1
|
Hao J, Yang R, Ma X. Combatting Acinetobacter baumannii pneumonia by multidrug codelivery of curcumin nanosuspensions and sulfamethoxazole with polymyxin B for targeted pediatric pulmonary inflammation and disease management. Microb Pathog 2025; 203:107454. [PMID: 40086739 DOI: 10.1016/j.micpath.2025.107454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 02/10/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The bacterium Acinetobacter baumannii (A. baumannii) poses a severe clinical challenge due to its high drug resistance, driven by genetic adaptability, biofilm formation, enzymatic degradation, and facilitating healthcare-associated infections. In our present study, we described and to developed an innovative strategy for A. baumannii pneumonia using multidrug codelivery of curcumin nanosuspensions and sulfamethoxazole (SZ) with polymyxin B (PB) (CNS@SZ/PB). The physicochemical properties, drug release kinetics, antibacterial efficacy, and anti-biofilm activity of CNS@SZ/PB were comprehensively evaluated. In-vitro anti-bacterial analysis of CNS@SZ/PB exhibited potent bacterial inhibition against A. baumannii with a minimum inhibitory concentration (MIC) of 60 μg/mL, it also effectively prevented biofilm formation at 50 μg/mL. Furthermore, the pediatric pulmonary targeting ability of CNS@SZ/PB demonstrated effective moderation of lung inflammation, offering a promising way for managing pulmonary-associated diseases. In-vivo studies on mice infected with multidrug-resistant A. baumannii validated the significant protective effects of CNS@SZ/PB, attributed to its regulation of inflammatory factors. Overall, our findings highlight the therapeutic potential of CNS@SZ/PB as a promising approach for combating A. baumannii infections, providing a solid foundation for further experimental analysis of pediatric pulmonary inflammation and disease.
Collapse
Affiliation(s)
- Jingjing Hao
- Department of Neonatal, Yan'an University Affiliated Hospital, Yan'an, 716000, China
| | - Runhong Yang
- Department of Imaging, Yan'an University Affiliated Hospital, Yan'an, 716000, China
| | - Xiaxia Ma
- Department of Neonatal, Yan'an University Affiliated Hospital, Yan'an, 716000, China.
| |
Collapse
|
2
|
Fang Y, Huang C, Jang T, Lin S, Wang J, Huang Y, Tsai MH. Pharmacokinetic study of polymyxin B in healthy subjects and subjects with renal insufficiency. Clin Transl Sci 2024; 17:e70110. [PMID: 39673151 PMCID: PMC11645446 DOI: 10.1111/cts.70110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/18/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024] Open
Abstract
Polymyxin B is a viable option for treating antibiotic-resistant infections; however, current data on its pharmacokinetics, particularly in patients with renal insufficiency, remain inconclusive and necessitates further investigation. To address this gap, we conducted an open-label, single-center, single-dose, parallel-group pharmacokinetic study. Participants received an intravenous dose of 0.75 mg/kg of polymyxin B and were categorized based on their renal function: those with normal function (creatinine clearance [CLcr] ≥ 90 mL/min), mild renal insufficiency (CLcr 60-89 mL/min), and end-stage kidney disease patients on intermittent hemodialysis (IHD) (CLcr < 10 mL/min). The pharmacokinetic parameters assessed included the area under the curve (AUC), maximum concentration (Cmax), clearance rate (CL), volume of distribution (Vz), and half-life (t1/2). Results indicated that subjects with mild renal insufficiency exhibited pharmacokinetic profiles similar to healthy individuals. Nevertheless, in patients undergoing long-term IHD, we observed significant differences: the AUC was 58% higher, Cmax was 29% lower, CL was 42% lower, Vz was 60% larger, and t1/2 was extended by 10 h compared to healthy controls. Secondary outcomes revealed good tolerability of polymyxin B across all groups, with no serious adverse effects related to renal function. In summary, while kidney function may have a slight impact on the pharmacokinetic of polymyxin B, it does not compromise the drug's therapeutic effectiveness.
Collapse
Affiliation(s)
- Yu‐Wei Fang
- Division of Nephrology, Department of MedicineShin Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Department of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Chien‐Hsien Huang
- Department of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan
- Division of Infectious Disease, Department of Internal MedicineShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | - Tsrang‐Neng Jang
- Department of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan
- Department of Internal MedicineShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | - Shih‐Sen Lin
- Division of Chest Medicine, Department of Internal MedicineShin Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | - Jing‐Tong Wang
- Division of Nephrology, Department of MedicineShin Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | - Yen‐Ta Huang
- Department of Surgery, College of MedicineNational Cheng Kung University Hospital, National Cheng Kung UniversityTainan CityTaiwan
| | - Ming Hsien Tsai
- Division of Nephrology, Department of MedicineShin Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Department of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan
| |
Collapse
|
3
|
Salvaterra Pasquini JP, Queiroz PA, Rodrigues do Amaral PH, da Silva TC, de Souza Bonfim Mendonça P, Vandresen F, Carvalho Ceolis JP, de Lima Scodro RB, Caleffi-Ferracioli KR, Cardoso RF, Dias Siqueira VL. Polymyxin B adjuvants against polymyxin B- and carbapenem-resistant Gram-negative bacteria. Future Microbiol 2024; 19:1445-1454. [PMID: 39258398 PMCID: PMC11492663 DOI: 10.1080/17460913.2024.2398312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Aim: Polymyxin B (PMB) is one of the few therapeutic options for treating infections caused by carbapenem-resistant Gram-negative bacteria (CR-GNB). However, the emergence of PMB-resistant CR-GNB strains has prompted the exploration of antibiotic adjuvants as potential therapeutic avenues. Thus, this study evaluates the potential of 3,5-dinitrobenzoic acid derivatives (DNH01, DNH11, DNH13 and DNH20) and isoniazid-N-acylhydrazones (INZ1-7, INZ9 and INZ11) as adjuvants to enhance PMB efficacy against CR-GNB.Materials & methods: MIC, MBC and drug combination assays were conducted using multidrug-resistant clinical isolates of Enterobacterales and Acinetobacter baumannii. In addition, the effects of PMB and PMB + DNH derivatives were assessed through flow cytometry and scanning electron microscopy (SEM).Results: DNH01, DNH11 and DNH20, unlike the INH-acylhydrazones, significantly restored PMB activity (MIC ≤ 2 μg/ml) in 80% of the tested isolates. Flow cytometry and SEM assays confirmed that DNH derivatives rescued the activity of PMB, yielding results comparable to those expected for PMB alone but at 256-fold lower concentrations.Conclusion: These findings suggest DNH derivatives hold substantial promise as PMB adjuvants to combat PMB-resistant CR-GNB infections.
Collapse
Affiliation(s)
| | - Paula Assis Queiroz
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringa, Maringa, Parana, Brazil
| | | | - Thalita Camilo da Silva
- Department of Clinical Analysis & Biomedicine, State University of Maringa, Maringa, Parana, Brazil
| | | | - Fábio Vandresen
- Department of Chemistry, Federal Technological University of Parana, Londrina, Parana, Brazil
| | | | - Regiane Bertin de Lima Scodro
- Postgraduate Program in Health Sciences, State University of Maringa, Parana, Brazil
- Department of Clinical Analysis & Biomedicine, State University of Maringa, Maringa, Parana, Brazil
| | - Katiany Rizzieri Caleffi-Ferracioli
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringa, Maringa, Parana, Brazil
- Department of Clinical Analysis & Biomedicine, State University of Maringa, Maringa, Parana, Brazil
| | - Rosilene Fressatti Cardoso
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringa, Maringa, Parana, Brazil
- Department of Clinical Analysis & Biomedicine, State University of Maringa, Maringa, Parana, Brazil
| | - Vera Lúcia Dias Siqueira
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringa, Maringa, Parana, Brazil
- Department of Clinical Analysis & Biomedicine, State University of Maringa, Maringa, Parana, Brazil
| |
Collapse
|
4
|
Marchant P, Vivanco E, Silva A, Nevermann J, Fuentes I, Barrera B, Otero C, Calderón IL, Gil F, Fuentes JA. β-lactam-induced OMV release promotes polymyxin tolerance in Salmonella enterica sv. Typhi. Front Microbiol 2024; 15:1389663. [PMID: 38591031 PMCID: PMC10999688 DOI: 10.3389/fmicb.2024.1389663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
The rise of multidrug-resistant bacteria is a global concern, leading to a renewed reliance on older antibiotics like polymyxins as a last resort. Polymyxins, cationic cyclic peptides synthesized nonribosomally, feature a hydrophobic acyl tail and positively charged residues. Their antimicrobial mechanism involves initial interaction with Gram-negative bacterial outer-membrane components through polar and hydrophobic interactions. Outer membrane vesicles (OMVs), nano-sized proteoliposomes secreted from the outer membrane of Gram-negative bacteria, play a crucial role in tolerating harmful molecules, including cationic peptides such as polymyxins. Existing literature has documented environmental changes' impact on modulating OMV properties in Salmonella Typhimurium. However, less information exists regarding OMV production and characteristics in Salmonella Typhi. A previous study in our laboratory showed that S. Typhi ΔmrcB, a mutant associated with penicillin-binding protein (PBP, a β-lactam antibiotic target), exhibited hypervesiculation. Consequently, this study investigated the potential impact of β-lactam antibiotics on promoting polymyxin tolerance via OMVs in S. Typhi. Our results demonstrated that sub-lethal doses of β-lactams increased bacterial survival against polymyxin B in S. Typhi. This phenomenon stems from β-lactam antibiotics inducing hypervesiculation of OMVs with higher affinity for polymyxin B, capturing and diminishing its biologically effective concentration. These findings suggest that β-lactam antibiotic use may inadvertently contribute to decreased polymyxin effectivity against S. Typhi or other Gram-negative bacteria, complicating the effective treatment of infections caused by these pathogens. This study emphasizes the importance of evaluating the influence of β-lactam antibiotics on the interaction between OMVs and other antimicrobial agents.
Collapse
Affiliation(s)
- Pedro Marchant
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Erika Vivanco
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Andrés Silva
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Jan Nevermann
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Ignacio Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Boris Barrera
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Santiago, Chile
| | - Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Iván L. Calderón
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Fernando Gil
- Microbiota-Host Interactions and Clostridia Research Group, Universidad Andres Bello, Santiago, Chile
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
5
|
Lobertti CA, Gizzi FO, Magni C, Rial A, Chabalgoity JA, Yim L, Blancato VS, Asquith CRM, García Véscovi E. Enhancing colistin efficacy against Salmonella infections with a quinazoline-based dual therapeutic strategy. Sci Rep 2024; 14:5148. [PMID: 38429351 PMCID: PMC10907601 DOI: 10.1038/s41598-024-55793-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Colistin remains one of the last-resort therapies for combating infections caused by multidrug-resistant (MDR) Enterobacterales, despite its adverse nephro- and neuro-toxic effects. This study elucidates the mechanism of action of a non-antibiotic 4-anilinoquinazoline-based compound that synergistically enhances the effectiveness of colistin against Salmonella enterica. The quinazoline sensitizes Salmonella by deactivating intrinsic, mutational, and transferable resistance mechanisms that enable Salmonella to counteract the antibiotic impact colistin, together with an induced disruption to the electrochemical balance of the bacterial membrane. The attenuation of colistin resistance via the combined treatment approach also proves efficacious against E. coli, Klebsiella, and Acinetobacter strains. The dual therapy reduces the mortality of Galleria mellonella larvae undergoing a systemic Salmonella infection when compared to individual drug treatments. Overall, our findings unveil the potential of the quinazoline-colistin combined therapy as an innovative strategy against MDR bacteria.
Collapse
Affiliation(s)
- Carlos A Lobertti
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Predio CCT-CONICET Rosario, S2000, Santa Fe, Rosario, Argentina
| | - Fernán O Gizzi
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Predio CCT-CONICET Rosario, S2000, Santa Fe, Rosario, Argentina
| | - Christian Magni
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Predio CCT-CONICET Rosario, S2000, Santa Fe, Rosario, Argentina
| | - Analía Rial
- Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Universidad de La República, Avda. Alfredo Navarro 3051, 11600, Montevideo, Uruguay
| | - José A Chabalgoity
- Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Universidad de La República, Avda. Alfredo Navarro 3051, 11600, Montevideo, Uruguay
| | - Lucía Yim
- Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Universidad de La República, Avda. Alfredo Navarro 3051, 11600, Montevideo, Uruguay
| | - Víctor S Blancato
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Predio CCT-CONICET Rosario, S2000, Santa Fe, Rosario, Argentina
| | - Christopher R M Asquith
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Eleonora García Véscovi
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Predio CCT-CONICET Rosario, S2000, Santa Fe, Rosario, Argentina.
| |
Collapse
|
6
|
Wang J, Dong X, Wang F, Jiang J, Zhao Y, Gu J, Xu J, Mao X, Tu B. Molecular Characteristics and Genetic Analysis of Extensively Drug-Resistant Isolates with different Tn3 Mobile Genetic Elements. Curr Microbiol 2023; 80:246. [PMID: 37335402 DOI: 10.1007/s00284-023-03340-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/22/2023] [Indexed: 06/21/2023]
Abstract
Extensively drug-resistant (XDR) bacteria are the main caues for causing clinical infectious diseases. Our aim was to distinguish the present molecular epidemiological situation of XDR Klebsiella pneumoniae, Acinetobacter baumannii, and Escherichia coli isolates recovered from local hospitals in Changzhou. Antibiotic susceptibility and phenotypic analysis, multilocus sequence typing and Pulsed Field Gel Electrophoresis were performed to trace these isolates. Resistant phenotype and gene analysis from 29 XDR strains demonstrated that they mainly included TEM, CTX-M-1/2, OXA-48, and KPC products. A. baumannii strains belonged to sequence type (ST) ST224, and carrying the blaCTX-M-2/TEM gene. The quinolone genes aac(6')-ib-cr and qnrB were carrying only in A. baumannii and E.coli. Three (2.3%) of these strains were found to contain the blaNDM-1 or blaNDM-5 gene. A new genotype of K. pneumoniae was found as ST2639. Epidemic characteristics of the XDR clones showed that antibiotic resistance genes distributed unevenly in different wards in Changzhou's local hospitals. With the sequencing of blaNDM carrying isolates, the plasmids often carrying a highly conservative Tn3-relavent mobile genetic element. The especially coupled insert sequence ISKox3 may be a distinctive resistance gene transfer loci. The genotypic diversity variation of XDRs suggested that tracking and isolating the sources of antibiotic resistance especially MBL-encoding genes such as blaNDM-will help manage the risk of infection by these XDRs.
Collapse
Affiliation(s)
- Jiazhen Wang
- School of Public Health, Xuzhou Medical University, Xuzhou, 221004, China
| | - Xin Dong
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Fengming Wang
- School of Public Health, Xuzhou Medical University, Xuzhou, 221004, China
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Jinyi Jiang
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Ying Zhao
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Jingyue Gu
- School of Public Health, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jian Xu
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Xujian Mao
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Bowen Tu
- School of Public Health, Xuzhou Medical University, Xuzhou, 221004, China.
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| |
Collapse
|
7
|
Hanafin PO, Abdul Rahim N, Sharma R, Cess CG, Finley SD, Bergen PJ, Velkov T, Li J, Rao GG. Proof-of-concept for incorporating mechanistic insights from multi-omics analyses of polymyxin B in combination with chloramphenicol against Klebsiella pneumoniae. CPT Pharmacometrics Syst Pharmacol 2023; 12:387-400. [PMID: 36661181 PMCID: PMC10014067 DOI: 10.1002/psp4.12923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/21/2022] [Accepted: 12/30/2022] [Indexed: 01/21/2023] Open
Abstract
Carbapenemase-resistant Klebsiella pneumoniae (KP) resistant to multiple antibiotic classes necessitates optimized combination therapy. Our objective is to build a workflow leveraging omics and bacterial count data to identify antibiotic mechanisms that can be used to design and optimize combination regimens. For pharmacodynamic (PD) analysis, previously published static time-kill studies (J Antimicrob Chemother 70, 2015, 2589) were used with polymyxin B (PMB) and chloramphenicol (CHL) mono and combination therapy against three KP clinical isolates over 24 h. A mechanism-based model (MBM) was developed using time-kill data in S-ADAPT describing PMB-CHL PD activity against each isolate. Previously published results of PMB (1 mg/L continuous infusion) and CHL (Cmax : 8 mg/L; bolus q6h) mono and combination regimens were evaluated using an in vitro one-compartment dynamic infection model against a KP clinical isolate (108 CFU/ml inoculum) over 24 h to obtain bacterial samples for multi-omics analyses. The differentially expressed genes and metabolites in these bacterial samples served as input to develop a partial least squares regression (PLSR) in R that links PD responses with the multi-omics responses via a multi-omics pathway analysis. PMB efficacy was increased when combined with CHL, and the MBM described the observed PD well for all strains. The PLSR consisted of 29 omics inputs and predicted MBM PD response (R2 = 0.946). Our analysis found that CHL downregulated metabolites and genes pertinent to lipid A, hence limiting the emergence of PMB resistance. Our workflow linked insights from analysis of multi-omics data with MBM to identify biological mechanisms explaining observed PD activity in combination therapy.
Collapse
Affiliation(s)
- Patrick O Hanafin
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Rajnikant Sharma
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Colin G Cess
- Department of Biomedical Engineering Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Department of Biomedical Engineering Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Phillip J Bergen
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Tony Velkov
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jian Li
- Department of Microbiology, Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences and Biomedicine Discovery Institute, Monash University, Parkville, Victoria, Australia
| | - Gauri G Rao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Bray AS, Smith RD, Hudson AW, Hernandez GE, Young TM, George HE, Ernst RK, Zafar MA. MgrB-Dependent Colistin Resistance in Klebsiella pneumoniae Is Associated with an Increase in Host-to-Host Transmission. mBio 2022; 13:e0359521. [PMID: 35311534 PMCID: PMC9040857 DOI: 10.1128/mbio.03595-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
Due to its high transmissibility, Klebsiella pneumoniae is one of the leading causes of nosocomial infections. Here, we studied the biological cost of colistin resistance, an antibiotic of last resort, in this opportunistic pathogen using a murine model of gut colonization and transmission. Colistin resistance in K. pneumoniae is commonly the result of the inactivation of the small regulatory protein MgrB. Without a functional MgrB, the two-component system PhoPQ is constitutively active, leading to an increase in lipid A modifications and subsequent colistin resistance. Using an isogenic mgrB deletion mutant (MgrB-), we demonstrate that the mutant's colistin resistance is not associated with a fitness defect under in vitro growth conditions. However, in our murine model of K. pneumoniae gastrointestinal (GI) colonization, the MgrB- colonizes the gut poorly, allowing us to identify a fitness cost. Moreover, the MgrB- mutant has higher survival outside the host compared with the parental strain. We attribute this enhanced survivability to dysregulation of the PhoPQ two-component system and accumulation of the master stress regulator RpoS. The enhanced survival of MgrB- may be critical for its rapid host-to-host transmission observed in our model. Together, our data using multiple clinical isolates demonstrate that MgrB-dependent colistin resistance in K. pneumoniae comes with a biological cost in gut colonization. However, this cost is mitigated by enhanced survival outside the host and consequently increases its host-to-host transmission. Additionally, it underscores the importance of considering the entire life cycle of a pathogen to determine the actual biological cost associated with antibiotic resistance. IMPORTANCE The biological cost associated with colistin resistance in Klebsiella pneumoniae was examined using a murine model of K. pneumoniae gut colonization and fecal-oral transmission. A common mutation resulting in colistin resistance in K. pneumoniae is a loss-of-function mutation of the small regulatory protein MgrB that regulates the two-component system PhoPQ. Even though colistin resistance in K. pneumoniae comes with a fitness defect in gut colonization, it increases bacterial survival outside the host enabling it to transmit more effectively to a new host. The enhanced survival is dependent upon the accumulation of RpoS and dysregulation of the PhoPQ. Hence, our study expands our understanding of the underlying molecular mechanism contributing to the transmission of colistin-resistant K. pneumoniae.
Collapse
Affiliation(s)
- Andrew S. Bray
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Richard D. Smith
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, Baltimore, Maryland, USA
- Department of Pathology, University of Maryland, Baltimore, Baltimore, Maryland, USA
| | - Andrew W. Hudson
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Giovanna E. Hernandez
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Taylor M. Young
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | | | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, Baltimore, Maryland, USA
| | - M. Ammar Zafar
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
9
|
Cui X, Lü Y, Yue C. Development and Research Progress of Anti-Drug Resistant Bacteria Drugs. Infect Drug Resist 2022; 14:5575-5593. [PMID: 34992385 PMCID: PMC8711564 DOI: 10.2147/idr.s338987] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/12/2021] [Indexed: 01/10/2023] Open
Abstract
Bacterial resistance has become increasingly serious because of the widespread use and abuse of antibiotics. In particular, the emergence of multidrug-resistant bacteria has posed a serious threat to human public health and attracted the attention of the World Health Organization (WHO) and the governments of various countries. Therefore, the establishment of measures against bacterial resistance and the discovery of new antibacterial drugs are increasingly urgent to better contain the emergence of bacterial resistance and provide a reference for the development of new antibacterial drugs. In this review, we discuss some antibiotic drugs that have been approved for clinical use and a partial summary of the meaningful research results of anti-drug resistant bacterial drugs in different fields, including the antibiotic drugs approved by the FDA from 2015 to 2020, the potential drugs against drug-resistant bacteria, the new molecules synthesized by chemical modification, combination therapy, drug repurposing, immunotherapy and other therapies.
Collapse
Affiliation(s)
- Xiangyi Cui
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an, 716000, Shaanxi, People's Republic of China
| | - Yuhong Lü
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an, 716000, Shaanxi, People's Republic of China.,Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources, Yan'an University, Yan'an, 716000, Shaanxi, People's Republic of China
| | - Changwu Yue
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an, 716000, Shaanxi, People's Republic of China.,Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources, Yan'an University, Yan'an, 716000, Shaanxi, People's Republic of China
| |
Collapse
|
10
|
Marchant P, Carreño A, Vivanco E, Silva A, Nevermann J, Otero C, Araya E, Gil F, Calderón IL, Fuentes JA. "One for All": Functional Transfer of OMV-Mediated Polymyxin B Resistance From Salmonella enterica sv. Typhi Δ tolR and Δ degS to Susceptible Bacteria. Front Microbiol 2021; 12:672467. [PMID: 34025627 PMCID: PMC8131662 DOI: 10.3389/fmicb.2021.672467] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
The appearance of multi-resistant strains has contributed to reintroducing polymyxin as the last-line therapy. Although polymyxin resistance is based on bacterial envelope changes, other resistance mechanisms are being reported. Outer membrane vesicles (OMVs) are nanosized proteoliposomes secreted from the outer membrane of Gram-negative bacteria. In some bacteria, OMVs have shown to provide resistance to diverse antimicrobial agents either by sequestering and/or expelling the harmful agent from the bacterial envelope. Nevertheless, the participation of OMVs in polymyxin resistance has not yet been explored in S. Typhi, and neither OMVs derived from hypervesiculating mutants. In this work, we explored whether OMVs produced by the hypervesiculating strains Salmonella Typhi ΔrfaE (LPS synthesis), ΔtolR (bacterial envelope) and ΔdegS (misfolded proteins and σ E activation) exhibit protective properties against polymyxin B. We found that the OMVs extracted from S. Typhi ΔtolR and ΔdegS protect S. Typhi WT from polymyxin B in a concentration-depending manner. By contrast, the protective effect exerted by OMVs from S. Typhi WT and S. Typhi ΔrfaE is much lower. This effect is achieved by the sequestration of polymyxin B, as assessed by the more positive Zeta potential of OMVs with polymyxin B and the diminished antibiotic's availability when coincubated with OMVs. We also found that S. Typhi ΔtolR exhibited an increased MIC of polymyxin B. Finally, we determined that S. Typhi ΔtolR and S. Typhi ΔdegS, at a lesser level, can functionally and transiently transfer the OMV-mediated polymyxin B resistance to susceptible bacteria in cocultures. This work shows that mutants in genes related to OMVs biogenesis can release vesicles with improved abilities to protect bacteria against membrane-active agents. Since mutations affecting OMV biogenesis can involve the bacterial envelope, mutants with increased resistance to membrane-acting agents that, in turn, produce protective OMVs with a high vesiculation rate (e.g., S. Typhi ΔtolR) can arise. Such mutants can functionally transfer the resistance to surrounding bacteria via OMVs, diminishing the effective concentration of the antimicrobial agent and potentially favoring the selection of spontaneous resistant strains in the environment. This phenomenon might be considered the source for the emergence of polymyxin resistance in an entire bacterial community.
Collapse
Affiliation(s)
- Pedro Marchant
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Alexander Carreño
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Eduardo Vivanco
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Andrés Silva
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Jan Nevermann
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Eyleen Araya
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Santiago, Chile
| | - Fernando Gil
- Microbiota-Host Interactions and Clostridia Research Group, Universidad Andres Bello, Santiago, Chile.,ANID-Millennium Science Initiative Program-Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
| | - Iván L Calderón
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Juan A Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
11
|
Antimicrobial Stewardship and Its Impact on the Changing Epidemiology of Polymyxin Use in a South Indian Healthcare Setting. Antibiotics (Basel) 2021; 10:antibiotics10050470. [PMID: 33918994 PMCID: PMC8142974 DOI: 10.3390/antibiotics10050470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/22/2021] [Indexed: 11/17/2022] Open
Abstract
Polymyxins being last resort drugs to treat infections triggered by multidrug-resistant pathogens necessitates the implementation of antimicrobial stewardship program (ASP) initiatives to support its rational prescription across healthcare settings. Our study aims to describe the change in the epidemiology of polymyxins and patient outcomes following the implementation of ASP at our institution. The antimicrobial stewardship program initiated in February 2016 at our 1300 bed tertiary care center involved post-prescriptive audits tracking polymyxin consumption and evaluating prescription appropriateness in terms of the right indication, right frequency, right drug, right duration of therapy and administration of the right loading dose (LD) and maintenance dose (MD). Among the 2442 polymyxin prescriptions tracked over the entire study period ranging from February 2016 to January 2020, the number of prescriptions dropped from 772 prescriptions in the pre-implementation period to an average of 417 per year during the post-implementation period, recording a 45% reduction. The quarterly patient survival rates had a significant positive correlation with the quarterly prescription appropriateness rates (r = 0.4774, p = 0.02), right loading dose (r = 0.5228, p = 0.015) and right duration (r = 0.4361, p = 0.04). Our study on the epidemiology of polymyxin use demonstrated favorable effects on the appropriateness of prescriptions and mortality benefits after successful implementation of antimicrobial stewardship in a real-world setting.
Collapse
|
12
|
Sabnis A, Hagart KLH, Klöckner A, Becce M, Evans LE, Furniss RCD, Mavridou DAI, Murphy R, Stevens MM, Davies JC, Larrouy-Maumus GJ, Clarke TB, Edwards AM. Colistin kills bacteria by targeting lipopolysaccharide in the cytoplasmic membrane. eLife 2021; 10:e65836. [PMID: 33821795 PMCID: PMC8096433 DOI: 10.7554/elife.65836] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Colistin is an antibiotic of last resort, but has poor efficacy and resistance is a growing problem. Whilst it is well established that colistin disrupts the bacterial outer membrane (OM) by selectively targeting lipopolysaccharide (LPS), it was unclear how this led to bacterial killing. We discovered that MCR-1 mediated colistin resistance in Escherichia coli is due to modified LPS at the cytoplasmic rather than OM. In doing so, we also demonstrated that colistin exerts bactericidal activity by targeting LPS in the cytoplasmic membrane (CM). We then exploited this information to devise a new therapeutic approach. Using the LPS transport inhibitor murepavadin, we were able to cause LPS accumulation in the CM of Pseudomonas aeruginosa, which resulted in increased susceptibility to colistin in vitro and improved treatment efficacy in vivo. These findings reveal new insight into the mechanism by which colistin kills bacteria, providing the foundations for novel approaches to enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Akshay Sabnis
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
| | - Katheryn LH Hagart
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
| | - Anna Klöckner
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
- Department of Bioengineering, Imperial College LondonLondonUnited Kingdom
- Department of Materials, Imperial College LondonLondonUnited Kingdom
- Institute of Biomedical Engineering, Imperial College LondonLondonUnited Kingdom
| | - Michele Becce
- Department of Bioengineering, Imperial College LondonLondonUnited Kingdom
- Department of Materials, Imperial College LondonLondonUnited Kingdom
- Institute of Biomedical Engineering, Imperial College LondonLondonUnited Kingdom
| | - Lindsay E Evans
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
- Department of Chemistry, Imperial College London, Molecular Sciences Research HubLondonUnited Kingdom
| | - R Christopher D Furniss
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
| | - Despoina AI Mavridou
- Department of Molecular Biosciences, University of Texas at AustinAustinUnited States
| | - Ronan Murphy
- National Heart and Lung Institute, Imperial College LondonLondonUnited Kingdom
- Department of Paediatric Respiratory Medicine, Royal Brompton HospitalLondonUnited Kingdom
| | - Molly M Stevens
- Department of Bioengineering, Imperial College LondonLondonUnited Kingdom
- Department of Materials, Imperial College LondonLondonUnited Kingdom
- Institute of Biomedical Engineering, Imperial College LondonLondonUnited Kingdom
| | - Jane C Davies
- National Heart and Lung Institute, Imperial College LondonLondonUnited Kingdom
- Department of Paediatric Respiratory Medicine, Royal Brompton HospitalLondonUnited Kingdom
| | - Gérald J Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
| | - Thomas B Clarke
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
| | - Andrew M Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
13
|
Chen W, Liu H, Wang Q, Wang X, Kong X, Wang X, Zhang X, Zhan Q, Li P. Estimation of the area under concentration-time curve of polymyxin B based on limited sampling concentrations in Chinese patients with severe pneumonia. Eur J Clin Pharmacol 2020; 77:95-105. [PMID: 32875388 DOI: 10.1007/s00228-020-02986-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023]
Abstract
AIMS The efficacy and toxicity of polymyxin B (PB) are closely related to its pharmacokinetic/pharmacodynamic (PK/PD) index area under the concentration-time curve (AUC) to minimum inhibitory concentration (MIC) ratio. The purpose of this study was to obtain PK data for PB in Chinese severe pneumonia patients and establish appropriate blood sampling time points for the PB therapeutic drug monitoring (TDM). SUBJECT AND METHOD After treatment with at least four doses of PB (50 IU, q12h), the blood samples were collected immediately after the end of infusion (C0) and 1.5, 2, 4, 6, 8, and 12 h (C1.5, C2, C4, C6, C8, C12) after PB administration. The PB blood plasma concentrations were determined using an ultra-performance liquid chromatography-tandem mass spectrometer (UPLC-MS/MS). All 42 patients were randomly divided into modeling (n = 24) and validation (n = 18) groups. The relationship between AUCss,24h and PB plasma concentration at each time point in modeling group was analyzed using limited sampling strategy and a PK method based on one-compartment with correction model. RESULTS C6 scheme was found to provide the most accurate prediction of AUCss,24h values (r2 = 0.984) with the target value of 1.9-4.2 μg/ml at steady state to reach the 50-100 μg h/ml criteria of AUCss,24h. C0 with target value of 1.0-2.8 μg/ml can be considered an alternative sampling scheme (r2 = 0.900) but prediction deviation may exist. C0 and Cmax sampling scheme also demonstrated good predicting ability of AUC values using PK model. CONCLUSION This study provides a clear plan for the implementation of TDM of PB, which is useful for optimizing the dosing regimen and individualizing treatment in severe pneumonia patients.
Collapse
Affiliation(s)
- Wenqian Chen
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Huifang Liu
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Qianlin Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiaoxing Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xudong Kong
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiaoxue Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xianglin Zhang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Pengmei Li
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
14
|
Ongwae GM, Morrison KR, Allen RA, Kim S, Im W, Wuest WM, Pires MM. Broadening Activity of Polymyxin by Quaternary Ammonium Grafting. ACS Infect Dis 2020; 6:1427-1435. [PMID: 32212668 DOI: 10.1021/acsinfecdis.0c00037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bacterial pathogens continue to impose a tremendous health burden across the globe. Here, we describe a novel series of polymyxin-based agents grafted with membrane-active quaternary ammonium warheads to combine two important classes of Gram-negative antimicrobial scaffolds. The goal was to deliver a targeted quaternary ammonium warhead onto the surface of bacterial pathogens using the outer membrane homing properties of polymyxin. The most potent agents resulted in new scaffolds that retained the ability to target Gram-negative bacteria and had limited toxicity toward mammalian cells. We showed, using a molecular dynamics approach, that the new agents retained their ability to engage in specific interactions with lipopolysaccharide molecules. Significantly, the combination of quaternary ammonium and polymyxin widens the activity to the pathogen Staphylococcus aureus. Our results serve as an example of how two membrane-active agents can be combined to produce a class of novel scaffolds with potent biological activity.
Collapse
Affiliation(s)
- George M. Ongwae
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Kelly R. Morrison
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Ryan A. Allen
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Seonghoon Kim
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Wonpil Im
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Marcos M. Pires
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
15
|
Brown P, Abdulle O, Boakes S, Duperchy E, Moss S, Simonovic M, Stanway S, Wilson A, Dawson MJ. Direct modifications of the cyclic peptide Polymyxin B leading to analogues with enhanced in vitro antibacterial activity. Bioorg Med Chem Lett 2020; 30:127163. [PMID: 32273214 DOI: 10.1016/j.bmcl.2020.127163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/26/2020] [Accepted: 03/28/2020] [Indexed: 11/30/2022]
Abstract
Synthetic modifications have been made directly to the cyclic peptide core of polymyxin B, enabling the further understanding of structure activity relationships of this antimicrobial peptide. Such modified polymyxins are also substrates for enzymic hydrolysis, enabling the synthesis of a variety of semi-synthetic analogues, resulting in compounds with increased in vitro antibacterial activity.
Collapse
Affiliation(s)
- Pamela Brown
- Cantab Anti-Infectives Ltd, Welwyn Garden City AL7 3AX, UK; Spero Therapeutics, Inc, 675 Massachusetts Avenue, 14th Floor, Cambridge, MA 02139 USA.
| | - Omar Abdulle
- Cantab Anti-Infectives Ltd, Welwyn Garden City AL7 3AX, UK
| | - Steven Boakes
- Cantab Anti-Infectives Ltd, Welwyn Garden City AL7 3AX, UK
| | | | - Stephen Moss
- Eurofins Integrated Discovery UK Ltd, Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, UK
| | - Mona Simonovic
- Cantab Anti-Infectives Ltd, Welwyn Garden City AL7 3AX, UK
| | - Steven Stanway
- Eurofins Integrated Discovery UK Ltd, Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, UK
| | - Antoinette Wilson
- Eurofins Integrated Discovery UK Ltd, Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, UK
| | - Michael J Dawson
- Cantab Anti-Infectives Ltd, Welwyn Garden City AL7 3AX, UK; Spero Therapeutics, Inc, 675 Massachusetts Avenue, 14th Floor, Cambridge, MA 02139 USA
| |
Collapse
|
16
|
Kathayat D, Helmy YA, Deblais L, Rajashekara G. Novel small molecules affecting cell membrane as potential therapeutics for avian pathogenic Escherichia coli. Sci Rep 2018; 8:15329. [PMID: 30333507 PMCID: PMC6193035 DOI: 10.1038/s41598-018-33587-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/26/2018] [Indexed: 11/09/2022] Open
Abstract
Avian pathogenic Escherichia coli (APEC), a most common bacterial pathogen of poultry, causes multiple extra-intestinal diseases in poultry which results in significant economic losses to the poultry industry worldwide. In addition, APEC are a subgroup of extra-intestinal pathogenic E. coli (ExPEC), and APEC contaminated poultry products are a potential source of foodborne ExPEC infections to humans and transfer of antimicrobial resistant genes. The emergence of multi-drug resistant APEC strains and the limited efficacy of vaccines necessitate novel APEC control approaches. Here, we screened a small molecule (SM) library and identified 11 SMs bactericidal to APEC. The identified SMs were effective against multiple APEC serotypes, biofilm embedded APEC, antimicrobials resistant APECs, and other pathogenic E. coli strains. Microscopy revealed that these SMs affect the APEC cell membrane. Exposure of SMs to APEC revealed no resistance. Most SMs showed low toxicity towards chicken and human cells and reduced the intracellular APEC load. Treatment with most SMs extended the wax moth larval survival and reduced the intra-larval APEC load. Our studies could facilitate the development of antimicrobial therapeutics for the effective management of APEC infections in poultry as well as other E. coli related foodborne zoonosis, including APEC related ExPEC infections in humans.
Collapse
Affiliation(s)
- Dipak Kathayat
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA
| | - Yosra A Helmy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA
| | - Loic Deblais
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| |
Collapse
|
17
|
Craft KM, Gaddy JA, Townsend SD. Human Milk Oligosaccharides (HMOs) Sensitize Group B Streptococcus to Clindamycin, Erythromycin, Gentamicin, and Minocycline on a Strain Specific Basis. ACS Chem Biol 2018; 13:2020-2026. [PMID: 30071726 DOI: 10.1021/acschembio.8b00661] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human milk oligosaccharides (HMOs) possess antimicrobial and antibiofilm activity against Group B Streptococcus (GBS). HMOs were screened for their ability to potentiate antibiotic activity. We observed that HMOs potentiate the function of aminoglycosides, lincosamides, macrolides, and tetracyclines on a strain specific basis but not β-lactams or glycopeptides that inhibit cell wall synthesis. These findings are notable as GBS has evolved high levels of resistance toward aminoglycosides, macrolides, and tetracyclines. Finally, HMOs potentiate the function of aminoglycosides against both Staphylococcus aureus and Acinetobacter baumannii. On the basis of these observations, we hypothesized that HMOs act by increasing membrane permeability. This hypothesis was evaluated using a bacterial membrane permeability assay which revealed that HMOs increase membrane permeability toward propidium iodide.
Collapse
Affiliation(s)
- Kelly M. Craft
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Jennifer A. Gaddy
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, D-3100 Medical Center North, Nashville, Tennessee 37232, United States
- Tennessee Valley Healthcare Systems, Department of Veterans Affairs, 1310 24th Avenue South, Nashville, Tennessee 37212, United States
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, 896 Preston Research Building, Nashville, Tennessee 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Medical Center North A-5302, 1161 21st Ave South, Nashville, Tennessee 37232, United States
| |
Collapse
|
18
|
A Rapid and Semi-Quantitative Gold Nanoparticles Based Strip Sensor for Polymyxin B Sulfate Residues. NANOMATERIALS 2018; 8:nano8030144. [PMID: 29510541 PMCID: PMC5869635 DOI: 10.3390/nano8030144] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 12/02/2022]
Abstract
Increasing attention is now being directed to the utilization of polymyxin B (PMB) as a last-line treatment for life-threatening infections caused by multidrug resistant Gram-negative bacteria. Unfortunately, polymyxins resistance is also increasingly reported, leaving a serious threat to human health. Therefore, the establishment of rapid detection methods for PMB residues is highly essential to ensure public health. In this study, two monoclonal antibodies (mAb; 2A2 and 3C6) were obtained using PMB-bovine serum albumin as the immunogen and PMB-ovalbumin as the coating antigen, which were prepared with N-(γ-maleimidobutyryloxy) succinimide ester and glutaraldehyde as cross-linking agents, respectively. Through an indirect competitive enzyme-linked immunosorbent assay, resultant two mAbs were compared and the results indicated that 3C6 showed higher sensitivity with a half maximum inhibition concentration of 13.13 ng/mL. Based on 3C6, a gold nanoparticles (AuNPs)-based immunochromatographic test (ICT) strip was then established, the mechanism of which is that free PMB competes with the fixed coating antigen to combine with mAb labeled by AuNPs. Using ICT strip to detect milk and animal feed samples revealed the visible detection limits were 25 ng/mL and 500 μg/kg, respectively and the cutoff limits were 100 ng/mL and 1000 μg/kg, respectively. The ICT strip provides results within 15 min, facilitating rapid and semi-quantitative analysis of PMB residues in milk and animal feed.
Collapse
|