201
|
Li W, Separovic F, O'Brien-Simpson NM, Wade JD. Chemically modified and conjugated antimicrobial peptides against superbugs. Chem Soc Rev 2021; 50:4932-4973. [PMID: 33710195 DOI: 10.1039/d0cs01026j] [Citation(s) in RCA: 259] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antimicrobial resistance (AMR) is one of the greatest threats to human health that, by 2050, will lead to more deaths from bacterial infections than cancer. New antimicrobial agents, both broad-spectrum and selective, that do not induce AMR are urgently required. Antimicrobial peptides (AMPs) are a novel class of alternatives that possess potent activity against a wide range of Gram-negative and positive bacteria with little or no capacity to induce AMR. This has stimulated substantial chemical development of novel peptide-based antibiotics possessing improved therapeutic index. This review summarises recent synthetic efforts and their impact on analogue design as well as their various applications in AMP development. It includes modifications that have been reported to enhance antimicrobial activity including lipidation, glycosylation and multimerization through to the broad application of novel bio-orthogonal chemistry, as well as perspectives on the direction of future research. The subject area is primarily the development of next-generation antimicrobial agents through selective, rational chemical modification of AMPs. The review further serves as a guide toward the most promising directions in this field to stimulate broad scientific attention, and will lead to new, effective and selective solutions for the several biomedical challenges to which antimicrobial peptidomimetics are being applied.
Collapse
Affiliation(s)
- Wenyi Li
- Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, VIC 3010, Australia. and Bio21 Institute, University of Melbourne, VIC 3010, Australia
| | - Frances Separovic
- Bio21 Institute, University of Melbourne, VIC 3010, Australia and School of Chemistry, University of Melbourne, VIC 3010, Australia
| | - Neil M O'Brien-Simpson
- Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, VIC 3010, Australia. and Bio21 Institute, University of Melbourne, VIC 3010, Australia
| | - John D Wade
- School of Chemistry, University of Melbourne, VIC 3010, Australia and The Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
202
|
Muñoz-López J, Oliveira JCL, Michel DAGR, Ferreira CS, Neto FG, Salnikov ES, Verly RM, Bechinger B, Resende JM. Membrane interactions of Ocellatins. Where do antimicrobial gaps stem from? Amino Acids 2021; 53:1241-1256. [PMID: 34251525 DOI: 10.1007/s00726-021-03029-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/25/2021] [Indexed: 10/20/2022]
Abstract
The antimicrobial peptides Ocellatin-LB1, -LB2 and -F1, isolated from frogs, are identical from residue 1 to 22, which correspond to the -LB1 sequence, whereas -LB2 carries an extra N and -F1 additional NKL residues at their C-termini. Despite the similar sequences, previous investigations showed different spectra of activities and biophysical investigations indicated a direct correlation between both membrane-disruptive properties and activities, i.e., ocellatin-F1 > ocellatin-LB1 > ocellatin-LB2. This study presents experimental evidence as well as results from theoretical studies that contribute to a deeper understanding on how these peptides exert their antimicrobial activities and how small differences in the amino acid composition and their secondary structure can be correlated to these activity gaps. Solid-state NMR experiments allied to the simulation of anisotropic NMR parameters allowed the determination of the membrane topologies of these ocellatins. Interestingly, the extra Asn residue at the Ocellatin-LB2 C-terminus results in increased topological flexibility, which is mainly related to wobbling of the helix main axis as noticed by molecular dynamics simulations. Binding kinetics and thermodynamics of the interactions have also been assessed by Surface Plasmon Resonance and Isothermal Titration Calorimetry. Therefore, these investigations allowed to understand in atomic detail the relationships between peptide structure and membrane topology, which are in tune within the series -F1 > > -LB1 ≥ -LB2, as well as how peptide dynamics can affect membrane topology, insertion and binding.
Collapse
Affiliation(s)
- José Muñoz-López
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, P.O. Box 486, Belo Horizonte, MG, 31270-901, Brazil.,UMR7177, Institut de Chimie, Université de Strasbourg/CNRS, 4, rue Blaise Pascal , 67000, Strasbourg, France
| | - Jade C L Oliveira
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, P.O. Box 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Daniel A G R Michel
- Departamento de Química, Universidade Federal Dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000, Brazil
| | - Carolina S Ferreira
- Departamento de Química, Universidade Federal Dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000, Brazil
| | - Francisco Gomes Neto
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, 21040-900, Brazil
| | - Evgeniy S Salnikov
- UMR7177, Institut de Chimie, Université de Strasbourg/CNRS, 4, rue Blaise Pascal , 67000, Strasbourg, France
| | - Rodrigo M Verly
- Departamento de Química, Universidade Federal Dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000, Brazil
| | - Burkhard Bechinger
- UMR7177, Institut de Chimie, Université de Strasbourg/CNRS, 4, rue Blaise Pascal , 67000, Strasbourg, France.,Institut Universitaire de France, 75005, Paris, France
| | - Jarbas M Resende
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, P.O. Box 486, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
203
|
Huebinger RM, Do DH, Carlson DL, Yao X, Stones DH, De Souza Santos M, Vaz DP, Keen E, Wolf SE, Minei JP, Francis KP, Orth K, Krachler AM. Bacterial adhesion inhibitor prevents infection in a rodent surgical incision model. Virulence 2021; 11:695-706. [PMID: 32490711 PMCID: PMC7550027 DOI: 10.1080/21505594.2020.1772652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Surgical site infection risk continues to increase due to lack of efficacy in current standard of care drugs. New methods to treat or prevent antibiotic-resistant bacterial infections are needed. Multivalent Adhesion Molecules (MAM) are bacterial adhesins required for virulence. We developed a bacterial adhesion inhibitor using recombinant MAM fragment bound to polymer scaffold, mimicking MAM7 display on the bacterial surface. Here, we test MAM7 inhibitor efficacy to prevent Gram-positive and Gram-negative infections. Using a rodent model of surgical infection, incision sites were infected with antibiotic-resistant bioluminescent strains of Staphylococcus aureus or Pseudomonas aeruginosa. Infections were treated with MAM7 inhibitor or control suspension. Bacterial abundance was quantified for nine days post infection. Inflammatory responses and histology were characterized using fixed tissue sections. MAM7 inhibitor treatment decreased burden of S. aureus and P. aeruginosa below detection threshold. Bacterial load of groups treated with control were significantly higher than MAM7 inhibitor-treated groups. Treatment with inhibitor reduced colonization of clinically-relevant pathogens in an in vivo model of surgical infection. Use of MAM7 inhibitor to block initial adhesion of bacteria to tissue in surgical incisions may reduce infection rates, presenting a strategy to mitigate overuse of antibiotics to prevent surgical site infections.
Collapse
Affiliation(s)
- R M Huebinger
- Department of Surgery, Division of General and Acute Care Surgery, University of Texas Southwestern Medical Center , Dallas, TX, USA
| | - D H Do
- Department of Surgery, Division of General and Acute Care Surgery, University of Texas Southwestern Medical Center , Dallas, TX, USA
| | - D L Carlson
- Department of Surgery, Division of General and Acute Care Surgery, University of Texas Southwestern Medical Center , Dallas, TX, USA
| | - X Yao
- Department of Surgery, Division of General and Acute Care Surgery, University of Texas Southwestern Medical Center , Dallas, TX, USA
| | - D H Stones
- School of Biosciences, Institute of Microbiology and Infection, University of Birmingham , Birmingham, UK.,University of Gloucestershire, School of Natural and Social Sciences , Cheltenham, UK
| | - M De Souza Santos
- Department of Molecular Biology, University of Texas Southwestern Medical Center , Dallas, TX, USA
| | - D P Vaz
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, McGovern Medical School , Houston, TX, USA
| | - E Keen
- School of Biosciences, Institute of Microbiology and Infection, University of Birmingham , Birmingham, UK
| | - S E Wolf
- Department of Surgery, Division of General and Acute Care Surgery, University of Texas Southwestern Medical Center , Dallas, TX, USA.,UTMB Department of Surgery, Shriners Hospitals for Children , Galveston, TX, USA
| | - J P Minei
- Department of Surgery, Division of General and Acute Care Surgery, University of Texas Southwestern Medical Center , Dallas, TX, USA
| | | | - K Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center , Dallas, TX, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, TX, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center , Dallas, TX, USA
| | - A M Krachler
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, McGovern Medical School , Houston, TX, USA
| |
Collapse
|
204
|
Zommiti M, Chikindas ML, Ferchichi M. Probiotics-Live Biotherapeutics: a Story of Success, Limitations, and Future Prospects-Not Only for Humans. Probiotics Antimicrob Proteins 2021; 12:1266-1289. [PMID: 31376026 DOI: 10.1007/s12602-019-09570-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In livestock production, lactic acid bacteria (LAB) represent the most widespread microorganisms used as probiotics. For such critical use, these bacteria must be correctly identified and characterized to ensure their safety and efficiency. Recently, probiotics have become highly recognized as supplements for humans and in particular for animals because of their beneficial outcome on health improvement and well-being maintenance. Various factors, encompassing dietary and management constraints, have been demonstrated to tremendously influence the structure, composition, and activities of gut microbial communities in farm animals. Previous investigations reported the potential of probiotics in animal diets and nutrition. But a high rate of inconsistency in the efficiency of probiotics has been reported. This may be due, in a major part, to the dynamics of the gastrointestinal microbial communities. Under stressing surroundings, the direct-fed microbials may play a key role as the salient limiting factor of the severity of the dysbiosis caused by disruption of the normal intestinal balance. Probiotics are live microorganisms, which confer health benefits on the host by positively modifying the intestinal microflora. Thus, the aim of this review is to summarize and to highlight the positive influence of probiotics and potential probiotic microbe supplementation in animal feed with mention of several limitations.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université de Tunis El-Manar, 1006, Tunis, Tunisia
| | - Michael L Chikindas
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA.,Center for Digestive Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ, USA
| | - Mounir Ferchichi
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université de Tunis El-Manar, 1006, Tunis, Tunisia.
| |
Collapse
|
205
|
Buroni S, Chiarelli LR. Antivirulence compounds: a future direction to overcome antibiotic resistance? Future Microbiol 2021; 15:299-301. [PMID: 32286100 DOI: 10.2217/fmb-2019-0294] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Silvia Buroni
- Department of Biology & Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, 27100, Italy
| | - Laurent R Chiarelli
- Department of Biology & Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, 27100, Italy
| |
Collapse
|
206
|
Roussin M, Salcedo SP. NAD+-targeting by bacteria: an emerging weapon in pathogenesis. FEMS Microbiol Rev 2021; 45:6315328. [PMID: 34223888 DOI: 10.1093/femsre/fuab037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/01/2021] [Indexed: 11/14/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a major cofactor in redox reactions in all lifeforms. A stable level of NAD+ is vital to ensure cellular homeostasis. Some pathogens can modulate NAD+ metabolism to their advantage and even utilize or cleave NAD+ from the host using specialized effectors known as ADP-ribosyltransferase toxins and NADases, leading to energy store depletion, immune evasion, or even cell death. This review explores recent advances in the field of bacterial NAD+-targeting toxins, highlighting the relevance of NAD+ modulation as an emerging pathogenesis strategy. In addition, we discuss the role of specific NAD+-targeting toxins in niche colonization and bacterial lifestyle as components of Toxin/Antitoxin systems and key players in inter-bacterial competition. Understanding the mechanisms of toxicity, regulation, and secretion of these toxins will provide interesting leads in the search for new antimicrobial treatments in the fight against infectious diseases.
Collapse
Affiliation(s)
- Morgane Roussin
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique UMR5086, Université de Lyon, Lyon, France
| | - Suzana P Salcedo
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique UMR5086, Université de Lyon, Lyon, France
| |
Collapse
|
207
|
Asempa TE, DeRosa NA, Cassino C, Lehoux D, Schuch R, Nicolau DP. Efficacy assessment of lysin CF-296 in addition to daptomycin or vancomycin against Staphylococcus aureus in the murine thigh infection model. J Antimicrob Chemother 2021; 76:2622-2628. [PMID: 34223628 DOI: 10.1093/jac/dkab206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/24/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES CF-296 is a lysin in pre-clinical development for the treatment of MSSA and MRSA infections, used in addition to standard-of-care (SOC) antibiotics. We evaluated the efficacy of CF-296 alone and in addition to daptomycin or vancomycin against Staphylococcus aureus in the neutropenic mouse thigh infection model. METHODS Eight isolates (one MSSA and seven MRSA) were studied. Mice were administered five CF-296 monotherapy doses ranging from 0.5 to 50 mg/kg intravenously. To assess adjunctive therapy, mice received sub-therapeutic daptomycin alone, sub-therapeutic vancomycin alone, or the five CF-296 doses in addition to either daptomycin or vancomycin. RESULTS Relative to starting inoculum (5.80 ± 0.31 log10 cfu/thigh), bacterial density in vehicle controls increased by +2.49 ± 0.98 across all eight strains. Relative to 24 h controls, a dose-response in bacterial killing (range -0.22 ± 0.87 to -2.01 ± 1.71 log10 cfu/thigh) was observed with increasing CF-296 monotherapy against the eight isolates. Daptomycin and vancomycin resulted in -1.36 ± 0.77 and -1.37 ± 1.01 log10 cfu/thigh bacteria reduction, respectively, relative to 24 h controls. Escalating CF-296 exposures (0.5-50 mg/kg) in addition to daptomycin resulted in an enhanced dose-response, ranging from bacterial killing of -0.69 to -2.13 log10 cfu/thigh, relative to daptomycin alone. Similarly, in addition to vancomycin, escalating CF-296 exposures resulted in bacterial reduction ranging from -1.37 to -2.29 log10 cfu/thigh, relative to vancomycin alone. CONCLUSIONS Relative to SOC antibiotics (daptomycin or vancomycin), addition of CF-296 resulted in robust and enhanced antibacterial dose-response, achieving ≥1 log10 cfu/thigh decrease across most doses, highlighting a potential role for CF-296 adjunctive therapy against MSSA and MRSA isolates.
Collapse
Affiliation(s)
- Tomefa E Asempa
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - Nicole A DeRosa
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | | | | | | | - David P Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA.,Division of Infectious Diseases, Hartford Hospital, Hartford, CT, USA
| |
Collapse
|
208
|
Wan F, Xu L, Ruan Z, Luo Q. Genomic and Transcriptomic Analysis of Colistin-Susceptible and Colistin-Resistant Isolates Identify Two-Component System EvgS/EvgA Associated with Colistin Resistance in Escherichia coli. Infect Drug Resist 2021; 14:2437-2447. [PMID: 34234474 PMCID: PMC8254184 DOI: 10.2147/idr.s316963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/15/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose Colistin is one of the last-resort antimicrobial agents that combat the increasing threat of multi-drug resistant (MDR) gram-negative bacteria. Based on the known mechanism of colistin resistance which contributes to chromosomal mutations involved in the synthesis and modification of lipopolysaccharide (LPS), we explored the regulatory genes mediate colistin resistance, by whole genome sequencing and transcriptome analysis. Materials and Methods In this study, a colistin-resistant (Colr) strain Escherichia coli ATCC 25922-R was generated from colistin-sensible (Cols) strain E. coli ATCC 25922 by colistin induction. We compared the genome and transcriptome sequencing result from Cols and Colr strain. MALDI-TOF mass spectrometry was used to detect LPS. Results Genomic analysis and complementation experiment demonstrated the PmrB amino acid substitution in ATCC 25922-R (L14R) conferred the colistin resistance phenotype. Results of RNA sequencing (RNA-Seq) and comparative transcriptome analysis indicated that the two-component system EvgS/EvgA is highly involved in the global regulation of colistin resistance. Conclusion This study demonstrated that PmrB L14R amino acid substitution resulted in colistin resistance, and two-component system EvgS/EvgA might participate in colistin resistance in E. coli.
Collapse
Affiliation(s)
- Fen Wan
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Linna Xu
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Zhi Ruan
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, People's Republic of China
| | - Qixia Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Medical School, College of medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| |
Collapse
|
209
|
PhaLP: A Database for the Study of Phage Lytic Proteins and Their Evolution. Viruses 2021; 13:v13071240. [PMID: 34206969 PMCID: PMC8310338 DOI: 10.3390/v13071240] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 01/22/2023] Open
Abstract
Phage lytic proteins are a clinically advanced class of novel enzyme-based antibiotics, so-called enzybiotics. A growing community of researchers develops phage lytic proteins with the perspective of their use as enzybiotics. A successful translation of enzybiotics to the market requires well-considered selections of phage lytic proteins in early research stages. Here, we introduce PhaLP, a database of phage lytic proteins, which serves as an open portal to facilitate the development of phage lytic proteins. PhaLP is a comprehensive, easily accessible and automatically updated database (currently 16,095 entries). Capitalizing on the rich content of PhaLP, we have mapped the high diversity of natural phage lytic proteins and conducted analyses at three levels to gain insight in their host-specific evolution. First, we provide an overview of the modular diversity. Secondly, datamining and interpretable machine learning approaches were adopted to reveal host-specific design rules for domain architectures in endolysins. Lastly, the evolution of phage lytic proteins on the protein sequence level was explored, revealing host-specific clusters. In sum, PhaLP can act as a starting point for the broad community of enzybiotic researchers, while the steadily improving evolutionary insights will serve as a natural inspiration for protein engineers.
Collapse
|
210
|
Amaral JL, Souza PFN, Oliveira JTA, Freire VN, Sousa DOB. Computational approach, scanning electron and fluorescence microscopies revealed insights into the action mechanisms of anticandidal peptide Mo-CBP 3-PepIII. Life Sci 2021; 281:119775. [PMID: 34186044 DOI: 10.1016/j.lfs.2021.119775] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 06/09/2021] [Accepted: 06/20/2021] [Indexed: 11/17/2022]
Abstract
AIMS The Candida genus is composed of opportunistic pathogens that threaten public health. Given the increase in resistance to current drugs, it is necessary to develop new drugs to treat infections by these pathogens. Antimicrobial peptides are promising alternative molecules with low cost, broad action spectrum and low resistance induction. This study aimed to clarify the action mechanisms of synthetic peptides against Candida albicans. MAIN METHODS The mode of action of the anticandidal peptides Mo-CBP3-PepIII were analyzed through molecular dynamics and quantum biochemistry methods against Exo-β-1,3-glucanase (EXG), vital to cell wall metabolism. Furthermore, scanning electron (SEM) and fluorescence (FM) microscopies were employed to corroborate the in silico data. KEY FINDINGS Mo-CBP3-PepIII strongly interacted with EXG (-122.2 kcal mol-1) at the active site, higher than the commercial inhibitor pepstatin. Also, molecular dynamics revealed the insertion of Mo-CBP3-PepIII into the yeast membrane. SEM analyses revealed that Mo-CBP3-PepIII induced cracks and scars of the cell wall and FM analyses confirmed the pore formation on the Candida membrane. SIGNIFICANCE Mo-CBP3-PepIII has strong potential as a new drug with a broad spectrum of action, given its different mode of action compared to conventional drugs.
Collapse
Affiliation(s)
- Jackson L Amaral
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil; Department of Physics, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil.
| | - Pedro F N Souza
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil
| | - Jose T A Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil
| | - Valder N Freire
- Department of Physics, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil
| | - Daniele O B Sousa
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil.
| |
Collapse
|
211
|
De Oliveira DMP, Bohlmann L, Conroy T, Jen FEC, Everest-Dass A, Hansford KA, Bolisetti R, El-Deeb IM, Forde BM, Phan MD, Lacey JA, Tan A, Rivera-Hernandez T, Brouwer S, Keller N, Kidd TJ, Cork AJ, Bauer MJ, Cook GM, Davies MR, Beatson SA, Paterson DL, McEwan AG, Li J, Schembri MA, Blaskovich MAT, Jennings MP, McDevitt CA, von Itzstein M, Walker MJ. Repurposing a neurodegenerative disease drug to treat Gram-negative antibiotic-resistant bacterial sepsis. Sci Transl Med 2021; 12:12/570/eabb3791. [PMID: 33208501 DOI: 10.1126/scitranslmed.abb3791] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022]
Abstract
The emergence of polymyxin resistance in carbapenem-resistant and extended-spectrum β-lactamase (ESBL)-producing bacteria is a critical threat to human health, and alternative treatment strategies are urgently required. We investigated the ability of the hydroxyquinoline analog ionophore PBT2 to restore antibiotic sensitivity in polymyxin-resistant, ESBL-producing, carbapenem-resistant Gram-negative human pathogens. PBT2 resensitized Klebsiella pneumoniae, Escherichia coli, Acinetobacter baumannii, and Pseudomonas aeruginosa to last-resort polymyxin class antibiotics, including the less toxic next-generation polymyxin derivative FADDI-287, in vitro. We were unable to select for mutants resistant to PBT2 + FADDI-287 in polymyxin-resistant E. coli containing a plasmid-borne mcr-1 gene or K. pneumoniae carrying a chromosomal mgrB mutation. Using a highly invasive K. pneumoniae strain engineered for polymyxin resistance through mgrB mutation, we successfully demonstrated the efficacy of PBT2 + polymyxin (colistin or FADDI-287) for the treatment of Gram-negative sepsis in immunocompetent mice. In comparison to polymyxin alone, the combination of PBT2 + polymyxin improved survival and reduced bacterial dissemination to the lungs and spleen of infected mice. These data present a treatment modality to break antibiotic resistance in high-priority polymyxin-resistant Gram-negative pathogens.
Collapse
Affiliation(s)
- David M P De Oliveira
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Lisa Bohlmann
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Trent Conroy
- Institute for Glycomics, Griffith University, Queensland 4222, Australia
| | - Freda E-C Jen
- Institute for Glycomics, Griffith University, Queensland 4222, Australia
| | - Arun Everest-Dass
- Institute for Glycomics, Griffith University, Queensland 4222, Australia
| | - Karl A Hansford
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Raghu Bolisetti
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ibrahim M El-Deeb
- Institute for Glycomics, Griffith University, Queensland 4222, Australia
| | - Brian M Forde
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia.,Centre for Clinical Research and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4029, Australia
| | - Minh-Duy Phan
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Jake A Lacey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria 3000, Australia
| | - Aimee Tan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria 3000, Australia
| | - Tania Rivera-Hernandez
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia.,Consejo Nacional de Ciencia y Tecnología-Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Stephan Brouwer
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Nadia Keller
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Timothy J Kidd
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Amanda J Cork
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Michelle J Bauer
- Centre for Clinical Research and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4029, Australia
| | - Gregory M Cook
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand
| | - Mark R Davies
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria 3000, Australia
| | - Scott A Beatson
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - David L Paterson
- Centre for Clinical Research and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4029, Australia
| | - Alastair G McEwan
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria 3800, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Michael P Jennings
- Institute for Glycomics, Griffith University, Queensland 4222, Australia
| | - Christopher A McDevitt
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria 3000, Australia
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Queensland 4222, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia.
| |
Collapse
|
212
|
Plantaricin NC8 αβ prevents Staphylococcus aureus-mediated cytotoxicity and inflammatory responses of human keratinocytes. Sci Rep 2021; 11:12514. [PMID: 34131160 PMCID: PMC8206081 DOI: 10.1038/s41598-021-91682-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Multidrug resistance bacteria constitue an increasing global health problem and the development of novel therapeutic strategies to face this challenge is urgent. Antimicrobial peptides have been proven as potent agents against pathogenic bacteria shown by promising in vitro results. The aim of this study was to characterize the antimicrobial effects of PLNC8 αβ on cell signaling pathways and inflammatory responses of human keratinocytes infected with S. aureus. PLNC8 αβ did not affect the viability of human keratinocytes but upregulated several cytokines (IL-1β, IL-6, CXCL8), MMPs (MMP1, MMP2, MMP9, MMP10) and growth factors (VEGF and PDGF-AA), which are essential in cell regeneration. S. aureus induced the expression of several inflammatory mediators at the gene and protein level and PLNC8 αβ was able to significantly suppress these effects. Intracellular signaling events involved primarily c-Jun via JNK, c-Fos and NFκB, suggesting their essential role in the initiation of inflammatory responses in human keratinocytes. PLNC8 αβ was shown to modulate early keratinocyte responses, without affecting their viability. The peptides have high selectivity towards S. aureus and were efficient at eliminating the bacteria and counteracting their inflammatory and cytotoxic effects, alone and in combination with low concentrations of gentamicin. We propose that PLNC8 αβ may be developed to combat infections caused by Staphylococcus spp.
Collapse
|
213
|
Comparison of a Short Linear Antimicrobial Peptide with Its Disulfide-Cyclized and Cyclotide-Grafted Variants against Clinically Relevant Pathogens. Microorganisms 2021; 9:microorganisms9061249. [PMID: 34201398 PMCID: PMC8228819 DOI: 10.3390/microorganisms9061249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
According to the World Health Organization (WHO) the development of resistance against antibiotics by microbes is one of the most pressing health concerns. The situation will intensify since only a few pharmacological companies are currently developing novel antimicrobial compounds. Discovery and development of novel antimicrobial compounds with new modes of action are urgently needed. Antimicrobial peptides (AMPs) are known to be able to kill multidrug-resistant bacteria and, therefore, of interest to be developed into antimicrobial drugs. Proteolytic stability and toxicities of these peptides are challenges to overcome, and one strategy frequently used to address stability is cyclization. Here we introduced a disulfide-bond to cyclize a potent and nontoxic 9mer peptide and, in addition, as a proof-of-concept study, grafted this peptide into loop 6 of the cyclotide MCoTI-II. This is the first time an antimicrobial peptide has been successfully grafted onto the cyclotide scaffold. The disulfide-cyclized and grafted cyclotide showed moderate activity in broth and strong activity in 1/5 broth against clinically relevant resistant pathogens. The linear peptide showed superior activity in both conditions. The half-life time in 100% human serum was determined, for the linear peptide, to be 13 min, for the simple disulfide-cyclized peptide, 9 min, and, for the grafted cyclotide 7 h 15 min. The addition of 10% human serum led to a loss of antimicrobial activity for the different organisms, ranging from 1 to >8-fold for the cyclotide. For the disulfide-cyclized version and the linear version, activity also dropped to different degrees, 2 to 18-fold, and 1 to 30-fold respectively. Despite the massive difference in stability, the linear peptide still showed superior antimicrobial activity. The cyclotide and the disulfide-cyclized version demonstrated a slower bactericidal effect than the linear version. All three peptides were stable at high and low pH, and had very low hemolytic and cytotoxic activity.
Collapse
|
214
|
Ferriol-González C, Domingo-Calap P. Phage Therapy in Livestock and Companion Animals. Antibiotics (Basel) 2021; 10:559. [PMID: 34064754 PMCID: PMC8150778 DOI: 10.3390/antibiotics10050559] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
The irrational use of antibiotics has led to a high emergence of multi-drug resistant (MDR) bacteria. The traditional overuse of antibiotics in the animal feed industry plays a crucial role in the emergence of these pathogens that pose both economic and health problems. In addition, antibiotics have also recently experienced an increase to treat companion animal infections, promoting the emergence of MDR bacteria in pets, which can reach humans. Phages have been proposed as an alternative for antibiotics for the treatment of livestock and companion animal infections due to their multiple advantages as adaptative drugs, such as their ability to evolve, to multiply at the site of infections, and their high specificity. Moreover, phage-derived enzymes may also be an interesting approach. However, the lack of regulation for this type of pharmaceutical hinders its potential commercialization. In this review, we summarize the main recent studies on phage therapy in livestock and companion animals, providing an insight into current advances in this area and the future of treatments for bacterial infections.
Collapse
Affiliation(s)
| | - Pilar Domingo-Calap
- Department of Genetics, Universitat de València, 46100 Valencia, Spain;
- Institute for Integrative Systems Biology, I2SysBio, Universitat de València-CSIC, 46980 Valencia, Spain
| |
Collapse
|
215
|
López-Siles M, Corral-Lugo A, McConnell MJ. Vaccines for multidrug resistant Gram negative bacteria: lessons from the past for guiding future success. FEMS Microbiol Rev 2021; 45:fuaa054. [PMID: 33289833 DOI: 10.1093/femsre/fuaa054] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial resistance is a major threat to global public health. Vaccination is an effective approach for preventing bacterial infections, however it has not been successfully applied to infections caused by some of the most problematic multidrug resistant pathogens. In this review, the potential for vaccines to contribute to reducing the burden of disease of infections caused by multidrug resistant Gram negative bacteria is presented. Technical, logistical and societal hurdles that have limited successful vaccine development for these infections in the past are identified, and recent advances that can contribute to overcoming these challenges are assessed. A synthesis of vaccine technologies that have been employed in the development of vaccines for key multidrug resistant Gram negative bacteria is included, and emerging technologies that may contribute to future successes are discussed. Finally, a comprehensive review of vaccine development efforts over the last 40 years for three of the most worrisome multidrug resistant Gram negative pathogens, Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa is presented, with a focus on recent and ongoing studies. Finally, future directions for the vaccine development field are highlighted.
Collapse
Affiliation(s)
- Mireia López-Siles
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Andrés Corral-Lugo
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Michael J McConnell
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
216
|
Coburn J, Bone F, Hopkins MM, Stirling A, Mestre-Ferrandiz J, Arapostathis S, Llewelyn MJ. Appraising research policy instrument mixes: a multicriteria mapping study in six European countries of diagnostic innovation to manage antimicrobial resistance. RESEARCH POLICY 2021; 50:104140. [PMID: 33941992 PMCID: PMC8039188 DOI: 10.1016/j.respol.2020.104140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The following contributions are provided: A general process for conducting systematic prospective appraisal of policy options within the field of research policy, using multicriteria mapping (MCM). Novel methods for systematically analysing MCM data using pairwise comparison and merit orders which improve the utility of the MCM method. Quantitative findings (derived using MCM) on the appraisal of policy options for antimicrobial resistance (AMR) by 47 experts from six European countries, providing seven stakeholder perspectives, and identifying most and least favoured policy options. Qualitative findings (derived using MCM) addressing different modes of reasoning as to why contrasting policy options may be expected to perform well or poorly in different settings and from different perspectives. Qualitative findings (derived using MCM) underpinning a framework for policy design that may be useful for research policy around AMR. Quantitative findings (derived not using MCM) concerning those particular options that interviewees identified as most complementary to each other, supported by qualitative illustrations and qualifications.
This article provides prospective appraisal of key policy instruments intended to stimulate innovation to combat antimicrobial resistance (AMR). AMR refers to the ability of microbes to evolve resistance to those treatments designed to kill them, and is associated with the overuse or misuse of medicines such as antibiotics. AMR is an emerging global challenge with major implications for healthcare and society as a whole. Diagnostic tests for infectious diseases can guide decision making when prescribing medicines, so reducing inappropriate drug use. In the context of growing international interest in policies to stimulate innovation in AMR diagnostics, this study uses multicriteria mapping (MCM) to appraise a range of policy instruments in order to understand their potential performance while also highlighting the uncertainties that stakeholders hold about such interventions in complex contexts. A contribution of the article is the demonstration of a novel method to analyse and visualise MCM data in order to reveal stakeholder inclinations towards particular options while exploring interviewees’ uncertainties about the effectiveness of each instrument's design or implementation. The article reports results from six European countries (Germany, Greece, Italy, the Netherlands, Spain and the UK). The findings reveal which policy instruments are deemed most likely to perform well, and why, across stakeholder groups and national settings, with areas of common ground and difference being identified. Importantly, the conclusions presented here differ from prominent policy discourse, with international implications for the design of mixes of policy instruments to combat AMR. Strategic and practical methodological implications also emerge for general appraisal of innovation policy instrument mixes.
Collapse
Affiliation(s)
- Josie Coburn
- Science Policy Research Unit, University of Sussex Business School, University of Sussex, Falmer, Brighton, United Kingdom
| | - Frederique Bone
- Science Policy Research Unit, University of Sussex Business School, University of Sussex, Falmer, Brighton, United Kingdom
| | - Michael M Hopkins
- Science Policy Research Unit, University of Sussex Business School, University of Sussex, Falmer, Brighton, United Kingdom
| | - Andy Stirling
- Science Policy Research Unit, University of Sussex Business School, University of Sussex, Falmer, Brighton, United Kingdom
| | | | - Stathis Arapostathis
- Department of History and Philosophy of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Martin J Llewelyn
- Brighton and Sussex Medical School, Falmer, Brighton, United Kingdom
| |
Collapse
|
217
|
Zhu J, Hu C, Zeng Z, Deng X, Zeng L, Xie S, Fang Y, Jin Y, Alezra V, Wan Y. Polymyxin B-inspired non-hemolytic tyrocidine A analogues with significantly enhanced activity against gram-negative bacteria: How cationicity impacts cell specificity and antibacterial mechanism. Eur J Med Chem 2021; 221:113488. [PMID: 33991963 DOI: 10.1016/j.ejmech.2021.113488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/31/2021] [Accepted: 04/18/2021] [Indexed: 11/25/2022]
Abstract
Naturally occurring cyclic antimicrobial peptides (AMPs) such as tyrocidine A (Tyrc A) and gramicidin S (GS) are appealing targets for the development of novel antibiotics. However, their therapeutic potentials are limited by undesired hemolytic activity and relatively poor activity against Gram-negative bacteria. Inspired by polycationic lipopeptide polymyxin B (PMB), the so called 'last-resort' antibiotic for the treatment of infections caused by multidrug-resistant Gram-negative bacteria, we synthesized and biologically evaluated a series of polycationic analogues derived from Tyrc A. We were able to obtain peptide 8 that possesses 5 positive charges exhibiting potent activities against both Gram-negative and Gram-positive bacteria along with totally diminished hemolytic activity. Intriguingly, antibacterial mechanism studies revealed that, rather than the 'pore forming' model that possessed by Tyrc A, peptide 8 likely diffuses membrane in a 'detergent-like' manner. Furthermore, when treating mice with peritonitis-sepsis, peptide 8 showed excellent antibacterial and anti-inflammatory activities in vivo.
Collapse
Affiliation(s)
- Jibao Zhu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China
| | - Chengfei Hu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China
| | - Zizhen Zeng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China
| | - Xiaoyu Deng
- Minist Educ, Key Lab Modern Preparat TCM, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China
| | - Lingbing Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Donghu, Nanchang, 330006, PR China
| | - Saisai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China
| | - Yuanying Fang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China
| | - Valérie Alezra
- Laboratoire de Méthodologie, Synthèse et Molécules Thérapeutiques (ICMMO), UMR 8182, CNRS, Université Paris-Saclay, Bât 410, Facultédes Sciences D'Orsay, Orsay, 291405, France
| | - Yang Wan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, PR China; Laboratoire de Méthodologie, Synthèse et Molécules Thérapeutiques (ICMMO), UMR 8182, CNRS, Université Paris-Saclay, Bât 410, Facultédes Sciences D'Orsay, Orsay, 291405, France; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, 15 Yuchai Road, Guilin, 541004, PR China.
| |
Collapse
|
218
|
Michelet R, Ursino M, Boulet S, Franck S, Casilag F, Baldry M, Rolff J, van Dyk M, Wicha SG, Sirard JC, Comets E, Zohar S, Kloft C. The Use of Translational Modelling and Simulation to Develop Immunomodulatory Therapy as an Adjunct to Antibiotic Treatment in the Context of Pneumonia. Pharmaceutics 2021; 13:601. [PMID: 33922017 PMCID: PMC8143524 DOI: 10.3390/pharmaceutics13050601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
The treatment of respiratory tract infections is threatened by the emergence of bacterial resistance. Immunomodulatory drugs, which enhance airway innate immune defenses, may improve therapeutic outcome. In this concept paper, we aim to highlight the utility of pharmacometrics and Bayesian inference in the development of immunomodulatory therapeutic agents as an adjunct to antibiotics in the context of pneumonia. For this, two case studies of translational modelling and simulation frameworks are introduced for these types of drugs up to clinical use. First, we evaluate the pharmacokinetic/pharmacodynamic relationship of an experimental combination of amoxicillin and a TLR4 agonist, monophosphoryl lipid A, by developing a pharmacometric model accounting for interaction and potential translation to humans. Capitalizing on this knowledge and associating clinical trial extrapolation and statistical modelling approaches, we then investigate the TLR5 agonist flagellin. The resulting workflow combines expert and prior knowledge on the compound with the in vitro and in vivo data generated during exploratory studies in order to construct high-dimensional models considering the pharmacokinetics and pharmacodynamics of the compound. This workflow can be used to refine preclinical experiments, estimate the best doses for human studies, and create an adaptive knowledge-based design for the next phases of clinical development.
Collapse
Affiliation(s)
- Robin Michelet
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| | - Moreno Ursino
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, Université de Paris, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, F-75019 Paris, France;
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
| | - Sandrine Boulet
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Sebastian Franck
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Fiordiligie Casilag
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Mara Baldry
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Jens Rolff
- Department of Evolutionary Biology, Institute of Biology, Freie Universitaet Berlin, 14195 Berlin, Germany;
| | - Madelé van Dyk
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia;
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Jean-Claude Sirard
- CNRS, Inserm, CHU Lille, Institute Pasteur de Lille, U1019-UMR9017-CIIL-Centre for Infection and Immunity of Lille, Université de Lille, F-59000 Lille, France; (F.C.); (M.B.); (J.-C.S.)
| | - Emmanuelle Comets
- INSERM, University Rennes-1, CIC 1414, F-35000 Rennes, France;
- INSERM, IAME, Université de Paris, F-75006 Paris, France
| | - Sarah Zohar
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, F-75006 Paris, France; (S.B.); (S.Z.)
- HeKA, Inria, F-75006 Paris, France
| | - Charlotte Kloft
- Department of Clinical Pharmacy & Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (C.K.)
| |
Collapse
|
219
|
Zigangirova NA, Nesterenko LN, Sheremet AB, Soloveva AV, Luyksaar SI, Zayakin ES, Balunets DV, Gintsburg AL. Fluorothiazinon, a small-molecular inhibitor of T3SS, suppresses salmonella oral infection in mice. J Antibiot (Tokyo) 2021; 74:244-254. [PMID: 33479520 DOI: 10.1038/s41429-020-00396-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/05/2020] [Accepted: 11/18/2020] [Indexed: 01/29/2023]
Abstract
Therapeutic strategies that target bacterial virulence have received considerable attention. The type III secretion system (T3SS) is important for bacterial virulence and represents an attractive therapeutic target. Recently, we developed a new small-molecule inhibitor belonging to a class 2,4-disubstituted-4H-[1,3,4]-thiadiazine-5-ones, Fluorothiazinon (FT-previously called CL-55). FT effectively suppressed T3SS of Chlamydia spp., Pseudomonas aeruginosa, and Salmonella without affecting bacterial growth in vitro. FT was previously characterized by low toxicity, stability, and therapeutic efficacy in animal models. Salmonella T3SS inhibition by FT was studied using in vitro assays for effector proteins detection and estimation of salmonella replication in peritoneal macrophages. The antibacterial effect of FT in vivo was investigated in murine models of salmonella chronic systemic and acute infection. Oral administration of the virulent strain of Salmonella enterica serovar Typhimurium to mice-induced chronic systemic infection with the pathogen persistence in different lymphoid organs such as spleens, Peyer's plaques, and mesenteric lymph nodes. We found that FT suppressed orally induced salmonella infection both with therapeutic and prophylactic administration. Treatment by FT at a dose of 50 mg/kg for 4 days starting from day 7 post-infection (therapy) as well as for 4 days before infection (prevention) led to practically complete eradication of salmonella in mice. FT shows a strong potential for antibacterial therapy and could be used as a substance in the design of antibacterial drugs for pharmaceutical intervention including therapy of antibiotic-resistant infections.
Collapse
Affiliation(s)
- Nailya A Zigangirova
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia.
| | - Ludmila N Nesterenko
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Anna B Sheremet
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Anna V Soloveva
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Sergey I Luyksaar
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Egor S Zayakin
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Denis V Balunets
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| | - Alexandr L Gintsburg
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, Gamaleya str.18, Moscow, 123098, Russia
| |
Collapse
|
220
|
Bamgbose T, Anvikar AR, Alberdi P, Abdullahi IO, Inabo HI, Bello M, Cabezas-Cruz A, de la Fuente J. Functional Food for the Stimulation of the Immune System Against Malaria. Probiotics Antimicrob Proteins 2021; 13:1254-1266. [PMID: 33791994 PMCID: PMC8012070 DOI: 10.1007/s12602-021-09780-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 12/20/2022]
Abstract
Drug resistance has become a threat to global health, and new interventions are needed to control major infectious diseases. The composition of gut microbiota has been linked to human health and has been associated with severity of malaria. Fermented foods contribute to the community of healthy gut bacteria. Despite the studies connecting gut microbiota to the prevention of malaria transmission and severity, research on developing functional foods for the purpose of manipulating the gut microbiota for malaria control is limited. This review summarizes recent knowledge on the role of the gut microbiota in malaria prevention and treatment. This information should encourage the search for lactic acid bacteria expressing α-Gal and those that exhibit the desired immune stimulating properties for the development of functional food and probiotics for malaria control.
Collapse
Affiliation(s)
- Timothy Bamgbose
- ICMR, -National Institute of Malaria Research, Sector 8, Dwarka, New Delhi, India
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Anupkumar R Anvikar
- ICMR, -National Institute of Malaria Research, Sector 8, Dwarka, New Delhi, India
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Isa O Abdullahi
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Helen I Inabo
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Mohammed Bello
- Department of Veterinary Public Health and Preventive Medicine, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire D'Alfort, Université Paris-Est, 94700, Maisons-Alfort, France
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain.
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
221
|
Grigonyte AM, Hapeshi A, Constantinidou C, Millard A. Modification of Bacteriophages to Increase Their Association with Lung Epithelium Cells In Vitro. Pharmaceuticals (Basel) 2021; 14:308. [PMID: 33915737 PMCID: PMC8067280 DOI: 10.3390/ph14040308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
There is currently a renaissance in research on bacteriophages as alternatives to antibiotics. Phage specificity to their bacterial host, in addition to a plethora of other advantages, makes them ideal candidates for a broad range of applications, including bacterial detection, drug delivery, and phage therapy in particular. One issue obstructing phage efficiency in phage therapy settings is their poor localization to the site of infection in the human body. Here, we engineered phage T7 with lung tissue targeting homing peptides. We then used in vitro studies to demonstrate that the engineered T7 phages had a more significant association with the lung epithelium cells than wild-type T7. In addition, we showed that, in general, there was a trend of increased association of engineered phages with the lung epithelium cells but not mouse fibroblast cells, allowing for targeted tissue specificity. These results indicate that appending phages with homing peptides would potentially allow for greater phage concentrations and greater efficacy at the infection site.
Collapse
Affiliation(s)
- Aurelija M. Grigonyte
- Warwick Integrative Synthetic Biology Centre (WISB) and School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK;
| | - Alexia Hapeshi
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK;
| | | | - Andrew Millard
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| |
Collapse
|
222
|
Franck S, Michelet R, Casilag F, Sirard JC, Wicha SG, Kloft C. A Model-Based Pharmacokinetic/Pharmacodynamic Analysis of the Combination of Amoxicillin and Monophosphoryl Lipid A Against S. pneumoniae in Mice. Pharmaceutics 2021; 13:469. [PMID: 33808396 PMCID: PMC8065677 DOI: 10.3390/pharmaceutics13040469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
Combining amoxicillin with the immunostimulatory toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) represents an innovative approach for enhancing antibacterial treatment success. Exploiting pharmacokinetic and pharmacodynamic data from an infection model of Streptococcus pneumoniae infected mice, we aimed to evaluate the preclinical exposure-response relationship of amoxicillin with MPLA coadministration and establish a link to survival. Antibiotic serum concentrations, bacterial numbers in lung and spleen and survival data of mice being untreated or treated with amoxicillin (four dose levels), MPLA, or their combination were analyzed by nonlinear mixed-effects modelling and time-to-event analysis using NONMEM® to characterize these treatment regimens. On top of a pharmacokinetic interaction, regarding the pharmacodynamic effects the combined treatment was superior to both monotherapies: The amoxicillin efficacy at highest dose was increased by a bacterial reduction of 1.74 log10 CFU/lung after 36 h and survival was increased 1.35-fold to 90.3% after 14 days both compared to amoxicillin alone. The developed pharmacometric pharmacokinetic/pharmacodynamic disease-treatment-survival models provided quantitative insights into a novel treatment option against pneumonia revealing a pharmacokinetic interaction and enhanced activity of amoxicillin and the immune system stimulator MPLA in combination. Further development of this drug combination flanked with pharmacometrics towards the clinical setting seems promising.
Collapse
Affiliation(s)
- Sebastian Franck
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (R.M.)
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (R.M.)
| | - Fiordiligie Casilag
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR8204-CIIL-Center of Infection and Immunity of Lille, University Lille, 59019 Lille, France; (F.C.); (J.-C.S.)
| | - Jean-Claude Sirard
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR8204-CIIL-Center of Infection and Immunity of Lille, University Lille, 59019 Lille, France; (F.C.); (J.-C.S.)
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (R.M.)
| |
Collapse
|
223
|
Broendum SS, Williams DE, Hayes BK, Kraus F, Fodor J, Clifton BE, Geert Volbeda A, Codee JDC, Riley BT, Drinkwater N, Farrow KA, Tsyganov K, Heselpoth RD, Nelson DC, Jackson CJ, Buckle AM, McGowan S. High avidity drives the interaction between the streptococcal C1 phage endolysin, PlyC, with the cell surface carbohydrates of Group A Streptococcus. Mol Microbiol 2021; 116:397-415. [PMID: 33756056 DOI: 10.1111/mmi.14719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 01/03/2023]
Abstract
Endolysin enzymes from bacteriophage cause bacterial lysis by degrading the peptidoglycan cell wall. The streptococcal C1 phage endolysin PlyC, is the most potent endolysin described to date and can rapidly lyse group A, C, and E streptococci. PlyC is known to bind the Group A streptococcal cell wall, but the specific molecular target or the binding site within PlyC remain uncharacterized. Here we report for the first time, that the polyrhamnose backbone of the Group A streptococcal cell wall is the binding target of PlyC. We have also characterized the putative rhamnose binding groove of PlyC and found four key residues that were critical to either the folding or the cell wall binding action of PlyC. Based on our results, we suggest that the interaction between PlyC and the cell wall may not be a high-affinity interaction as previously proposed, but rather a high avidity one, allowing for PlyC's remarkable lytic activity. Resistance to our current antibiotics is reaching crisis levels and there is an urgent need to develop the antibacterial agents with new modes of action. A detailed understanding of this potent endolysin may facilitate future developments of PlyC as a tool against the rise of antibiotic resistance.
Collapse
Affiliation(s)
- Sebastian S Broendum
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia.,Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Daniel E Williams
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Brooke K Hayes
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Felix Kraus
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - James Fodor
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia.,Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Ben E Clifton
- Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Anne Geert Volbeda
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Jeroen D C Codee
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Blake T Riley
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia.,Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, USA
| | - Nyssa Drinkwater
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Kylie A Farrow
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Kirill Tsyganov
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia.,Monash Bioinformatics Platform, Monash University, Melbourne, VIC, Australia
| | - Ryan D Heselpoth
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, MD, USA
| | - Daniel C Nelson
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Rockville, MD, USA
| | - Colin J Jackson
- Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Ashley M Buckle
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Sheena McGowan
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
224
|
Vinuesa V, McConnell MJ. Recent Advances in Iron Chelation and Gallium-Based Therapies for Antibiotic Resistant Bacterial Infections. Int J Mol Sci 2021; 22:2876. [PMID: 33809032 PMCID: PMC8000330 DOI: 10.3390/ijms22062876] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Iron is essential for multiple bacterial processes and is thus required for host colonization and infection. The antimicrobial activity of multiple iron chelators and gallium-based therapies against different bacterial species has been characterized in preclinical studies. In this review, we provide a synthesis of studies characterizing the antimicrobial activity of the major classes of iron chelators (hydroxamates, aminocarboxylates and hydroxypyridinones) and gallium compounds. Special emphasis is placed on recent in-vitro and in-vivo studies with the novel iron chelator DIBI. Limitations associated with iron chelation and gallium-based therapies are presented, with emphasis on limitations of preclinical models, lack of understanding regarding mechanisms of action, and potential host toxicity. Collectively, these studies demonstrate potential for iron chelators and gallium to be used as antimicrobial agents, particularly in combination with existing antibiotics. Additional studies are needed in order to characterize the activity of these compounds under physiologic conditions and address potential limitations associated with their clinical use as antimicrobial agents.
Collapse
Affiliation(s)
| | - Michael J. McConnell
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28220 Madrid, Spain;
| |
Collapse
|
225
|
Rapid and High-Throughput Evaluation of Diverse Configurations of Engineered Lysins Using the VersaTile Technique. Antibiotics (Basel) 2021; 10:antibiotics10030293. [PMID: 33799561 PMCID: PMC7998686 DOI: 10.3390/antibiotics10030293] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/17/2022] Open
Abstract
Bacteriophage-encoded lysins are an emerging class of antibacterial enzymes based on peptidoglycan degradation. The modular composition of lysins is a hallmark feature enabling optimization of antibacterial and pharmacological properties by engineering of lysin candidates based on lysin and non-lysin modules. In this regard, the recent introduction of the VersaTile technique allows the rapid construction of large modular lysin libraries based on a premade repository of building blocks. In this study, we perform a high-throughput construction and screening of five combinatorial lysin libraries with different configurations, targeting Klebsiella pneumoniae. An elaborate analysis of the activity distribution of 940 variants and sequencing data of 74 top hits inhibiting the growth of Klebsiella pneumoniae could be associated with specific design rules. Specific outer membrane permeabilizing peptides (OMPs) and enzymatically active domains (EADs) are significantly overrepresented among the top hits, while cell wall binding domains (CBDs) are equally represented. Especially libraries with the configuration (OMP-linker-CBD-EAD) and the inverse configuration (CBD-EAD-linker-OMP) yield the most active variants, with discernible clusters of variants that emerge above the remaining variants. The approach implemented here provides a blueprint for discovery campaigns of engineered lysins starting from libraries with different configurations and compositions.
Collapse
|
226
|
Han H, Teng D, Mao R, Hao Y, Yang N, Wang Z, Li T, Wang X, Wang J. Marine Peptide-N6NH2 and Its Derivative-GUON6NH2 Have Potent Antimicrobial Activity Against Intracellular Edwardsiella tarda in vitro and in vivo. Front Microbiol 2021; 12:637427. [PMID: 33767681 PMCID: PMC7985170 DOI: 10.3389/fmicb.2021.637427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/10/2021] [Indexed: 12/03/2022] Open
Abstract
Edwardsiella tarda is a facultative intracellular pathogen in humans and animals. There is no effective way except vaccine candidates to eradicate intracellular E. tarda. In this study, four derivatives of marine peptide-N6NH2 were designed by an introduction of unnatural residues or substitution of natural ones, and their intracellular activities against E. tarda were evaluated in macrophages and in mice, respectively. The minimum inhibitory concentration (MIC) value of N6NH2 and GUON6NH2 against E. tarda was 8 μg/mL. GUON6NH2 showed higher stability to trypsin, lower toxicity (<1%) and longer post-antibiotic effect (PAE) than N6NH2 and other derivatives. Antibacterial mechanism results showed that GUON6NH2 could bind to LPS and destroyed outer/inner cell membranes of E. tarda, superior to N6NH2 and norfloxacin. Both N6NH2 and GUON6NH2 were internalized into macrophages mainly via lipid rafts, micropinocytosis, and microtubule polymerization, respectively, and distributed in the cytoplasm. The intracellular inhibition rate of GUON6NH2 against E. tarda was 97.05–100%, higher than that in case of N6NH2 (96.82–100%). In the E. tarda-induced peritonitis mouse model, after treatment with of 1 μmol/kg N6NH2 and GUON6NH2, intracellular bacterial numbers were reduced by 1.54- and 1.97-Log10 CFU, respectively, higher than norfloxacin (0.35-Log10 CFU). These results suggest that GUON6NH2 may be an excellent candidate for novel antimicrobial agents to treat infectious diseases caused by intracellular E. tarda.
Collapse
Affiliation(s)
- Huihui Han
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Zhenlong Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ting Li
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Xiumin Wang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China.,Chinese Herbal Medicine Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
227
|
Liu Y, Tong Z, Shi J, Li R, Upton M, Wang Z. Drug repurposing for next-generation combination therapies against multidrug-resistant bacteria. Theranostics 2021; 11:4910-4928. [PMID: 33754035 PMCID: PMC7978324 DOI: 10.7150/thno.56205] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial resistance has been a global health challenge that threatens our ability to control and treat life-threatening bacterial infections. Despite ongoing efforts to identify new drugs or alternatives to antibiotics, no new classes of antibiotic or their alternatives have been clinically approved in the last three decades. A combination of antibiotics and non-antibiotic compounds that could inhibit bacterial resistance determinants or enhance antibiotic activity offers a sustainable and effective strategy to confront multidrug-resistant bacteria. In this review, we provide a brief overview of the co-evolution of antibiotic discovery and the development of bacterial resistance. We summarize drug-drug interactions and uncover the art of repurposing non-antibiotic drugs as potential antibiotic adjuvants, including discussing classification and mechanisms of action, as well as reporting novel screening platforms. A pathogen-by-pathogen approach is then proposed to highlight the critical value of drug repurposing and its therapeutic potential. Finally, general advantages, challenges and development trends of drug combination strategy are discussed.
Collapse
Affiliation(s)
- Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ziwen Tong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jingru Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ruichao Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Mathew Upton
- School of Biomedical Sciences, University of Plymouth, Drake Circus, Plymouth, UK
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
228
|
d-Leucine Modified CM11 Peptide has Admissible Antibacterial Activity with Low Cytotoxic Properties Against Vero Cell Line. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10182-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
229
|
Aghaee BL, Khan Mirzaei M, Alikhani MY, Mojtahedi A, Maurice CF. Improving the Inhibitory Effect of Phages against Pseudomonas aeruginosa Isolated from a Burn Patient Using a Combination of Phages and Antibiotics. Viruses 2021; 13:334. [PMID: 33670028 PMCID: PMC7926668 DOI: 10.3390/v13020334] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
Antibiotic resistance causes around 700,000 deaths a year worldwide. Without immediate action, we are fast approaching a post-antibiotic era in which common infections can result in death. Pseudomonas aeruginosa is the leading cause of nosocomial infection and is also one of the three bacterial pathogens in the WHO list of priority bacteria for developing new antibiotics against. A viable alternative to antibiotics is to use phages, which are bacterial viruses. Yet, the isolation of phages that efficiently kill their target bacteria has proven difficult. Using a combination of phages and antibiotics might increase treatment efficacy and prevent the development of resistance against phages and/or antibiotics, as evidenced by previous studies. Here, in vitro populations of a Pseudomonas aeruginosa strain isolated from a burn patient were treated with a single phage, a mixture of two phages (used simultaneously and sequentially), and the combination of phages and antibiotics (at sub-minimum inhibitory concentration (MIC) and MIC levels). In addition, we tested the stability of these phages at different temperatures, pH values, and in two burn ointments. Our results show that the two-phages-one-antibiotic combination had the highest killing efficiency against the P. aeruginosa strain. The phages tested showed low stability at high temperatures, acidic pH values, and in the two ointments. This work provides additional support for the potential of using combinations of phage-antibiotic cocktails at sub-MIC levels for the treatment of multidrug-resistant P. aeruginosa infections.
Collapse
Affiliation(s)
- Bahareh Lashtoo Aghaee
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan 65178-38678, Iran;
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Center Munich and Technical University of Munich, 85764 Neuherberg, Germany;
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 0B1, Canada
| | - Mohammad Yousef Alikhani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan 65178-38678, Iran;
| | - Ali Mojtahedi
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht 41938-33697, Iran
| | - Corinne F. Maurice
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 0B1, Canada
| |
Collapse
|
230
|
Splichalova A, Donovan SM, Tlaskalova-Hogenova H, Stranak Z, Splichalova Z, Splichal I. Monoassociation of Preterm Germ-Free Piglets with Bifidobacterium animalis Subsp. lactis BB-12 and Its Impact on Infection with Salmonella Typhimurium. Biomedicines 2021; 9:biomedicines9020183. [PMID: 33670419 PMCID: PMC7917597 DOI: 10.3390/biomedicines9020183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023] Open
Abstract
Preterm germ-free piglets were monoassociated with probiotic Bifidobacterium animalis subsp. lactis BB-12 (BB12) to verify its safety and to investigate possible protection against subsequent infection with Salmonella Typhimurium strain LT2 (LT2). Clinical signs of salmonellosis, bacterial colonization in the intestine, bacterial translocation to mesenteric lymph nodes (MLN), blood, liver, spleen, and lungs, histopathological changes in the ileum, claudin-1 and occludin mRNA expression in the ileum and colon, intestinal and plasma concentrations of IL-8, TNF-α, and IL-10 were evaluated. Both BB12 and LT2 colonized the intestine of the monoassociated piglets. BB12 did not translocate in the BB12-monoassociated piglets. BB12 was detected in some cases in the MLN of piglets, consequently infected with LT2, but reduced LT2 counts in the ileum and liver of these piglets. LT2 damaged the luminal structure of the ileum, but a previous association with BB12 mildly alleviated these changes. LT2 infection upregulated claudin-1 mRNA in the ileum and colon and downregulated occludin mRNA in the colon. Infection with LT2 increased levels of IL-8, TNF-α, and IL-10 in the intestine and plasma, and BB12 mildly downregulated them compared to LT2 alone. Despite reductions in bacterial translocation and inflammatory cytokines, clinical signs of LT2 infection were not significantly affected by the probiotic BB12. Thus, we hypothesize that multistrain bacterial colonization of preterm gnotobiotic piglets may be needed to enhance the protective effect against the infection with S. Typhimurium LT2.
Collapse
Affiliation(s)
- Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.S.)
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA;
| | - Helena Tlaskalova-Hogenova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic;
| | - Zbynek Stranak
- Department of Neonatology, Institute for the Care of Mother and Child, 147 00 Prague, Czech Republic;
| | - Zdislava Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.S.)
| | - Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.S.)
- Correspondence: ; Tel.: +420-491-418-539
| |
Collapse
|
231
|
Dhouib R, Vagenas D, Hong Y, Verderosa AD, Martin JL, Heras B, Totsika M. Antivirulence DsbA inhibitors attenuate Salmonella enterica serovar Typhimurium fitness without detectable resistance. FASEB Bioadv 2021; 3:231-242. [PMID: 33842848 PMCID: PMC8019255 DOI: 10.1096/fba.2020-00100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 11/15/2022] Open
Abstract
Inhibition of the DiSulfide Bond (DSB) oxidative protein folding machinery, a major facilitator of virulence in Gram‐negative bacteria, represents a promising antivirulence strategy. We previously developed small molecule inhibitors of DsbA from Escherichia coli K‐12 (EcDsbA) and showed that they attenuate virulence of Gram‐negative pathogens by directly inhibiting multiple diverse DsbA homologues. Here we tested the evolutionary robustness of DsbA inhibitors as antivirulence antimicrobials against Salmonella enterica serovar Typhimurium under pathophysiological conditions in vitro. We show that phenylthiophene DsbA inhibitors slow S. Typhimurium growth in minimal media, phenocopying S. Typhimurium isogenic dsbA null mutants. Through passaging experiments, we found that DsbA inhibitor resistance was not induced under conditions that rapidly induced resistance to ciprofloxacin, an antibiotic commonly used to treat Salmonella infections. Furthermore, no mutations were identified in the dsbA gene of inhibitor‐treated S. Typhimurium, and S. Typhimurium virulence remained susceptible to DsbA inhibitors. Our work demonstrates that under in vitro pathophysiological conditions, DsbA inhibitors can have both antivirulence and antibiotic action. Importantly, our finding that DsbA inhibitors appear to be evolutionarily robust offers promise for their further development as next‐generation antimicrobials against Gram‐negative pathogens.
Collapse
Affiliation(s)
- Rabeb Dhouib
- Institute of Health and Biomedical Innovation School of Biomedical Sciences Queensland University of Technology Herston QLD Australia.,Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology Herston QLD Australia
| | - Dimitrios Vagenas
- Institute of Health and Biomedical Innovation School of Biomedical Sciences Queensland University of Technology Herston QLD Australia
| | - Yaoqin Hong
- Institute of Health and Biomedical Innovation School of Biomedical Sciences Queensland University of Technology Herston QLD Australia.,Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology Herston QLD Australia
| | - Anthony D Verderosa
- Institute of Health and Biomedical Innovation School of Biomedical Sciences Queensland University of Technology Herston QLD Australia.,Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology Herston QLD Australia
| | - Jennifer L Martin
- Griffith Institute for Drug Discovery Griffith University Nathan QLD Australia.,University of Wollongong Wollongong NSW Australia
| | - Begoña Heras
- La Trobe Institute for Molecular Science La Trobe University Bundoora VIC Australia
| | - Makrina Totsika
- Institute of Health and Biomedical Innovation School of Biomedical Sciences Queensland University of Technology Herston QLD Australia.,Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology Herston QLD Australia
| |
Collapse
|
232
|
Zrelovs N, Kurbatska V, Rudevica Z, Leonchiks A, Fridmanis D. Sorting out the Superbugs: Potential of Sortase A Inhibitors among Other Antimicrobial Strategies to Tackle the Problem of Antibiotic Resistance. Antibiotics (Basel) 2021; 10:164. [PMID: 33562778 PMCID: PMC7916047 DOI: 10.3390/antibiotics10020164] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/22/2022] Open
Abstract
Rapid spread of antibiotic resistance throughout the kingdom bacteria is inevitably bringing humanity towards the "post-antibiotic" era. The emergence of so-called "superbugs"-pathogen strains that develop resistance to multiple conventional antibiotics-is urging researchers around the globe to work on the development or perfecting of alternative means of tackling the pathogenic bacteria infections. Although various conceptually different approaches are being considered, each comes with its advantages and drawbacks. While drug-resistant pathogens are undoubtedly represented by both Gram(+) and Gram(-) bacteria, possible target spectrum across the proposed alternative approaches of tackling them is variable. Numerous anti-virulence strategies aimed at reducing the pathogenicity of target bacteria rather than eliminating them are being considered among such alternative approaches. Sortase A (SrtA) is a membrane-associated cysteine protease that catalyzes a cell wall sorting reaction by which surface proteins, including virulence factors, are anchored to the bacterial cell wall of Gram(+) bacteria. Although SrtA inhibition seems perspective among the Gram-positive pathogen-targeted antivirulence strategies, it still remains less popular than other alternatives. A decrease in virulence due to inactivation of SrtA activity has been extensively studied in Staphylococcus aureus, but it has also been demonstrated in other Gram(+) species. In this manuscript, results of past studies on the discovery of novel SrtA inhibitory compounds and evaluation of their potency were summarized and commented on. Here, we discussed the rationale behind the inhibition of SrtA, raised some concerns on the comparability of the results from different studies, and touched upon the possible resistance mechanisms as a response to implementation of such therapy in practice. The goal of this article is to encourage further studies of SrtA inhibitory compounds.
Collapse
Affiliation(s)
| | | | | | | | - Davids Fridmanis
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k1, LV-1067 Riga, Latvia; (N.Z.); (V.K.); (Z.R.); (A.L.)
| |
Collapse
|
233
|
Goli A, Khademi-Zare H, Tavakkoli-Moghaddam R, Sadeghieh A, Sasanian M, Malekalipour Kordestanizadeh R. An integrated approach based on artificial intelligence and novel meta-heuristic algorithms to predict demand for dairy products: a case study. NETWORK (BRISTOL, ENGLAND) 2021; 32:1-35. [PMID: 33390063 DOI: 10.1080/0954898x.2020.1849841] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 12/20/2019] [Accepted: 11/05/2020] [Indexed: 05/20/2023]
Abstract
This research specifically addresses the prediction of dairy product demand (DPD). Since dairy products have a short consumption period, it is important to have accurate information about their future demand. The main contribution of this research is to provide an integrated framework based on statistical tests, time-series neural networks, and improved MLP, ANFIS, and SVR with novel meta-heuristic algorithms in order to obtain the best prediction of DPD in Iran. At first, a series of economic and social indicators that seemed to be effective in the demand for dairy products is identified. Then, the ineffective indices are eliminated by using the Pearson correlation coefficient, and statistically significant variables are determined. Since the regression relation is not able to predict this demand properly, the artificial intelligence tools including MLP, ANFIS, and SVR are implemented and improved with the help of novel meta-heuristic algorithms such as grey wolf optimization (GWO), invasive weed optimization (IWO), cultural algorithm (CA), and particle swarm optimization (PSO). The designed hybrid method is used to predict the DPD in Iran by using data from 2013 to 2017. The high accurate results confirm that the proposed hybrid methods have the ability to improve the prediction of the demand for various products.
Collapse
Affiliation(s)
- Alireza Goli
- Department of Industrial Engineering, Yazd University, Saffayieh, Iran
| | | | | | - Ahmad Sadeghieh
- Department of Industrial Engineering, Yazd University, Saffayieh, Iran
| | - Mazyar Sasanian
- Department of Industrial Engineering, Mazandaran University of Science and Technology, Mazandaran, Iran
| | | |
Collapse
|
234
|
Pires DP, Monteiro R, Mil-Homens D, Fialho A, Lu TK, Azeredo J. Designing P. aeruginosa synthetic phages with reduced genomes. Sci Rep 2021; 11:2164. [PMID: 33495501 PMCID: PMC7835345 DOI: 10.1038/s41598-021-81580-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/29/2020] [Indexed: 12/18/2022] Open
Abstract
In the era where antibiotic resistance is considered one of the major worldwide concerns, bacteriophages have emerged as a promising therapeutic approach to deal with this problem. Genetically engineered bacteriophages can enable enhanced anti-bacterial functionalities, but require cloning additional genes into the phage genomes, which might be challenging due to the DNA encapsulation capacity of a phage. To tackle this issue, we designed and assembled for the first time synthetic phages with smaller genomes by knocking out up to 48% of the genes encoding hypothetical proteins from the genome of the newly isolated Pseudomonas aeruginosa phage vB_PaeP_PE3. The antibacterial efficacy of the wild-type and the synthetic phages was assessed in vitro as well as in vivo using a Galleria mellonella infection model. Overall, both in vitro and in vivo studies revealed that the knock-outs made in phage genome do not impair the antibacterial properties of the synthetic phages, indicating that this could be a good strategy to clear space from phage genomes in order to enable the introduction of other genes of interest that can potentiate the future treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Diana P Pires
- CEB - Centre of Biological Engineering, Universidade Do Minho, Campus de Gualtar, Braga, Portugal.
| | - Rodrigo Monteiro
- CEB - Centre of Biological Engineering, Universidade Do Minho, Campus de Gualtar, Braga, Portugal
| | - Dalila Mil-Homens
- Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Lisboa, Portugal
| | - Arsénio Fialho
- Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Lisboa, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Timothy K Lu
- Department of Electrical Engineering and Computer Science and Department of Biological Engineering, Synthetic Biology Center, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Joana Azeredo
- CEB - Centre of Biological Engineering, Universidade Do Minho, Campus de Gualtar, Braga, Portugal.
| |
Collapse
|
235
|
Nale JY, Vinner GK, Lopez VC, Thanki AM, Phothaworn P, Thiennimitr P, Garcia A, AbuOun M, Anjum MF, Korbsrisate S, Galyov EE, Malik DJ, Clokie MRJ. An Optimized Bacteriophage Cocktail Can Effectively Control Salmonella in vitro and in Galleria mellonella. Front Microbiol 2021; 11:609955. [PMID: 33552020 PMCID: PMC7858669 DOI: 10.3389/fmicb.2020.609955] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
Salmonella spp. is a leading cause of gastrointestinal enteritis in humans where it is largely contracted via contaminated poultry and pork. Phages can be used to control Salmonella infection in the animals, which could break the cycle of infection before the products are accessible for consumption. Here, the potential of 21 myoviruses and a siphovirus to eliminate Salmonella in vitro and in vivo was examined with the aim of developing a biocontrol strategy to curtail the infection in poultry and swine. Together, the phages targeted the twenty-three poultry and ten swine prevalent Salmonella serotype isolates tested. Although individual phages significantly reduced bacterial growth of representative isolates within 6 h post-infection, bacterial regrowth occurred 1 h later, indicating proliferation of resistant strains. To curtail bacteriophage resistance, a novel three-phage cocktail was developed in vitro, and further investigated in an optimized Galleria mellonella larva Salmonella infection model colonized with representative swine, chicken and laboratory strains. For all the strains examined, G. mellonella larvae given phages 2 h prior to bacterial exposure (prophylactic regimen) survived and Salmonella was undetectable 24 h post-phage treatment and throughout the experimental time (72 h). Administering phages with bacteria (co-infection), or 2 h post-bacterial exposure (remedial regimen) also improved survival (73-100% and 15-88%, respectively), but was less effective than prophylaxis application. These pre-livestock data support the future application of this cocktail for further development to effectively treat Salmonella infection in poultry and pigs. Future work will focus on cocktail formulation to ensure stability and incorporation into feeds and used to treat the infection in target animals.
Collapse
Affiliation(s)
- Janet Y Nale
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Gurinder K Vinner
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Viviana C Lopez
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Anisha M Thanki
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Preeda Phothaworn
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parameth Thiennimitr
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Angela Garcia
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Manal AbuOun
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Muna F Anjum
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Sunee Korbsrisate
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Edouard E Galyov
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Danish J Malik
- Department of Chemical Engineering, Loughborough University, Loughborough, United Kingdom
| | - Martha R J Clokie
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
236
|
Atterbury RJ, Tyson J. Predatory bacteria as living antibiotics - where are we now? MICROBIOLOGY-SGM 2021; 167. [PMID: 33465024 DOI: 10.1099/mic.0.001025] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Antimicrobial resistance (AMR) is a global health and economic crisis. With too few antibiotics in development to meet current and anticipated needs, there is a critical need for new therapies to treat Gram-negative infections. One potential approach is the use of living predatory bacteria, such as Bdellovibrio bacteriovorus (small Gram-negative bacteria that naturally invade and kill Gram-negative pathogens of humans, animals and plants). Moving toward the use of Bdellovibrio as a 'living antibiotic' demands the investigation and characterization of these bacterial predators in biologically relevant systems. We review the fundamental science supporting the feasibility of predatory bacteria as alternatives to antibiotics.
Collapse
Affiliation(s)
- Robert J Atterbury
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| | - Jess Tyson
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| |
Collapse
|
237
|
Cecchini ME, Paoloni C, Campra N, Picco N, Grosso MC, Soriano Perez ML, Alustiza F, Cariddi N, Bellingeri R. Nanoemulsion of Minthostachys verticillata essential oil. In-vitro evaluation of its antibacterial activity. Heliyon 2021; 7:e05896. [PMID: 33521347 PMCID: PMC7820482 DOI: 10.1016/j.heliyon.2021.e05896] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/28/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
Infectious diseases constitute a problem of great importance for animal and human health, as well as the increasing bacterial resistance to antibiotics. In this context, medicinal plants emerge as an effective alternative to replace the use antibiotics. The essential oil (EO) of Minthostachys verticillata (Griseb.) Epling (Lamiaceae) has demonstrated a strong antimicrobial activity. However, its instability and hydrophobicity under normal storage conditions are limitations to its use. Nanoemulsion technology is an excellent way to solubilize, microencapsulate, and protect this compound. This study aimed to obtain a nanoemulsion based on M. verticillata EO and evaluate its antibacterial activity against Staphylococcus aureus. The EO was obtained by steam distillation. Identification and quantification of their components were determined by GC-MS revealing that the dominated chemical group was oxygenated monoterpenes. Nanoemulsions (NE) were characterized by measuring pH, transmittance, separation percentage, release profile, and morphology. The effect of NE on the growth of S. aureus and cyto-compatibility was also evaluated. The results showed that NE containing a higher percentage of tween 20 exhibited higher stability with an approximated droplet size of 10 nm. The effect of encapsulation process was evaluated by GC-MS revealing that the volatile components in EO were no affected. After 24 h, 74.24 ± 0.75% of EO was released from NE and the antibacterial activity of EO was enhanced considerably by its encapsulation. The incubation of S. aureus with the NE and pure EO, show a bacterial growth inhibition of 58.87% ± 0.99 and 46.72% ± 3.32 (p < 0.05), respectively. In addition, nanoemulsión did not cause toxicity to porcine and equine red blood cells. The results obtained showed that NE could be a potential vehicle for M. verticillata EO with promissory properties to emerge as a tool for developing advanced therapies to control and combat infections.
Collapse
Affiliation(s)
- M E Cecchini
- Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Laboratorio de Inmunología, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina.,Instituto de Biotecnología Ambiental y Salud (INBIAS) CONICET, Universidad Nacional de Río Cuarto, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - C Paoloni
- Universidad Nacional de Río Cuarto, Facultad de Agronomía y Veterinaria, Departamento de Anatomía Animal, Laboratorio de Biotecnología Animal, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - N Campra
- Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Laboratorio de Inmunología, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina.,Instituto de Biotecnología Ambiental y Salud (INBIAS) CONICET, Universidad Nacional de Río Cuarto, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - N Picco
- Universidad Nacional de Río Cuarto, Facultad de Agronomía y Veterinaria, Departamento de Anatomía Animal, Laboratorio de Biotecnología Animal, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - M C Grosso
- Universidad Nacional de Río Cuarto, Facultad de Agronomía y Veterinaria, Departamento de Anatomía Animal, Laboratorio de Biotecnología Animal, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - M L Soriano Perez
- Instituto Nacional de Tecnología Agropecuaria (INTA), Estación Experimental Marcos Juárez, Marcos Juárez, X2580, Córdoba, Argentina
| | - F Alustiza
- Instituto Nacional de Tecnología Agropecuaria (INTA), Estación Experimental Marcos Juárez, Marcos Juárez, X2580, Córdoba, Argentina
| | - N Cariddi
- Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Laboratorio de Inmunología, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina.,Instituto de Biotecnología Ambiental y Salud (INBIAS) CONICET, Universidad Nacional de Río Cuarto, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| | - R Bellingeri
- Universidad Nacional de Río Cuarto, Facultad de Agronomía y Veterinaria, Departamento de Anatomía Animal, Laboratorio de Biotecnología Animal, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina.,Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA) CONICET, Universidad Nacional de Río Cuarto, Ruta 36 Km 601, Río Cuarto, Córdoba, 5800, Argentina
| |
Collapse
|
238
|
Kowalczyk P, Szymczak M, Maciejewska M, Laskowski Ł, Laskowska M, Ostaszewski R, Skiba G, Franiak-Pietryga I. All That Glitters Is Not Silver-A New Look at Microbiological and Medical Applications of Silver Nanoparticles. Int J Mol Sci 2021; 22:E854. [PMID: 33467032 PMCID: PMC7830466 DOI: 10.3390/ijms22020854] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/23/2022] Open
Abstract
Silver and its nanoparticles (AgNPs) have different faces, providing different applications. In recent years, the number of positive nanosilver applications has increased substantially. It has been proven that AgNPs inhibit the growth and survival of bacteria, including human and animal pathogens, as well as fungi, protozoa and arthropods. Silver nanoparticles are known from their antiviral and anti-cancer properties; however, they are also very popular in medical and pharmaceutical nanoengineering as carriers for precise delivery of therapeutic compounds, in the diagnostics of different diseases and in optics and chemistry, where they act as sensors, conductors and substrates for various syntheses. The activity of AgNPs has not been fully discovered; therefore, we need interdisciplinary research to fulfil this knowledge. New forms of products with silver will certainly find application in the future treatment of many complicated and difficult to treat diseases. There is still a lack of appropriate and precise legal condition regarding the circulation of nanomaterials and the rules governing their safety use. The relatively low toxicity, relative biocompatibility and selectivity of nanoparticle interaction combined with the unusual biological properties allow their use in animal production as well as in bioengineering and medicine. Despite a quite big knowledge on this topic, there is still a need to organize the data on AgNPs in relation to specific microorganisms such as bacteria, viruses or fungi. We decided to put this knowledge together and try to show positive and negative effects on prokaryotic and eukaryotic cells.
Collapse
Affiliation(s)
- Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Mateusz Szymczak
- Department of Molecular Virology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland;
| | - Magdalena Maciejewska
- Institute of Polymer and Dye Technology, Lodz University of Technology, Stefanowskiego 12/16, 90-924 Łódź, Poland;
| | - Łukasz Laskowski
- Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Krakow, Poland; (Ł.L.); (M.L.)
| | - Magdalena Laskowska
- Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Krakow, Poland; (Ł.L.); (M.L.)
| | | | - Grzegorz Skiba
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Ida Franiak-Pietryga
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Dr., La Jolla, CA 92037, USA
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, 251 Pomorska Str., 92-213 Łódź, Poland
| |
Collapse
|
239
|
Investigating the mechanism of action of aggregation-inducing antimicrobial Pept-ins. Cell Chem Biol 2021; 28:524-536.e4. [PMID: 33434517 DOI: 10.1016/j.chembiol.2020.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/20/2020] [Accepted: 12/16/2020] [Indexed: 12/24/2022]
Abstract
Aggregation can be selectively induced by aggregation-prone regions (APRs) contained in the target proteins. Aggregation-inducing antimicrobial peptides (Pept-ins) contain sequences homologous to APRs of target proteins and exert their bactericidal effect by causing aggregation of a large number of proteins. To better understand the mechanism of action of Pept-ins and the resistance mechanisms, we analyzed the phenotypic, lipidomic, and transcriptomic as well as genotypic changes in laboratory-derived Pept-in-resistant E. coli mutator cells. The analysis showed that the Pept-in resistance mechanism is dominated by a decreased Pept-in uptake, in both laboratory-derived mutator cells and clinical isolates. Our data indicate that Pept-in uptake involves an electrostatic attraction between the Pept-in and the bacterial membrane and follows a complex mechanism potentially involving many transporters. Furthermore, it seems more challenging for bacteria to become resistant toward Pept-ins that are less dependent on electrostatic attraction for uptake, suggesting that future Pept-ins should be selected for this property.
Collapse
|
240
|
Xu B, Fu J, Zhu L, Li Z, Jin M, Wang Y. Overall assessment of antibiotic substitutes for pigs: a set of meta-analyses. J Anim Sci Biotechnol 2021; 12:3. [PMID: 33413687 PMCID: PMC7792336 DOI: 10.1186/s40104-020-00534-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 12/01/2020] [Indexed: 01/28/2023] Open
Abstract
Background Antibiotic growth promoters are widely used to improve weight gain. However, the abuse of antibiotics can have many negative effects on people. Developing alternatives to antibiotics is an urgent need in livestock production. We aimed to perform a meta-analysis and network meta-analysis (NMA) to investigate the effects of feed additives as potential antibiotic substitutes (ASs) on bacteriostasis, growth performance, intestinal morphology and immunity. Furthermore, the primary, secondary, and tertiary ASs were defined by comparing their results with the results of antibiotics. Results Among 16,309 identified studies, 37 were summarized to study the bacteriostasis effects of feed additives, and 89 were included in the meta-analysis and NMA (10,228 pigs). We summarized 268 associations of 57 interventions with 32 bacteria. The order of bacteriostasis effects was as follows: antimicrobial peptides (AMPs) ≈ antibiotics>organic acids>plant extracts>oligosaccharides. We detected associations of 11 feed additives and 11 outcomes. Compared with a basal diet, plant extract, AMPs, probiotics, microelements, organic acids, bacteriophages, lysozyme, zymin, and oligosaccharides significantly improved growth performance (P < 0.05); organic acids, probiotics, microelements, lysozyme, and AMPs remarkably increased the villus height:crypt depth ratio (V/C) (P < 0.05); and plant extracts, zymin, microelements, probiotics, and organic acids notably improved immunity (P < 0.05). The optimal AMP, bacteriophage, lysozyme, microelements, oligosaccharides, organic acids, plants, plant extracts, probiotics, and zymin doses were 0.100%, 0.150%, 0.012%, 0.010%, 0.050%, 0.750%, 0.20%, 0.040%, 0.180%, and 0.100%, respectively. Compared with antibiotics, all investigated feed additives exhibited no significant difference in effects on growth performance, IgG, and diarrhoea index/rate (P > 0.05); AMPs and microelements significantly increased V/C (P < 0.05); and zymin significantly improved lymphocyte levels (P < 0.05). Furthermore, linear weighting sum models were used to comprehensively estimate the overall impact of each feed additive on pig growth and health. Conclusions Our findings suggest that AMPs and plant extracts can be used as primary ASs for weaned piglets and growing pigs, respectively. Bacteriophages, zymin, plants, probiotics, oligosaccharides, lysozyme, and microelements can be regarded as secondary ASs. Nucleotides and organic acids can be considered as tertiary ASs. Future studies should further assess the alternative effects of combinational feed additives.
Collapse
Affiliation(s)
- Bocheng Xu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Jie Fu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Luoyi Zhu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Zhi Li
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Mingliang Jin
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Yizhen Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Nutrition and Feed of Ministry of Agriculture, Key Laboratory of Animal Nutrition and Feed Science of Zhejiang Province, Institute of Feed Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China.
| |
Collapse
|
241
|
Rebucci R, Comi M, Ghiringhelli M, Giorgi S, Cheli F, Bontempo V. Lauric acid saponified with calcium ameliorates indices of intestinal function and gut health in weaned piglets. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1944338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Raffaella Rebucci
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, University of Milan, Milan, Italy
| | - Marcello Comi
- Dipartimento di Scienze Umane e di Promozione della Qualita’ della Vita, University San Raffaele Roma, Roma, Italy
| | - Matteo Ghiringhelli
- Sohnis Research laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Silvia Giorgi
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, University of Milan, Milan, Italy
| | - Federica Cheli
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, University of Milan, Milan, Italy
- CRC I-WE (Coordinating Research Centre: Innovation for Well-Being and Environment), University of Milan, Milan, Italy
| | - Valentino Bontempo
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, University of Milan, Milan, Italy
| |
Collapse
|
242
|
Kustov AV, Krestyaninov MA, Kruchin SO, Shukhto OV, Kustova TV, Belykh DV, Khudyaeva IS, Koifman MO, Razgovorov PB, Berezin DB. Interaction of cationic chlorin photosensitizers with non-ionic surfactant Tween 80. MENDELEEV COMMUNICATIONS 2021. [DOI: 10.1016/j.mencom.2021.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
243
|
|
244
|
Holbein BE, Ang MTC, Allan DS, Chen W, Lehmann C. Iron-withdrawing anti-infectives for new host-directed therapies based on iron dependence, the Achilles' heel of antibiotic-resistant microbes. ENVIRONMENTAL CHEMISTRY LETTERS 2021; 19:2789-2808. [PMID: 33907538 PMCID: PMC8062846 DOI: 10.1007/s10311-021-01242-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/13/2021] [Indexed: 05/02/2023]
Abstract
The iron dependence of antibiotic-resistant microbes represents an Achilles' heel that can be exploited broadly. The growing global problem of antibiotic resistance of microbial pathogens wherein microbes become resistant to the very antibiotics used against them during infection is linked not only to our health uses but also to agribusiness practices and the changing environment. Here we review mechanisms of microbial iron acquisition and host iron withdrawal defense, and the influence of iron withdrawal on the antimicrobial activity of antibiotics. Antibiotic-resistant microbes are unaltered in their iron requirements, but iron withdrawal from microbes enhances the activities of various antibiotics and importantly suppresses outgrowth of antibiotic-exposed resistant microbial survivors. Of the three therapeutic approaches available to exploit microbial iron susceptibility, including (1) use of gallium as a non-functional iron analogue, (2) Trojan horse conjugates of microbial siderophores carrying antibiotics, and (3) new generation iron chelators, purposely designed as anti-microbials, the latter offers various advantages. For instance, these novel anti-microbial chelators overcome the limitations of conventional clinically-used hematological chelators which display host toxicity and are not useful antimicrobials. 3-Hydroxypyridin-4-one-containing polymeric chelators appear to have the highest potential. DIBI (developmental code name) is a well-developed lead candidate, being a low molecular weight, water-soluble copolymer with enhanced iron binding characteristics, strong anti-microbial and anti-inflammatory activities, low toxicity for animals and demonstrated freedom from microbial resistance development. DIBI has been shown to enhance antibiotic efficacy for antibiotic-resistant microbes during infection, and it also prevents recovery growth and resistance development during microbe exposure to various antibiotics. Because DIBI bolsters innate iron withdrawal defenses of the infected host, it has potential to provide a host-directed anti-infective therapy.
Collapse
Affiliation(s)
- Bruce E. Holbein
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
- Department of Microbiology and Immunology, Dalhousie University, 5859 College St., Halifax, NS B3H 1X5 Canada
| | - M. Trisha C. Ang
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
| | - David S. Allan
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6 Canada
| | - Christian Lehmann
- Department of Microbiology and Immunology, Dalhousie University, 5859 College St., Halifax, NS B3H 1X5 Canada
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
245
|
Liu L, Zhao L, Liu L, Yue S, Wang J, Cao Z. Influence of Different Aromatic Hydrophobic Residues on the Antimicrobial Activity and Membrane Selectivity of BRBR-NH 2 Tetrapeptide. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:15331-15342. [PMID: 33295774 DOI: 10.1021/acs.langmuir.0c02777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The ultrashort linear antimicrobial tetrapeptide BRBR-NH2 with an unnatural residue biphenylalanine (B) has potent and rapid antimethicillin-resistant Staphylococcus aureus (MRSA) activity but lacks hemolytic activity. The anti-MRSA activity of BRBR-NH2 is 8-fold more potent than that of WRWR-NH2 and 16-fold more potent than that of FRFR-NH2. However, how to influence their antimicrobial activities and mechanisms through the substitution of different aromatic hydrophobic residues is still unclear. In this work, to study the effects of varying hydrophobic interactions and membrane selectivities of BRBR-NH2, we performed multiple long-time (1000 ns) molecular dynamics (MD) simulations to investigate the interactions of a red blood cell (RBC) membrane and a Gram-positive bacterial cell membrane with three different tetrapeptides (BRBR-NH2, WRWR-NH2, and FRFR-NH2) under different ratios of peptides and lipids and also explored the changes in the membrane and structural characteristics of peptides. The binding energy results show that BRBR-NH2 interacts weakly with the RBC membrane, while not all BRBR-NH2 can be adsorbed to the RBC membrane surface. The MD simulation results produced significant local membrane thinning of multiBRBR-NH2 peptides in the Gram-positive bacterial cell membrane. An in-depth analysis of structural features and peptide-membrane interactions suggests that the aggregation of BRBR-NH2 on the membrane surface plays a crucial role in the destruction of the cell membrane. Taken together with the observed local membrane thinning, the in-depth analysis demonstrated that the interactions between the lipid bilayer and the BRBR-NH2 aggregation surface result in a local disturbance of the membrane structure. It can be concluded that the high anti-MRSA activity of BRBR-NH2 is attributed to the aggregation of BRBR-NH2 on the membrane surface. On the other hand, WRWR-NH2 and FRFR-NH2 peptides tend to bind with the membrane surface in a monomeric form and cover the membrane surface in a carpet-like manner. Therefore, these results provide an advanced microscopic understanding of how hydrophobic interactions or hydrophobic residues affect the antimicrobial activity and mechanism of antimicrobial peptides (AMPs).
Collapse
Affiliation(s)
- Lei Liu
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| | - Liling Zhao
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| | - Lixia Liu
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| | - Shizhong Yue
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| | - Jihua Wang
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| | - Zanxia Cao
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China
| |
Collapse
|
246
|
Improved Stability and Activity of a Marine Peptide-N6NH2 against Edwardsiella tarda and Its Preliminary Application in Fish. Mar Drugs 2020; 18:md18120650. [PMID: 33348729 PMCID: PMC7766155 DOI: 10.3390/md18120650] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/28/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Edwardsiella tarda can cause fatal gastro-/extraintestinal diseases in fish and humans. Overuse of antibiotics has led to antibiotic resistance and contamination in the environment, which highlights the need to find new antimicrobial agents. In this study, the marine peptide-N6 was amidated at its C-terminus to generate N6NH2. The antibacterial activity of N6 and N6NH2 against E. tarda was evaluated in vitro and in vivo; their stability, toxicity and mode of action were also determined. Minimal inhibitory concentrations (MICs) of N6 and N6NH2 against E. tarda were 1.29–3.2 μM. Both N6 and N6NH2 killed bacteria by destroying the cell membrane of E. tarda and binding to lipopolysaccharide (LPS) and genomic DNA. In contrast with N6, N6NH2 improved the stability toward trypsin, reduced hemolysis (by 0.19% at a concentration of 256 μg/mL) and enhanced the ability to penetrate the bacterial outer and inner membrane. In the model of fish peritonitis caused by E. tarda, superior to norfloxacin, N6NH2 improved the survival rate of fish, reduced the bacterial load on the organs, alleviated the organ injury and regulated the immunity of the liver and kidney. These data suggest that the marine peptide N6NH2 may be a candidate for novel antimicrobial agents against E. tarda infections.
Collapse
|
247
|
Gan BH, Cai X, Javor S, Köhler T, Reymond JL. Synergistic Effect of Propidium Iodide and Small Molecule Antibiotics with the Antimicrobial Peptide Dendrimer G3KL against Gram-Negative Bacteria. Molecules 2020; 25:E5643. [PMID: 33266085 PMCID: PMC7730455 DOI: 10.3390/molecules25235643] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/22/2022] Open
Abstract
There is an urgent need to develop new antibiotics against multidrug-resistant bacteria. Many antimicrobial peptides (AMPs) are active against such bacteria and often act by destabilizing membranes, a mechanism that can also be used to permeabilize bacteria to other antibiotics, resulting in synergistic effects. We recently showed that G3KL, an AMP with a multibranched dendritic topology of the peptide chain, permeabilizes the inner and outer membranes of Gram-negative bacteria including multidrug-resistant strains, leading to efficient bacterial killing. Here, we show that permeabilization of the outer and inner membranes of Pseudomonas aeruginosa by G3KL, initially detected using the DNA-binding fluorogenic dye propidium iodide (PI), also leads to a synergistic effect between G3KL and PI in this bacterium. We also identify a synergistic effect between G3KL and six different antibiotics against the Gram-negative Klebsiella pneumoniae, against which G3KL is inactive.
Collapse
Affiliation(s)
- Bee-Ha Gan
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland; (B.-H.G.); (X.C.); (S.J.)
| | - Xingguang Cai
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland; (B.-H.G.); (X.C.); (S.J.)
| | - Sacha Javor
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland; (B.-H.G.); (X.C.); (S.J.)
| | - Thilo Köhler
- Department of Microbiology and Molecular Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Service of Infectious Diseases, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Jean-Louis Reymond
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland; (B.-H.G.); (X.C.); (S.J.)
| |
Collapse
|
248
|
Manzo G, Hind CK, Ferguson PM, Amison RT, Hodgson-Casson AC, Ciazynska KA, Weller BJ, Clarke M, Lam C, Man RCH, Shaughnessy BGO, Clifford M, Bui TT, Drake AF, Atkinson RA, Lam JKW, Pitchford SC, Page CP, Phoenix DA, Lorenz CD, Sutton JM, Mason AJ. A pleurocidin analogue with greater conformational flexibility, enhanced antimicrobial potency and in vivo therapeutic efficacy. Commun Biol 2020; 3:697. [PMID: 33247193 PMCID: PMC7699649 DOI: 10.1038/s42003-020-01420-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 10/22/2020] [Indexed: 01/08/2023] Open
Abstract
Antimicrobial peptides (AMPs) are a potential alternative to classical antibiotics that are yet to achieve a therapeutic breakthrough for treatment of systemic infections. The antibacterial potency of pleurocidin, an AMP from Winter Flounder, is linked to its ability to cross bacterial plasma membranes and seek intracellular targets while also causing membrane damage. Here we describe modification strategies that generate pleurocidin analogues with substantially improved, broad spectrum, antibacterial properties, which are effective in murine models of bacterial lung infection. Increasing peptide-lipid intermolecular hydrogen bonding capabilities enhances conformational flexibility, associated with membrane translocation, but also membrane damage and potency, most notably against Gram-positive bacteria. This negates their ability to metabolically adapt to the AMP threat. An analogue comprising D-amino acids was well tolerated at an intravenous dose of 15 mg/kg and similarly effective as vancomycin in reducing EMRSA-15 lung CFU. This highlights the therapeutic potential of systemically delivered, bactericidal AMPs.
Collapse
Affiliation(s)
- Giorgia Manzo
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Charlotte K Hind
- Technology Development Group, National Infection Service, Public Health England, Salisbury, UK
| | - Philip M Ferguson
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Richard T Amison
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
- Sackler Institute of Pulmonary Pharmacology, King's College London, London, UK
| | - Alice C Hodgson-Casson
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Katarzyna A Ciazynska
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Bethany J Weller
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Maria Clarke
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Carolyn Lam
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Rico C H Man
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Blaze G O' Shaughnessy
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
- Sackler Institute of Pulmonary Pharmacology, King's College London, London, UK
| | - Melanie Clifford
- Technology Development Group, National Infection Service, Public Health England, Salisbury, UK
| | - Tam T Bui
- Centre for Biomolecular Spectroscopy and Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, London, SE1 1UL, UK
| | - Alex F Drake
- Centre for Biomolecular Spectroscopy and Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, London, SE1 1UL, UK
| | - R Andrew Atkinson
- Centre for Biomolecular Spectroscopy and Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, London, SE1 1UL, UK
| | - Jenny K W Lam
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Simon C Pitchford
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
- Sackler Institute of Pulmonary Pharmacology, King's College London, London, UK
| | - Clive P Page
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
- Sackler Institute of Pulmonary Pharmacology, King's College London, London, UK
| | - David A Phoenix
- School of Applied Science, London South Bank University, 103 Borough Road, London, SE1 0AA, UK
| | | | - J Mark Sutton
- Technology Development Group, National Infection Service, Public Health England, Salisbury, UK.
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
249
|
Zheng L, Li S, Luo J, Wang X. Latest Advances on Bacterial Cellulose-Based Antibacterial Materials as Wound Dressings. Front Bioeng Biotechnol 2020; 8:593768. [PMID: 33330424 PMCID: PMC7732461 DOI: 10.3389/fbioe.2020.593768] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/27/2020] [Indexed: 12/17/2022] Open
Abstract
At present, there are various wound dressings that can protect the wound from further injury or isolate the external environment in wound treatment. Whereas, infection and slow self-healing still exist in wound healing process. Therefore, it is urgent to develop an ideal wound dressing with good biocompatibility and strong antibacterial activity to promote wound healing. Bacterial cellulose is a kind of promising biopolymer because it can control wound exudate and provide a moist environment for wound healing. However, the lack of antibacterial activity limits its application. In this paper, the advantages of bacterial cellulose as wound dressings were introduced, and the preparation and research progress of bacterial cellulose-based antibacterial composites in recent years were reviewed, including adding antibiotics, combining with inorganic antibacterial agents or organic antibacterial agents. Finally, the existing problems and future development direction of bacterial cellulose-based antibacterial wound dressings were discussed.
Collapse
Affiliation(s)
- Lu Zheng
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, China
| | - Shanshan Li
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, China
| | - Jiwen Luo
- Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, School of Chemistry and Environment, South China Normal University, Guangzhou, China
| | - Xiaoying Wang
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
250
|
Davies JE, Behroozian S. An ancient solution to a modern problem. Mol Microbiol 2020; 113:546-549. [PMID: 32185831 DOI: 10.1111/mmi.14481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 11/30/2022]
Abstract
The dearth of new antibiotics and escalating emergence of multidrug resistant bacteria have created a global healthcare crisis and highlight the drastic need for novel antimicrobial agents. Complementary and alternative strategies including the investigation of ancient medicinals could address this problem. Natural clay minerals with a long history of medicinal and biomedical applications have become an interest due to their broad-spectrum antimicrobial activity. Such untapped natural sources may provide new therapeutic agents in the battle against infectious diseases in the post-antibiotic era.
Collapse
Affiliation(s)
- Julian E Davies
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Shekooh Behroozian
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|