens have become more common. Combining antibiotics such as polymyxin B or the broad-spectrum tetracycline and minocycline with various FDA-approved non-antibiotic drugs have emerged as a novel combination strategy against otherwise untreatable XDR pathogens. This review surveys the available literature on the potential benefits of employing antibiotic-non-antibiotic drug combination therapy. The apex of this review highlights the clinical utility of this novel therapeutic strategy for combating infections caused by 'superbugs'.
Collapse
|
24
|
Intrinsic, adaptive and acquired antimicrobial resistance in Gram-negative bacteria. Essays Biochem 2017; 61:49-59. [DOI: 10.1042/ebc20160063] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 12/31/2022]
Abstract
Gram-negative bacteria are responsible for a large proportion of antimicrobial-resistant infections in humans and animals. Among this class of bacteria are also some of the most successful environmental organisms. Part of this success is their adaptability to a variety of different niches, their intrinsic resistance to antimicrobial drugs and their ability to rapidly acquire resistance mechanisms. These mechanisms of resistance are not exclusive and the interplay of several mechanisms causes high levels of resistance. In this review, we explore the molecular mechanisms underlying resistance in Gram-negative organisms and how these different mechanisms enable them to survive many different stress conditions.
Collapse
|
25
|
Silver LL. A Gestalt approach to Gram-negative entry. Bioorg Med Chem 2016; 24:6379-6389. [DOI: 10.1016/j.bmc.2016.06.044] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/12/2016] [Accepted: 06/22/2016] [Indexed: 01/01/2023]
|
26
|
Trimble MJ, Mlynárčik P, Kolář M, Hancock REW. Polymyxin: Alternative Mechanisms of Action and Resistance. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a025288. [PMID: 27503996 DOI: 10.1101/cshperspect.a025288] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Antibiotic resistance among pathogenic bacteria is an ever-increasing issue worldwide. Unfortunately, very little has been achieved in the pharmaceutical industry to combat this problem. This has led researchers and the medical field to revisit past drugs that were deemed too toxic for clinical use. In particular, the cyclic cationic peptides polymyxin B and colistin, which are specific for Gram-negative bacteria, have been used as "last resort" antimicrobials. Before the 1980s, these drugs were known for their renal and neural toxicities; however, new clinical practices and possibly improved manufacturing have made them safer to use. Previously suggested to primarily attack the membranes of Gram-negative bacteria and to not easily select for resistant mutants, recent research exploring resistance and mechanisms of action has provided new perspectives. This review focuses primarily on the proposed alternative mechanisms of action, known resistance mechanisms, and how these support the alternative mechanisms of action.
Collapse
Affiliation(s)
- Michael J Trimble
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Patrik Mlynárčik
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University, 771 47 Olomouc, Czech Republic
| | - Milan Kolář
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University, 771 47 Olomouc, Czech Republic
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
27
|
Shavrina M, Zimin A, Molochkov N, Chernyshov S, Machulin A, Mikoulinskaia G. In vitro
study of the antibacterial effect of the bacteriophage T5 thermostable endolysin on Escherichia coli
cells. J Appl Microbiol 2016; 121:1282-1290. [DOI: 10.1111/jam.13251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/26/2016] [Accepted: 08/03/2016] [Indexed: 01/07/2023]
Affiliation(s)
- M.S. Shavrina
- Branch of Shemyakin & Ovchinnikov's Institute of Bioorganic Chemistry RAS; Pushchino Moscow Russia
| | - A.A. Zimin
- Skryabin's Institute of Biochemistry and Physiology of Micro-organisms RAS; Pushchino Moscow Russia
| | - N.V. Molochkov
- Institute of Theoretical and Experimental Biophysics RAS; Pushchino Moscow Russia
| | - S.V. Chernyshov
- Branch of Shemyakin & Ovchinnikov's Institute of Bioorganic Chemistry RAS; Pushchino Moscow Russia
| | - A.V. Machulin
- Skryabin's Institute of Biochemistry and Physiology of Micro-organisms RAS; Pushchino Moscow Russia
| | - G.V. Mikoulinskaia
- Branch of Shemyakin & Ovchinnikov's Institute of Bioorganic Chemistry RAS; Pushchino Moscow Russia
| |
Collapse
|
28
|
Polyamine derivatives: a revival of an old neglected scaffold to fight resistant Gram-negative bacteria? Future Med Chem 2016; 8:963-73. [DOI: 10.4155/fmc-2016-0011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Emergence of multidrug-resistant pathogens was responsible for microbial infections and inefficacy of numerous antimicrobial therapies has induced a need for the research of new classes of antibiotics. In this review, we will focus our interest toward the biological properties of polyamino antimicrobial agents.
Collapse
|
29
|
Arunachalam K, Ascêncio SD, Soares IM, Souza Aguiar RW, da Silva LI, de Oliveira RG, Balogun SO, de Oliveira Martins DT. Gallesia integrifolia (Spreng.) Harms: In vitro and in vivo antibacterial activities and mode of action. JOURNAL OF ETHNOPHARMACOLOGY 2016; 184:128-137. [PMID: 26945980 DOI: 10.1016/j.jep.2016.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 02/16/2016] [Accepted: 03/01/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gallesia integrifolia (Phytolaccaceae) is commonly known as "pau-d'alho" in Brazil or "garlic plant" due to the strong scent of garlic peculiar to all parts of the plant. The bark decoction is used for the treatment of microbial infections among other diseases by different ethnic groups in Brazil, Peruvian Amazonians, Bolivia and Mosetene Indians. This study aimed to advance in the antibacterial activity and characterize the mode of action of the hydroethanolic extract of the inner stem bark of G. integrifolia (HEGi) using in vivo and in vitro experimental models. MATERIALS AND METHODS The qualitative and quantitative phytochemical analyzes of HEGi were carried out using colorimetric and HPLC technique. The cytotoxic potential of HEGi was evaluated against CHO-K1 cells by Alamar blue assay and its acute toxicity was assessed by the Hippocratic screening test using Swiss-Webster mice. The antibacterial activity was evaluated by micro- dilution method against ten strains of Gram-positive and Gram-negative bacteria. The mode of action of HEGi was investigated by outer membrane permeability, nucleotide leakage and potassium efflux assays. In vivo infection model was established by using Staphylococcus aureus infection model Wistar rats. RESULTS Qualitative phytochemical analysis of HEGi revealed the presence of saponins, alkaloids, phenolic compounds and flavonoids. Phytochemical quantification of HEGi showed that higher total phenolic (80.10±0.62mg GAE/g) and flavonoid (16.10±0.03mg RE/g) contents. HPLC fingerprint analysis revealed the presence of gallic acid, rutin, and morin. In the Alamar blue assay no cytotoxic effect of HEGi in CHO-K1 cells was observed up to 200µg/mL, and no signs or symptoms of acute toxicity were observed in mice of both sexes at higher doses of up to 2000mg/kg, p.o. HEGi demonstrated bacteriostatic effect against selected Gram positive and Gram negative bacterial pathogens. Its mode of action is associated, at least partly, with changes in the permeability of bacterial membranes, evidenced by the increased entry of hydrophobic antibiotic in Pseudomonas aeruginosa, intense K(+) efflux and nucleotides leakage in Shigella flexneri, Streptococcus pyogenes and S. aureus. HEGi attenuated the experimental blood borne S. aureus infection in rats at all the tested doses levels (10, 50 and 250mg/kg). CONCLUSION HEGi is safe at the dose tested when used acutely, and it presented broad antibacterial effect, which support its traditional use in the treatment of bacterial infections. It contains well known important phytochemicals, recognized to be active against bacterial pathogens in vitro and might be collectively responsible for the antibacterial activity of HEGi. It is bacteriostatic in nature, with membrane perturbation being one of it mode of action. HEGi represent a potential phytotherapic antibacterial agent.
Collapse
Affiliation(s)
- Karuppusamy Arunachalam
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso (UFMT), Av. Fernando Correa da Costa, no. 2367, Coxipó, Cuiabá, Mato Grosso 78060-900, Brazil
| | - Sérgio Donizeti Ascêncio
- Research Laboratory of Natural Products, Federal University of Tocantins, Faculty of Medicine, Palmas, Tocantins 77020-210, Brazil
| | - Ilsamar Mendes Soares
- Research Laboratory of Natural Products, Federal University of Tocantins, Faculty of Medicine, Palmas, Tocantins 77020-210, Brazil
| | | | - Larissa Irene da Silva
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso (UFMT), Av. Fernando Correa da Costa, no. 2367, Coxipó, Cuiabá, Mato Grosso 78060-900, Brazil
| | - Ruberlei Godinho de Oliveira
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso (UFMT), Av. Fernando Correa da Costa, no. 2367, Coxipó, Cuiabá, Mato Grosso 78060-900, Brazil
| | - Sikiru Olaitan Balogun
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso (UFMT), Av. Fernando Correa da Costa, no. 2367, Coxipó, Cuiabá, Mato Grosso 78060-900, Brazil
| | - Domingos Tabajara de Oliveira Martins
- Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso (UFMT), Av. Fernando Correa da Costa, no. 2367, Coxipó, Cuiabá, Mato Grosso 78060-900, Brazil.
| |
Collapse
|
30
|
O'Dowd H, Erwin AL, Lewis JG. Lincosamide Antibacterials. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1002/9783527676545.ch07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
31
|
Miki T, Hardt WD. Outer membrane permeabilization is an essential step in the killing of gram-negative bacteria by the lectin RegIIIβ. PLoS One 2013; 8:e69901. [PMID: 23922847 PMCID: PMC3726741 DOI: 10.1371/journal.pone.0069901] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 06/17/2013] [Indexed: 02/06/2023] Open
Abstract
The C-type lectin RegIIIβ can kill certain Gram-positive and Gram-negative bacteria. The susceptibility of S. Typhimurium depends on the bacterial growth phase, i.e., bacteria from the logarithmic growth phase do bind RegIIIβ and are subsequently killed. Lipid A is one of the bacterial targets for RegIIIβ. However, at the molecular level, it is not understood how RegIIIβ interacts with and kills Gram-negative bacteria. Here, we show that RegIIIβ interacts with Gram-negative bacteria in two distinct steps. Initially, it binds to surface-exposed lipid A. The lipid A can be shielded by the O-antigen of lipopolysaccharide (LPS), as indicated by the exquisite susceptibility of wbaP mutants to RegIIIβ-mediated killing. Increased cell viability after incubation with an anti-lipid A antibody also supports this conclusion. This RegIIIβ-binding permeabilizes the outer membrane to hydrophobic dyes like Ethidium bromide or to bulky bacteriolytic enzymes like lysozyme. Conversely, compromising the outer membrane integrity by the mild detergent Triton X-100 enhances the antibacterial effect of RegIIIβ. Based on our observations, we conclude that RegIIIβ interacts with Gram-negative bacteria in two subsequent steps. Initially, it binds to the outer membrane thus leading to outer membrane permeabilization. This initial step is necessary for RegIIIβ to reach a second, still not well understood target site (presumably localized in the periplasm or the cytoplasmic membrane), thereby triggering bacterial death. This provides novel insights into the outer membrane-step of the bactericidal mechanism of RegIIIβ.
Collapse
Affiliation(s)
- Tsuyoshi Miki
- The Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | | |
Collapse
|
32
|
Pomares MF, Corbalán NS, Adler C, de Cristóbal R, Farías RN, Delgado MA, Vincent PA. Macrophage environment turns otherwise MccJ25-resistant Salmonella into sensitive. BMC Microbiol 2013; 13:95. [PMID: 23634875 PMCID: PMC3652737 DOI: 10.1186/1471-2180-13-95] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/22/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Microcin J25 (MccJ25) is a plasmid-encoded antibiotic peptide produced by Escherichia coli (E. coli). MccJ25 enters into the sensitive E. coli strains by the outer membrane receptor FhuA and the inner membrane proteins TonB, ExbB, ExbD and SbmA. The resistance of Salmonella enterica serovar Typhimurium (S. Typhimurium) to MccJ25 is attributed to the inability of its FhuA protein to incorporate the antibiotic into the cell. RESULTS In this work we demonstrate that S. Typhimurium becomes notably susceptible to MccJ25 when replicating within macrophages. In order to determine the possible cause of this phenomenon, we studied the sensitivity of S. Typhimurium to MccJ25 at conditions resembling those of the internal macrophage environment, such as low pH, low magnesium and iron deprivation. We observed that the strain was only sensitive to the antibiotic at low pH, leading us to attribute the bacterial sensitization to this condition. A MccJ25-resistant E. coli strain in which fhuA is deleted was also inhibited by the antibiotic at low pH. Then, we could assume that the MccJ25 sensitivity change observed in both E. coli fhuA and S. Typhimurium is mediated by a MccJ25 uptake independent of the FhuA receptor. Moreover, low pH incubation also sensitized S. Typhimurium to the hydrophobic antibiotic novobiocin, which does not affect enteric bacteria viability because it is unable to penetrate the bacterial outer membrane. This observation supports our hypothesis about low pH producing a modification in the bacterial membrane permeability that allows an unspecific MccJ25 uptake. On the other hand, MccJ25 inhibited S. Typhimurium when cells were preincubated in acidic pH medium and then treated at neutral pH with the antibiotic. CONCLUSIONS Our results suggest that acidic condition does not alter MccJ25 hydrophobicity but irreversibly modifies bacterial membrane permeability. This would allow an unspecific antibiotic uptake into the cell.From our data it is possible to infer that intracellular pathogenic strains, which are in vitro resistant to MccJ25, could become susceptible ones in vivo. Therefore, the MccJ25 action spectrum would be broader than what in vitro experiments indicate.
Collapse
Affiliation(s)
- María Fernanda Pomares
- Instituto Superior de Investigaciones Biológicas (Concejo Nacional de Investigación Científica y Técnica-Universidad Nacional de Tucumán) and Instituto de Química Biológica Dr, Bernabe Bloj, Chacabuco 461, San Miguel de Tucumán 4000 Tucumán, Argentina
| | | | | | | | | | | | | |
Collapse
|
33
|
Martínez de Tejada G, Sánchez-Gómez S, Rázquin-Olazaran I, Kowalski I, Kaconis Y, Heinbockel L, Andrä J, Schürholz T, Hornef M, Dupont A, Garidel P, Lohner K, Gutsmann T, David SA, Brandenburg K. Bacterial cell wall compounds as promising targets of antimicrobial agents I. Antimicrobial peptides and lipopolyamines. Curr Drug Targets 2012; 13:1121-30. [PMID: 22664072 DOI: 10.2174/138945012802002410] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 09/30/2011] [Accepted: 05/18/2012] [Indexed: 11/22/2022]
Abstract
The first barrier that an antimicrobial agent must overcome when interacting with its target is the microbial cell wall. In the case of Gram-negative bacteria, additional to the cytoplasmic membrane and the peptidoglycan layer, an outer membrane (OM) is the outermost barrier. The OM has an asymmetric distribution of the lipids with phospholipids and lipopolysaccharide (LPS) located in the inner and outer leaflets, respectively. In contrast, Gram-positive bacteria lack OM and possess a much thicker peptidoglycan layer compared to their Gram-negative counterparts. An additional class of amphiphiles exists in Gram-positives, the lipoteichoic acids (LTA), which may represent important structural components. These long molecules cross-bridge the entire cell envelope with their lipid component inserting into the outer leaflet of the cytoplasmic membrane and the teichoic acid portion penetrating into the peptidoglycan layer. Furthermore, both classes of bacteria have other important amphiphiles, such as lipoproteins, whose importance has become evident only recently. It is not known yet whether any of these amphiphilic components are able to stimulate the immune system under physiological conditions as constituents of intact bacteria. However, all of them have a very high pro-inflammatory activity when released from the cell. Such a release may take place through the interaction with the immune system, or with antibiotics (particularly with those targeting cell wall components), or simply by the bacterial division. Therefore, a given antimicrobial agent must ideally have a double character, namely, it must overcome the bacterial cell wall barrier, without inducing the liberation of the pro-inflammatory amphiphiles. Here, new data are presented which describe the development and use of membrane-active antimicrobial agents, in particular antimicrobial peptides (AMPs) and lipopolyamines. In this way, essential progress was achieved, in particular with respect to the inhibition of deleterious consequences of bacterial infections such as severe sepsis and septic shock.
Collapse
|
34
|
Shukla NM, Salunke DB, Yoo E, Mutz CA, Balakrishna R, David SA. Antibacterial activities of Groebke-Blackburn-Bienaymé-derived imidazo[1,2-a]pyridin-3-amines. Bioorg Med Chem 2012; 20:5850-63. [PMID: 22925449 PMCID: PMC3448811 DOI: 10.1016/j.bmc.2012.07.052] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/26/2012] [Accepted: 07/31/2012] [Indexed: 10/28/2022]
Abstract
We sought to explore the imidazo[1,2-a]pyridin-3-amines for TLR7 (or 8)-modulatory activities. This chemotype, readily accessed via the Groebke-Blackburn-Bienaymé multi-component reaction, resulted in compounds that were TLR7/8-inactive, but exhibited bacteriostatic activity against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA). To investigate the mechanism of antibacterial activity of this new chemotype, a resistant strain of S. aureus was generated by serially passaging the organism in escalating doses of the most active analogue. A comparison of minimum inhibitory concentrations (MICs) of known bacteriostatic agents in wild-type and resistant strains indicates a novel mechanism of action. Structure-activity relationship studies have led to the identification of positions on the scaffold for additional structural modifications that should allow for the introduction of probes designed to examine cognate binding partners and molecular targets, while not significantly compromising antibacterial potency.
Collapse
Affiliation(s)
| | | | - Euna Yoo
- Department of Medicinal Chemistry, University of Kansas
| | - Cole A. Mutz
- Department of Medicinal Chemistry, University of Kansas
| | | | | |
Collapse
|
35
|
Iino R, Nishino K, Noji H, Yamaguchi A, Matsumoto Y. A microfluidic device for simple and rapid evaluation of multidrug efflux pump inhibitors. Front Microbiol 2012; 3:40. [PMID: 22347225 PMCID: PMC3274760 DOI: 10.3389/fmicb.2012.00040] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 01/26/2012] [Indexed: 12/03/2022] Open
Abstract
Recently, multidrug-resistant pathogens have disseminated widely owing essentially to their increased multidrug efflux pump activity. Presently, there is a scarcity of new antibacterial agents, and hence, inhibitors of multidrug efflux pumps belonging to the resistance–nodulation–cell division (RND) family appear useful in the treatment of infections by multidrug-resistant pathogens. Moreover, recent progress in microfabrication technologies has expanded the application of nano/micro-devices to the field of human healthcare, such as the detection of infections and diagnosis of diseases. We developed a microfluidic channel device for a simple and rapid evaluation of bacterial drug efflux activity. By combining the microfluidic device with a fluorogenic compound, fluorescein-di-β-D-galactopyranoside, which is hydrolyzed to a fluorescent dye in the cytoplasm of Escherichia coli, we successfully evaluated the effects of inhibitors on the RND-type multidrug efflux pumps MexAB–OprM and MexXY–OprM from Pseudomonas aeruginosa in E. coli. Our new method successfully detected the MexB-specific inhibitory effect of D13-9001 and revealed an unexpected membrane-permeabilizing effect of Phe-Arg-β-naphthylamide, which has long been used as an efflux pump inhibitor.
Collapse
Affiliation(s)
- Ryota Iino
- Department of Applied Chemistry, Graduate School of Engineering, University of Tokyo Bunkyo-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
36
|
Kaconis Y, Kowalski I, Howe J, Brauser A, Richter W, Razquin-Olazarán I, Iñigo-Pestaña M, Garidel P, Rössle M, Martinez de Tejada G, Gutsmann T, Brandenburg K. Biophysical mechanisms of endotoxin neutralization by cationic amphiphilic peptides. Biophys J 2011; 100:2652-61. [PMID: 21641310 DOI: 10.1016/j.bpj.2011.04.041] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 04/05/2011] [Accepted: 04/06/2011] [Indexed: 11/25/2022] Open
Abstract
Bacterial endotoxins (lipopolysaccharides (LPS)) are strong elicitors of the human immune system by interacting with serum and membrane proteins such as lipopolysaccharide-binding protein (LBP) and CD14 with high specificity. At LPS concentrations as low as 0.3 ng/ml, such interactions may lead to severe pathophysiological effects, including sepsis and septic shock. One approach to inhibit an uncontrolled inflammatory reaction is the use of appropriate polycationic and amphiphilic antimicrobial peptides, here called synthetic anti-LPS peptides (SALPs). We designed various SALP structures and investigated their ability to inhibit LPS-induced cytokine secretion in vitro, their protective effect in a mouse model of sepsis, and their cytotoxicity in physiological human cells. Using a variety of biophysical techniques, we investigated selected SALPs with considerable differences in their biological responses to characterize and understand the mechanism of LPS inactivation by SALPs. Our investigations show that neutralization of LPS by peptides is associated with a fluidization of the LPS acyl chains, a strong exothermic Coulomb interaction between the two compounds, and a drastic change of the LPS aggregate type from cubic into multilamellar, with an increase in the aggregate sizes, inhibiting the binding of LBP and other mammalian proteins to the endotoxin. At the same time, peptide binding to phospholipids of human origin (e.g., phosphatidylcholine) does not cause essential structural changes, such as changes in membrane fluidity and bilayer structure. The absence of cytotoxicity is explained by the high specificity of the interaction of the peptides with LPS.
Collapse
Affiliation(s)
- Yani Kaconis
- Forschungszentrum Borstel, Leibniz-Zentrum für Medizin und Biowissenschaften, Borstel, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Peri F, Piazza M. Therapeutic targeting of innate immunity with Toll-like receptor 4 (TLR4) antagonists. Biotechnol Adv 2011; 30:251-60. [PMID: 21664961 DOI: 10.1016/j.biotechadv.2011.05.014] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 04/27/2011] [Accepted: 05/18/2011] [Indexed: 01/04/2023]
Abstract
Early recognition of invading bacteria by the innate immune system has a crucial function in antibacterial defense by triggering inflammatory responses that prevent the spread of infection and suppress bacterial growth. Toll-like receptor 4 (TLR4), the innate immunity receptor of bacterial endotoxins, plays a pivotal role in the induction of inflammatory responses. TLR4 activation by bacterial lipopolysaccharide (LPS) is achieved by the coordinate and sequential action of three other proteins, LBP, CD14 and MD-2 receptors, that bind lipopolysaccharide (LPS) and present it to TLR4 by forming the activated (TLR4-MD-2-LPS)(2) complex. Small molecules active in modulating the TLR4 activation process have great pharmacological interest as vaccine adjuvants, immunotherapeutics or antisepsis and anti-inflammatory agents. In this review we present natural and synthetic molecules active in inhibiting TLR4-mediated LPS signalling in humans and their therapeutic potential. New pharmacological applications of TLR4 antagonists will be also presented related to the recently discovered role of TLR4 in the insurgence and progression of neuropathic pain and sterile inflammations.
Collapse
Affiliation(s)
- Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy.
| | | |
Collapse
|
38
|
Evaluation of multidrug efflux pump inhibitors by a new method using microfluidic channels. PLoS One 2011; 6:e18547. [PMID: 21533264 PMCID: PMC3075257 DOI: 10.1371/journal.pone.0018547] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 03/03/2011] [Indexed: 11/19/2022] Open
Abstract
Fluorescein-di-β-d-galactopyranoside (FDG), a fluorogenic compound, is hydrolyzed by β-galactosidase in the cytoplasm of Escherichia coli to produce a fluorescent dye, fluorescein. We found that both FDG and fluorescein were substrates of efflux pumps, and have developed a new method to evaluate efflux-inhibitory activities in E. coli using FDG and a microfluidic channel device. We used E. coli MG1655 wild-type, ΔacrB (ΔB), ΔtolC (ΔC) and ΔacrBΔtolC (ΔBC) harboring plasmids carrying the mexAB-oprM (pABM) or mexXY-oprM (pXYM) genes of Pseudomonas aeruginosa. Two inhibitors, MexB-specific pyridopyrimidine (D13-9001) and non-specific Phe-Arg-β-naphthylamide (PAβN) were evaluated. The effects of inhibitors on pumps were observed using the microfluidic channel device under a fluorescence microscope. AcrAB-TolC and analogous pumps effectively prevented FDG influx in wild-type cells, resulting in no fluorescence. In contrast, ΔB or ΔC easily imported and hydrolyzed FDG to fluorescein, which was exported by residual pumps in ΔB. Consequently, fluorescent medium in ΔB and fluorescent cells of ΔC and ΔBC were observed in the microfluidic channels. D13-9001 substantially increased fluorescent cell number in ΔBC/pABM but not in ΔBC/pXYM. PAβN increased medium fluorescence in all strains, especially in the pump deletion mutants, and caused fluorescein accumulation to disappear in ΔC. The checkerboard method revealed that D13-9001 acts synergistically with aztreonam, ciprofloxacin, and erythromycin only against the MexAB-OprM producer (ΔBC/pABM), and PAβN acts synergistically, especially with erythromycin, in all strains including the pump deletion mutants. The results obtained from PAβN were similar to the results from membrane permeabilizer, polymyxin B or polymyxin B nonapeptide by concentration. The new method clarified that D13-9001 specifically inhibited MexAB-OprM in contrast to PAβN, which appeared to be a substrate of the pumps and permeabilized the membranes in E. coli.
Collapse
|
39
|
Sato Y, Shindo M, Sakura N, Uchida Y, Kato I. Novel Des-Fatty Acyl-Polymyxin B Derivatives with Pseudomonas aeruginosa-Specific Antimicrobial Activity. Chem Pharm Bull (Tokyo) 2011; 59:597-602. [DOI: 10.1248/cpb.59.597] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuki Sato
- Faculty of Pharmaceutical Sciences, Hokuriku University
| | - Mitsuno Shindo
- Department of Health and Nutrition, Osaka Shoin Women's University
| | | | - Yoshiki Uchida
- Department of Health and Nutrition, Osaka Shoin Women's University
| | - Ikuo Kato
- Faculty of Pharmaceutical Sciences, Hokuriku University
| |
Collapse
|
40
|
Yamada K, Kodaira M, Shinoda SS, Komagoe K, Oku H, Katakai R, Katsu T, Matsuo I. Structure–activity relationships of gramicidin S analogs containing (β-3-pyridyl)-α,β-dehydroalanine residues on membrane permeability. MEDCHEMCOMM 2011. [DOI: 10.1039/c1md00081k] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
Weatherspoon-Griffin N, Zhao G, Kong W, Kong Y, Morigen, Andrews-Polymenis H, McClelland M, Shi Y. The CpxR/CpxA two-component system up-regulates two Tat-dependent peptidoglycan amidases to confer bacterial resistance to antimicrobial peptide. J Biol Chem 2010; 286:5529-39. [PMID: 21149452 DOI: 10.1074/jbc.m110.200352] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We demonstrate that the twin arginine translocation (Tat) system contributes to bacterial resistance to cationic antimicrobial peptides (CAMPs). Our results show that a deletion at the tatC gene, which encodes a subunit of the Tat complex, caused Salmonella and Escherichia coli to become susceptible to protamine. We screened chromosomal loci that encode known and predicted Tat-dependent proteins and found that two N-acetylmuramoyl-l-alanine amidases, encoded by amiA and amiC, elevated bacterial resistance to protamine and α-helical peptides magainin 2 and melittin but not to β-sheet defensin HNP-1 and lipopeptide polymyxin B. Genetic analysis suggests that transcription of both amiA and amiC loci in Salmonella is up-regulated by the CpxR/CpxA two-component system when nlpE is overexpressed. A footprinting analysis reveals that CpxR protein can interact with amiA and amiC promoters at the CpxR box, which is localized between the predicted -10 and -35 regions but present on different strands in these two genes. In addition, our results show that activation of the CpxR/CpxA system can facilitate protamine resistance because nlpE overexpression elevates this resistance in the wild-type strain but not the cpxR deletion mutant. Thus, we uncover a new transcriptional regulation pathway in which the Cpx envelope stress response system modulates the integrity of the cell envelope in part by controlling peptidoglycan amidase activity, which confers bacterial resistance to protamine and α-helical CAMPs. Our studies have important implications for understanding transcriptional regulation of peptidoglycan metabolism and also provide new insights into the role of the bacterial envelope in CAMP resistance.
Collapse
|
42
|
Structural features governing the activity of lactoferricin-derived peptides that act in synergy with antibiotics against Pseudomonas aeruginosa in vitro and in vivo. Antimicrob Agents Chemother 2010; 55:218-28. [PMID: 20956602 DOI: 10.1128/aac.00904-10] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is naturally resistant to many antibiotics, and infections caused by this organism are a serious threat, especially to hospitalized patients. The intrinsic low permeability of P. aeruginosa to antibiotics results from the coordinated action of several mechanisms, such as the presence of restrictive porins and the expression of multidrug efflux pump systems. Our goal was to develop antimicrobial peptides with an improved bacterial membrane-permeabilizing ability, so that they enhance the antibacterial activity of antibiotics. We carried out a structure activity relationship analysis to investigate the parameters that govern the permeabilizing activity of short (8- to 12-amino-acid) lactoferricin-derived peptides. We used a new class of constitutional and sequence-dependent descriptors called PEDES (peptide descriptors from sequence) that allowed us to predict (Spearman's ρ = 0.74; P < 0.001) the permeabilizing activity of a new peptide generation. To study if peptide-mediated permeabilization could neutralize antibiotic resistance mechanisms, the most potent peptides were combined with antibiotics, and the antimicrobial activities of the combinations were determined on P. aeruginosa strains whose mechanisms of resistance to those antibiotics had been previously characterized. A subinhibitory concentration of compound P2-15 or P2-27 sensitized P. aeruginosa to most classes of antibiotics tested and counteracted several mechanisms of antibiotic resistance, including loss of the OprD porin and overexpression of several multidrug efflux pump systems. Using a mouse model of lethal infection, we demonstrated that whereas P2-15 and erythromycin were unable to protect mice when administered separately, concomitant administration of the compounds afforded long-lasting protection to one-third of the animals.
Collapse
|
43
|
Combined antibacterial and anti-inflammatory activity of a cationic disubstituted dexamethasone-spermine conjugate. Antimicrob Agents Chemother 2010; 54:2525-33. [PMID: 20308375 DOI: 10.1128/aac.01682-09] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The rising number of antibiotic-resistant bacterial strains represents an emerging health problem that has motivated efforts to develop new antibacterial agents. Endogenous cationic antibacterial peptides (CAPs) that are produced in tissues exposed to the external environment are one model for the design of novel antibacterial compounds. Here, we report evidence that disubstituted dexamethasone-spermine (D2S), a cationic corticosteroid derivative initially identified as a by-product of synthesis of dexamethasone-spermine (DS) for the purpose of improving cellular gene delivery, functions as an antibacterial peptide-mimicking molecule. This moiety exhibits bacterial killing activity against clinical isolates of Staphylococcus aureus, Pseudomonas aeruginosa present in cystic fibrosis (CF) sputa, and Pseudomonas aeruginosa biofilm. Although compromised in the presence of plasma, D2S antibacterial activity resists the proteolytic activity of pepsin and is maintained in ascites, cerebrospinal fluid, saliva, and bronchoalveolar lavage (BAL) fluid. D2S also enhances S. aureus susceptibility to antibiotics, such as amoxicillin (AMC), tetracycline (T), and amikacin (AN). Inhibition of interleukin-6 (IL-6) and IL-8 release from lipopolysaccharide (LPS)- or lipoteichoic acid (LTA)-treated neutrophils in the presence of D2S suggests that this molecule might also prevent systemic inflammation caused by bacterial wall products. D2S-mediated translocation of green fluorescent protein (GFP)-labeled glucocorticoid receptor (GR) in bovine aorta endothelial cells (BAECs) suggests that some of its anti-inflammatory activities involve engagement of glucocorticoid receptors. The combined antibacterial and anti-inflammatory activities of D2S suggest its potential as an alternative to natural CAPs in the prevention and treatment of some bacterial infections.
Collapse
|
44
|
Urakawa H, Yamada K, Komagoe K, Ando S, Oku H, Katsu T, Matsuo I. Structure-activity relationships of bacterial outer-membrane permeabilizers based on polymyxin B heptapeptides. Bioorg Med Chem Lett 2010; 20:1771-5. [PMID: 20138759 DOI: 10.1016/j.bmcl.2010.01.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Revised: 01/04/2010] [Accepted: 01/06/2010] [Indexed: 01/08/2023]
Abstract
A series of cationic cyclic heptapeptides based on polymyxin B have been synthesized for use as permeabilizers of the outer membrane of Gram-negative bacteria. Only analogs with the Dab(2)-d-Phe(3)-Leu(4)-Xxx(5) sequence (Xxx = Dab or Orn) showed a synergistic bactericidal effect when combined with conventional antibiotics, indicating that the Dab(2) residue plays a critical role in permeation of the outer membrane of Gram-negative bacteria.
Collapse
Affiliation(s)
- Hirotoshi Urakawa
- Department of Chemistry and Chemical Biology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Katsuma N, Sato Y, Ohki K, Okimura K, Ohnishi K, Sakura N. Development of des-fatty acyl-polymyxin B decapeptide analogs with Pseudomonas aeruginosa-specific antimicrobial activity. Chem Pharm Bull (Tokyo) 2009; 57:332-6. [PMID: 19336926 DOI: 10.1248/cpb.57.332] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Twelve N-terminal analogs of des-fatty acyl-polymyxin B (Des-FA-[X(1)]-PMB, X=various amino acids or peptides) were synthesized and examined for their antimicrobial activity against Escherichia coli (E. coli), Salmonella Typhimurium (S. Typhimurium) and Pseudomonas aeruginosa (P. aeruginosa). It was found that Des-FA-[Dap(1)]-, Des-FA-[Ser(1)]-, Des-FA-[Dab-Dab-Dab(1)]- and Des-FA-[Arg-Arg-Arg(1)]-PMB had potent activity only against P. aeruginosa, with MIC values of 0.5-1 nmol/ml. Analogs in which X was Lys, Arg, Leu or Ala did not have increased antimicrobial activity against the three bacterial species tested compared with the lead compounds Des-FA-[Dab(1)]-PMB and polymyxin B (PMB). Des-FA-[Trp(1)]-PMB and Des-FA-[Phe(1)]-PMB had reduced activity against P. aeruginosa. The results indicate that compact hydrophilic amino acids (C3) or basic tripeptides at the N-terminal provide specificity for bactericidal activity towards P. aeruginosa. For LPS-binding activity, Des-FA-[Dab-Dab-Dab(1)]-PMB and Des-FA-[Arg-Arg-Arg(1)]-PMB showed activity comparable to PMB, while Des-FA-[Ala-Ala-Ala(1)]-PMB showed very low activity. Reduced acute toxicity of Des-FA-[Dap(1)]-PMB and Des-FA-[Trp(1)]-PMB was demonstrated by a mouse tail intravenous administration test, with LD(50) values of 23.5 and 19.0 micromol/kg, respectively, in contrast to PMB (LD(50), 4.8 micromol/kg).
Collapse
Affiliation(s)
- Naoko Katsuma
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kunazawa, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Outer membrane permeability and antibiotic resistance. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2008; 1794:808-16. [PMID: 19100346 DOI: 10.1016/j.bbapap.2008.11.005] [Citation(s) in RCA: 1066] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 11/12/2008] [Accepted: 11/13/2008] [Indexed: 01/19/2023]
Abstract
To date most antibiotics are targeted at intracellular processes, and must be able to penetrate the bacterial cell envelope. In particular, the outer membrane of gram-negative bacteria provides a formidable barrier that must be overcome. There are essentially two pathways that antibiotics can take through the outer membrane: a lipid-mediated pathway for hydrophobic antibiotics, and general diffusion porins for hydrophilic antibiotics. The lipid and protein compositions of the outer membrane have a strong impact on the sensitivity of bacteria to many types of antibiotics, and drug resistance involving modifications of these macromolecules is common. This review will describe the molecular mechanisms for permeation of antibiotics through the outer membrane, and the strategies that bacteria have deployed to resist antibiotics by modifications of these pathways.
Collapse
|
47
|
Walther J, Bröcker MJ, Wätzlich D, Nimtz M, Rohde M, Jahn D, Moser J. Protochlorophyllide: a new photosensitizer for the photodynamic inactivation of Gram-positive and Gram-negative bacteria. FEMS Microbiol Lett 2008; 290:156-63. [PMID: 19025572 DOI: 10.1111/j.1574-6968.2008.01413.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The growing resistance against antibiotics demands the search for alternative treatment strategies. Photodynamic therapy is a promising candidate. The natural intermediate of chlorophyll biosynthesis, protochlorophyllide, was produced, purified and tested as a novel photosensitizer for the inactivation of five model organisms including Staphylococcus aureus, Listeria monocytogenes and Yersinia pseudotuberculosis, all responsible for serious clinical infections. When microorganisms were exposed to white light from a tungsten filament lamp (0.1 mW cm(-2)), Gram-positive S. aureus, L. monocytogenes and Bacillus subtilis were photochemically inactivated at concentrations of 0.5 mg L(-1) protochlorophyllide. Transmission electron microscopy revealed a disordered septum formation during cell division and the partial loss of the cytoplasmic cell contents. Gram-negative Y. pseudotuberculosis and Escherichia coli were found to be insensitive to protochlorophyllide treatment due to the permeability barrier of the outer membrane. However, the two bacteria were rendered susceptible to eradication by protochlorophyllide (10 mg L(-1)) upon addition of polymyxin B nonapeptide at 50 and 20 mg L(-1), respectively. The release of DNA and a detrimental rearrangement of the cytoplasm were observed.
Collapse
Affiliation(s)
- Johannes Walther
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Hemaiswarya S, Kruthiventi AK, Doble M. Synergism between natural products and antibiotics against infectious diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2008; 15:639-52. [PMID: 18599280 DOI: 10.1016/j.phymed.2008.06.008] [Citation(s) in RCA: 468] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Antibiotics have been effective in treating infectious diseases, but resistance to these drugs has led to the emergence of new and the reemergence of old infectious diseases. One strategy employed to overcome these resistance mechanisms is the use of combination of drugs, such as beta-lactams together with beta-lactamase inhibitors. Several plant extracts have exhibited synergistic activity against microorganisms. This review describes in detail, the observed synergy and mechanism of action between natural products including flavonoids and essential oils and synthetic drugs in effectively combating bacterial, fungal and mycobacterial infections. The mode of action of combination differs significantly than that of the same drugs acting individually; hence isolating a single component may lose its importance thereby simplifying the task of pharma industries.
Collapse
Affiliation(s)
- Shanmugam Hemaiswarya
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, India
| | | | | |
Collapse
|
49
|
Novel polymyxin derivatives carrying only three positive charges are effective antibacterial agents. Antimicrob Agents Chemother 2008; 52:3229-36. [PMID: 18591267 DOI: 10.1128/aac.00405-08] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The lack of novel antibiotics against gram-negative bacteria has reinstated polymyxins as the drugs of last resort to treat serious infections caused by extremely multiresistant gram-negative organisms. However, polymyxins are nephrotoxic, and this feature may complicate therapy or even require its discontinuation. Like that of aminoglycosides, the nephrotoxicity of polymyxins might be related to the highly cationic nature of the molecule. Colistin and polymyxin B carry five positive charges. Here we show that novel polymyxin derivatives carrying only three positive charges are effective antibacterial agents. NAB739 has a cyclic peptide portion identical to that of polymyxin B, but in the linear portion of the peptide, it carries the threonyl-D-serinyl residue (no cationic charges) instead of the diaminobutyryl-threonyl-diaminobutyryl residue (two cationic charges). The MICs of NAB739 for 17 strains of Escherichia coli were identical, or very close, to those of polymyxin B. Furthermore, NAB739 was effective against other polymyxin-susceptible strains of Enterobacteriaceae and against Acinetobacter baumannii. At subinhibitory concentrations, it dramatically sensitized A. baumannii to low concentrations of antibiotics such as rifampin, clarithromycin, vancomycin, fusidic acid, and meropenem. NAB739 methanesulfonate was a prodrug analogous to colistin methanesulfonate. NAB740 was the most active derivative against Pseudomonas aeruginosa. NAB7061 (linear portion of the peptide, threonyl-aminobutyryl) lacked direct antibacterial activity but sensitized the targets to hydrophobic antibiotics by factors up to 2,000. The affinities of the NAB compounds for isolated rat kidney brush border membrane were significantly lower than that of polymyxin B.
Collapse
|
50
|
|