1
|
Li W, Wang Y, Gao J, Wang A. Antimicrobial resistance and its risks evaluation in wetlands on the Qinghai-Tibetan Plateau. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116699. [PMID: 38981389 DOI: 10.1016/j.ecoenv.2024.116699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024]
Abstract
Amidst the global antimicrobial resistance (AMR) crisis, antibiotic resistance has permeated even the most remote environments. To understand the dissemination and evolution of AMR in minimally impacted ecosystems, the resistome and mobilome of wetlands across the Qinghai-Tibetan Plateau and its marginal regions were scrutinized using metagenomic sequencing techniques. The composition of wetland microbiomes exhibits significant variability, with dominant phyla including Proteobacteria, Actinobacteria, Bacteroidetes, and Verrucomicrobia. Notably, a substantial abundance of Antibiotic Resistance Genes (ARGs) and Mobile Genetic Elements (MGEs) was detected, encompassing 17 ARG types, 132 ARG subtypes, and 5 types of MGEs (Insertion Sequences, Insertions Sequences, Genomic Islands, Transposons, and Integrative Conjugative Elements). No significant variance was observed in the prevalence of resistome and mobilome across different wetland types (i.e., the Yellow River, other rivers, lakes, and marshes) (R=-0.5882, P=0.607). The co-occurrence of 74 ARG subtypes and 22 MGEs was identified, underscoring the pivotal role of MGEs in shaping ARG pools within the Qinghai-Tibetan Plateau wetlands. Metagenomic binning and analysis of assembled genomes (MAGs) revealed that 93 out of 206 MAGs harbored ARGs (45.15 %). Predominantly, Burkholderiales, Pseudomonadales, and Enterobacterales were identified as the primary hosts of these ARGs, many of which represent novel species. Notably, a substantial proportion of ARG-carrying MAGs also contained MGEs, reaffirming the significance of MGEs in AMR dissemination. Furthermore, utilizing the arg_ranker framework for risk assessment unveiled severe contamination of high-risk ARGs across most plateau wetlands. Moreover, some prevalent human pathogens were identified as potential hosts for these high-risk ARGs, posing substantial transmission risks. This study aims to investigate the prevalence of resistome and mobilome in wetlands, along with evaluating the risk posed by high-risk ARGs. Such insights are crucial for informing environmental protection strategies and facilitating the management of water resources on the Qinghai-Tibetan Plateau.
Collapse
Affiliation(s)
- Weiwei Li
- School of Life Sciences, Ludong University, Yantai, Shandong 264025, China
| | - Yanfang Wang
- School of Life Sciences, Ludong University, Yantai, Shandong 264025, China
| | - Jianxin Gao
- School of Life Sciences, Ludong University, Yantai, Shandong 264025, China
| | - Ailan Wang
- School of Life Sciences, Ludong University, Yantai, Shandong 264025, China.
| |
Collapse
|
2
|
Ying JP, Fu CM, Wu YC, Chen YM, Liu XY, Zhang QL, Liu H, Liang MZ. Combined analysis of transcriptomics and metabolomics provide insights into the antibacterial mechanism of bacteriocin XJS01 against multidrug-resistant Staphylococcus aureus. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 917:170412. [PMID: 38281634 DOI: 10.1016/j.scitotenv.2024.170412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Multidrug-resistant (MDR) bacteria are widespread in the environment and pose a serious threat to public health. It has been shown that bacteriocins have a great potential in controlling MDR pathogens, including Staphylococcus aureus. A previously reported Lactobacillus salivarius bacteriocin XJS01 exhibited good antibacterial activity against MDR S. aureus 2612:1606BL1486 (henceforth referred to as S. aureus_26), but its molecular mechanism remains unknown. Herein, we investigated the antibacterial mechanism of XJS01 on S. aureus_26 using an approach combining transcriptomics and metabolomics. The results showed that XJS01 induced significant changes at both transcriptional and metabolic levels in S. aureus_26. In total, 231 differentially expressed genes (DEGs) and 206 differentially abundance metabolites (DAMs) were identified in S. aureus_26 treated with 1 × MIC (minimum inhibition concentration) XJS01 compared with untreated (XJS01-free) cells (control). Functional analysis revealed that these DEGs and DAMs, alone with the related pathways and biological processes, were typically involved in stress response, being primarily related to metal uptake, cell virulence, self-help mechanism, amino acid and energy metabolism, bacterial stress response (e.g., two-component system), and membrane transport (e.g., phosphotransferase system). Overall, this study uncovered the multi-target effects of bacteriocins against MDR S. aureus at the genome-wide transcriptional and metabolic levels. These findings might be useful in the development of bacteriocins for the control of MDR S. aureus and other drug-resistant bacteria.
Collapse
Affiliation(s)
- Jian-Ping Ying
- Guangxi Key Laboratory of Marine Environmental Change and Disaster in Beibu Gulf, Beibu Gulf University, Guangxi, Qinzhou 535011, China; Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan, Kunming 650500, China
| | - Chao-Min Fu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan, Kunming 650500, China
| | - Yan-Chun Wu
- E.N.T. Department, The People's Hospital of Chuxiong Yi Autonomous Prefecture, Yunnan, Chuxiong 675000, China
| | - Ya-Mei Chen
- E.N.T. Department, The People's Hospital of Chuxiong Yi Autonomous Prefecture, Yunnan, Chuxiong 675000, China
| | - Xiao-Yu Liu
- E.N.T. Department, The People's Hospital of Chuxiong Yi Autonomous Prefecture, Yunnan, Chuxiong 675000, China
| | - Qi-Lin Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Yunnan, Kunming 650500, China.
| | - Hui Liu
- E.N.T. Department, The People's Hospital of Chuxiong Yi Autonomous Prefecture, Yunnan, Chuxiong 675000, China.
| | - Ming-Zhong Liang
- Guangxi Key Laboratory of Marine Environmental Change and Disaster in Beibu Gulf, Beibu Gulf University, Guangxi, Qinzhou 535011, China.
| |
Collapse
|
3
|
Saxena D, Maitra R, Bormon R, Czekanska M, Meiers J, Titz A, Verma S, Chopra S. Tackling the outer membrane: facilitating compound entry into Gram-negative bacterial pathogens. NPJ ANTIMICROBIALS AND RESISTANCE 2023; 1:17. [PMCID: PMC11721184 DOI: 10.1038/s44259-023-00016-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 10/23/2023] [Indexed: 01/17/2025]
Abstract
Emerging resistance to all available antibiotics highlights the need to develop new antibiotics with novel mechanisms of action. Most of the currently used antibiotics target Gram-positive bacteria while Gram-negative bacteria easily bypass the action of most drug molecules because of their unique outer membrane. This additional layer acts as a potent barrier restricting the entry of compounds into the cell. In this scenario, several approaches have been elucidated to increase the accumulation of compounds into Gram-negative bacteria. This review includes a brief description of the physicochemical properties that can aid compounds to enter and accumulate in Gram-negative bacteria and covers different strategies to target or bypass the outer membrane-mediated barrier in Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Deepanshi Saxena
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031 UP India
| | - Rahul Maitra
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031 UP India
| | - Rakhi Bormon
- Department of Chemistry, IIT Kanpur, Kanpur, 208016 UP India
| | - Marta Czekanska
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124 Standort Hannover-Braunschweig, Germany
| | - Joscha Meiers
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124 Standort Hannover-Braunschweig, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124 Standort Hannover-Braunschweig, Germany
| | - Sandeep Verma
- Department of Chemistry, IIT Kanpur, Kanpur, 208016 UP India
- Center for Nanoscience, IIT Kanpur, Kanpur, 208016 UP India
| | - Sidharth Chopra
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031 UP India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| |
Collapse
|
4
|
Thomson P, García P, del Río C, Castro R, Núñez A, Miranda C. Antimicrobial Resistance and Extended-Spectrum Beta-Lactamase Genes in Enterobacterales, Pseudomonas and Acinetobacter Isolates from the Uterus of Healthy Mares. Pathogens 2023; 12:1145. [PMID: 37764953 PMCID: PMC10535638 DOI: 10.3390/pathogens12091145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Antibiotic-resistant bacteria are a growing concern for human and animal health. The objective of this study was to determine the antimicrobial resistance and extended-spectrum beta-lactamase genes in Enterobacterales, Pseudomonas spp. and Acinetobacter spp. isolates from the uterus of healthy mares. For this purpose, 21 mares were swabbed for samples, which were later seeded on blood agar and MacConkey agar. The isolates were identified using MALDI-TOF and the antimicrobial susceptibility test was performed using the Kirby-Bauer technique. To characterize the resistance genes, a polymerase chain reaction (PCR) scheme was performed. Of the isolates identified as Gram-negative, 68.8% were Enterobacterales, represented by E. coli, Enterobacter cloacae, Citrobacter spp., and Klebsiella pneumoniae; 28.1% belonged to the genus Acinetobacter spp.; and 3.1% to Pseudomonas aeruginosa. A 9.3% of the isolates were multidrug-resistant (MDR), presenting resistance to antibiotics from three different classes, while 18.8% presented resistance to two or more classes of different antibiotics. The diversity of three genes that code for ESBL (blaTEM, blaCTX-M and blaSHV) was detected in 12.5% of the strains. The most frequent was blaSHV, while blaTEM and blaCTX-M were present in Citrobacter spp. and Klebsiella pneumoniae. These results are an alarm call for veterinarians and their environment and suggest taking measures to prevent the spread of these microorganisms.
Collapse
Affiliation(s)
- Pamela Thomson
- Laboratorio de Microbiología Clínica y Microbioma, Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370134, Chile;
| | - Patricia García
- Departamento de Laboratorios Clínicos, Escuela de Medicina, Pontificia Universidad Católica, Santiago 8940000, Chile;
| | - Camila del Río
- Laboratorio de Microbiología Clínica y Microbioma, Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370134, Chile;
| | - Rodrigo Castro
- Escuela de Medicina Veterinaria, Facultad de Recursos Naturales y Medicina Veterinaria, Universidad Santo Tomás, Talca 3473620, Chile
| | - Andrea Núñez
- Escuela de Medicina Veterinaria, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Curicó 3340000, Chile
- Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Santiago 7500975, Chile
| | - Carolina Miranda
- Laboratorio de Microbiología Red de Salud UC-CHRISTUS, Pontificia Universidad Católica, Santiago 8940000, Chile;
| |
Collapse
|
5
|
Al Tall Y, Al-Nassar B, Abualhaijaa A, Sabi SH, Almaaytah A. The design and functional characterization of a novel hybrid antimicrobial peptide from Esculentin-1a and melittin. PHARMACIA 2023. [DOI: 10.3897/pharmacia.70.e97116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Antimicrobial agents are one of the most widely used drugs in medicine. In the last fifty years, the misuse of these agents caused the emergence of resistant strains of bacteria that led to an increase in life-threatening infections. The need to develop new agents has become a priority, and antimicrobial peptides attained high consideration. The antimicrobial activities of a novel In-house designed hybrid cationic peptide (BKR1) were studied against different strains of Gram-negative bacteria. This was done using the broth dilution method as outlined by the Clinical and Laboratory Institute (CLSI). Checkerboard assy was employed to investigate the synergistic activity of BKR1 peptide with four antibiotics (Levofloxacin, chloramphenicol, rifampicin, and ampicillin). Finally, the cytotoxicity of BKR1 was evaluated against human blood cells and mammalian kidney cells (Vero cells). BKR1 displayed bactericidal activity against tested strains of Gram-negative bacteria, with zero hemolytic effects. It also acts as a strong adjuvant with levofloxacin, chloramphenicol, and rifampicin against resistant strains of P. aeruginosa and E. coli. This study represents the design and elucidation of the antimicrobial activities of a novel hybrid antimicrobial peptide named (BKR1). Our results indicate thar BKR1 is a promising candidate to treat resistant infectious diseases individually or as an adjuvant with conventional antibiotics.
Collapse
|
6
|
Stephen J, Salam F, Lekshmi M, Kumar SH, Varela MF. The Major Facilitator Superfamily and Antimicrobial Resistance Efflux Pumps of the ESKAPEE Pathogen Staphylococcus aureus. Antibiotics (Basel) 2023; 12:343. [PMID: 36830254 PMCID: PMC9952236 DOI: 10.3390/antibiotics12020343] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
The ESKAPEE bacterial pathogen Staphylococcus aureus has posed a serious public health concern for centuries. Throughout its evolutionary course, S. aureus has developed strains with resistance to antimicrobial agents. The bacterial pathogen has acquired multidrug resistance, causing, in many cases, untreatable infectious diseases and raising serious public safety and healthcare concerns. Amongst the various mechanisms for antimicrobial resistance, integral membrane proteins that serve as secondary active transporters from the major facilitator superfamily constitute a chief system of multidrug resistance. These MFS transporters actively export structurally different antimicrobial agents from the cells of S. aureus. This review article discusses the S. aureus-specific MFS multidrug efflux pump systems from a molecular mechanistic perspective, paying particular attention to structure-function relationships, modulation of antimicrobial resistance mediated by MFS drug efflux pumps, and direction for future investigation.
Collapse
Affiliation(s)
- Jerusha Stephen
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Fathima Salam
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Manjusha Lekshmi
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Sanath H. Kumar
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Manuel F. Varela
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA
| |
Collapse
|
7
|
The Potential of Antibiotics and Nanomaterial Combinations as Therapeutic Strategies in the Management of Multidrug-Resistant Infections: A Review. Int J Mol Sci 2022; 23:ijms232315038. [PMID: 36499363 PMCID: PMC9736695 DOI: 10.3390/ijms232315038] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022] Open
Abstract
Antibiotic resistance has become a major public health concern around the world. This is exacerbated by the non-discovery of novel drugs, the development of resistance mechanisms in most of the clinical isolates of bacteria, as well as recurring infections, hindering disease treatment efficacy. In vitro data has shown that antibiotic combinations can be effective when microorganisms are resistant to individual drugs. Recently, advances in the direction of combination therapy for the treatment of multidrug-resistant (MDR) bacterial infections have embraced antibiotic combinations and the use of nanoparticles conjugated with antibiotics. Nanoparticles (NPs) can penetrate the cellular membrane of disease-causing organisms and obstruct essential molecular pathways, showing unique antibacterial mechanisms. Combined with the optimal drugs, NPs have established synergy and may assist in regulating the general threat of emergent bacterial resistance. This review comprises a general overview of antibiotic combinations strategies for the treatment of microbial infections. The potential of antibiotic combinations with NPs as new entrants in the antimicrobial therapy domain is discussed.
Collapse
|
8
|
Li W, Yang Z, Hu J, Wang B, Rong H, Li Z, Sun Y, Wang Y, Zhang X, Wang M, Xu H. Evaluation of culturable 'last-resort' antibiotic resistant pathogens in hospital wastewater and implications on the risks of nosocomial antimicrobial resistance prevalence. JOURNAL OF HAZARDOUS MATERIALS 2022; 438:129477. [PMID: 35780736 DOI: 10.1016/j.jhazmat.2022.129477] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 05/02/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
Antimicrobial resistance has been recognized as an important emerging environmental pollutant. 'Last-resort' antibiotics including tigecycline, polymyxin E, daptomycin, vancomycin and linezolid are the 'last line of defence' for antibiotic resistant pathogen infections. Therefore, the presence of 'last-resort' antibiotic resistant pathogens in hospital environments and the nosocomial transmission of 'last-resort' antibiotic resistance poses a grave threat to the well-being of patients. In this work, the extent of resistance to 'last-resort' antibiotics in culturable pathogens in hospital wastewater was investigated. Resistance to 'last-resort' antibiotics were quantified for 1384 culturable Enterobacteriaceae, Enterococcus, Staphylococcus, and Pseudomonas strains. With these investigations, several significant findings were made: (1) a very high level of resistance to 'last-resort' antibiotics was found; (2) multiple resistance to antibiotics, including 'last-resort' antibiotics, was prevalent; (3) a high level of 'last-resort' antibiotic resistance phenotype-genotype inconsistency was found, suggesting knowledge gap for resistance mechanisms; 4) tet(X4)-containing tigecycline-resistant Gram-positive pathogens were found for the first time; 5) wastewater treatment processes are effective in preventing the release of 'last-resort' antibiotic resistant pathogens to the environment. This investigation reveals the severe situation on 'last-resort' resistance in the hospital environment, and implies high risk for nosocomial transmission of 'last-resort' antibiotic resistant pathogens.
Collapse
Affiliation(s)
- Weiwei Li
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China; Division of Science and Technology, Ludong University, Yantai, Shandong 264025, China
| | - Zhongjun Yang
- Department of Stomatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong 266035, China
| | - Jiamin Hu
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China
| | - Bianfang Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China
| | - Hao Rong
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China
| | - Ziyun Li
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China
| | - Yuqing Sun
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China
| | - Yunkun Wang
- Shandong Provincial Key Laboratory of Water Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong 266237, China
| | - Xuhua Zhang
- Laboratory Medicine Center, The Second Hospital of Shandong University, Jinan, Shandong 250000, China
| | - Mingyu Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China.
| | - Hai Xu
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
9
|
Bastian AA, Bastian M, Jäger M, Loznik M, Warszawik EM, Yang X, Tahiri N, Fodran P, Witte MD, Thoma A, Köhler J, Minnaard AJ, Herrmann A. Late-Stage Modification of Aminoglycoside Antibiotics Overcomes Bacterial Resistance Mediated by APH(3') Kinases. Chemistry 2022; 28:e202200883. [PMID: 35388562 PMCID: PMC9321007 DOI: 10.1002/chem.202200883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Indexed: 12/25/2022]
Abstract
The continuous emergence of antimicrobial resistance is causing a threat to patients infected by multidrug-resistant pathogens. In particular, the clinical use of aminoglycoside antibiotics, broad-spectrum antibacterials of last resort, is limited due to rising bacterial resistance. One of the major resistance mechanisms in Gram-positive and Gram-negative bacteria is phosphorylation of these amino sugars at the 3'-position by O-phosphotransferases [APH(3')s]. Structural alteration of these antibiotics at the 3'-position would be an obvious strategy to tackle this resistance mechanism. However, the access to such derivatives requires cumbersome multi-step synthesis, which is not appealing for pharma industry in this low-return-on-investment market. To overcome this obstacle and combat bacterial resistance mediated by APH(3')s, we introduce a novel regioselective modification of aminoglycosides in the 3'-position via palladium-catalyzed oxidation. To underline the effectiveness of our method for structural modification of aminoglycosides, we have developed two novel antibiotic candidates overcoming APH(3')s-mediated resistance employing only four synthetic steps.
Collapse
Affiliation(s)
- Andreas A. Bastian
- Department of Chemical BiologyStratingh Institute for ChemistryNijenborgh 79747 AGGroningen (TheNetherlands
- AGILeBiotics B.V.De Mudden 149747 AVGroningen (TheNetherlands
- Institute for Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 252074AachenGermany
| | - Maria Bastian
- AGILeBiotics B.V.De Mudden 149747 AVGroningen (TheNetherlands
| | - Manuel Jäger
- Department of Chemical BiologyStratingh Institute for ChemistryNijenborgh 79747 AGGroningen (TheNetherlands
| | - Mark Loznik
- Department of Polymer ChemistryZernike Institute for Advanced MaterialsNijenborgh 49747 AGGroningen (TheNetherlands
- DWI – Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052056AachenGermany
- Institute for Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 252074AachenGermany
| | - Eliza M. Warszawik
- Department of Polymer ChemistryZernike Institute for Advanced MaterialsNijenborgh 49747 AGGroningen (TheNetherlands
- Department of Biomedical Engineering-FB40W. J. Kolff Institute-FB41Antonius Deusinglaan 19713 AVGroningen (TheNetherlands
| | - Xintong Yang
- Department of Polymer ChemistryZernike Institute for Advanced MaterialsNijenborgh 49747 AGGroningen (TheNetherlands
- DWI – Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052056AachenGermany
| | - Nabil Tahiri
- Department of Chemical BiologyStratingh Institute for ChemistryNijenborgh 79747 AGGroningen (TheNetherlands
| | - Peter Fodran
- Department of Chemical BiologyStratingh Institute for ChemistryNijenborgh 79747 AGGroningen (TheNetherlands
| | - Martin D. Witte
- Department of Chemical BiologyStratingh Institute for ChemistryNijenborgh 79747 AGGroningen (TheNetherlands
| | - Anne Thoma
- DWI – Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052056AachenGermany
- Institute for Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 252074AachenGermany
| | - Jens Köhler
- DWI – Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052056AachenGermany
| | - Adriaan J. Minnaard
- Department of Chemical BiologyStratingh Institute for ChemistryNijenborgh 79747 AGGroningen (TheNetherlands
| | - Andreas Herrmann
- Department of Polymer ChemistryZernike Institute for Advanced MaterialsNijenborgh 49747 AGGroningen (TheNetherlands
- DWI – Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052056AachenGermany
- Institute for Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 252074AachenGermany
| |
Collapse
|
10
|
Torres-Martínez R, Moreno-León A, García-Rodríguez YM, Hernández-Delgado T, Delgado-Lamas G, Espinosa-García FJ. The Tagetes lucida Cav. essential oil and the mixture of its main compounds are antibacterial and modulate antibiotic resistance in multi-resistant pathogenic bacteria. Lett Appl Microbiol 2022; 75:210-223. [PMID: 35419861 DOI: 10.1111/lam.13721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/24/2022] [Accepted: 04/05/2022] [Indexed: 11/26/2022]
Abstract
We evaluated an essential oil (EO) of Tagetes lucida Cav. and the mixture of its main compounds against multi-drug resistant bacteria. We found that EO and the partially reconstituted blend of its main components have antibacterial activity and inhibit antibiotic resistance (ampicillin, chloramphenicol, nalidixic acid, vancomycin, and imipenem) in strains of Staphylococcus aureus ATCC 29213 and Pseudomonas aeruginosa HIM-MR01. The T. lucida EO alone or added to the antibiotics showed antimicrobial activity against S. aureus and P. aeruginosa. The EO main bioactive compounds were methyl eugenol (46.15%), estragole (32.93%), linalool (2.48%), and geraniol (0.33%). The mixture (PREO) of those compounds at those proportions inhibited the growth of P. aeruginosa in 45% at 683.62 µg mL-1 and that of S. aureus in 51.7% at 39.04 µg mL-1 . The PREO had higher antibacterial and modulatory activities than the original EO. In conclusion, we overcame the unpredictability of EO activity (due to their natural variability) by determining which EO components inhibited bacteria and then producing a PREO to generate a reproducible mixture with predictable antibacterial and modulation of resistance activities. Thus, the PREO, and its components, show potential as alternatives to manage multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Rafael Torres-Martínez
- Laboratorio de Ecología Química y Agroecología, Instituto de Investigaciones en Ecosistemas, Universidad Nacional Autónoma de México, Ex Hacienda de San José de la Huerta 58190, Morelia, Michoacán, México
| | - Agustín Moreno-León
- Laboratorio de Ecología Química y Agroecología, Instituto de Investigaciones en Ecosistemas, Universidad Nacional Autónoma de México, Ex Hacienda de San José de la Huerta 58190, Morelia, Michoacán, México
| | - Yolanda Magdalena García-Rodríguez
- Laboratorio de Ecología Química y Agroecología, Instituto de Investigaciones en Ecosistemas, Universidad Nacional Autónoma de México, Ex Hacienda de San José de la Huerta 58190, Morelia, Michoacán, México
| | - Tzasná Hernández-Delgado
- Laboratorio de Bioactividad de Productos Naturales, UBIPRO, FES-Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla, 54090, Estado de México, México
| | - Guillermo Delgado-Lamas
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Coyoacán, 04510, Ciudad de México, México
| | - Francisco Javier Espinosa-García
- Laboratorio de Ecología Química y Agroecología, Instituto de Investigaciones en Ecosistemas, Universidad Nacional Autónoma de México, Ex Hacienda de San José de la Huerta 58190, Morelia, Michoacán, México
| |
Collapse
|
11
|
Muhsin S, Bakir WE, Sabbah M. Identification and sequencing of ISAba2 of Acinetobacter baumannii isolated from baghdad hospitals. MUSTANSIRIYA MEDICAL JOURNAL 2022. [DOI: 10.4103/mj.mj_17_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
|
12
|
Detrimental Effect of Ozone on Pathogenic Bacteria. Microorganisms 2021; 10:microorganisms10010040. [PMID: 35056489 PMCID: PMC8779011 DOI: 10.3390/microorganisms10010040] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/11/2021] [Accepted: 12/19/2021] [Indexed: 12/13/2022] Open
Abstract
(1) Background: Disinfection of medical devices designed for clinical use associated or not with the growing area of tissue engineering is an urgent need. However, traditional disinfection methods are not always suitable for some biomaterials, especially those sensitive to chemical, thermal, or radiation. Therefore, the objective of this study was to evaluate the minimal concentration of ozone gas (O3) necessary to control and kill a set of sensitive or multi-resistant Gram-positive and Gram-negative bacteria. The cell viability, membrane permeability, and the levels of reactive intracellular oxygen (ROS) species were also investigated; (2) Material and Methods: Four standard strains and a clinical MDR strain were exposed to low doses of ozone at different concentrations and times. Bacterial inactivation (cultivability, membrane damage) was investigated using colony counts, resazurin as a metabolic indicator, and propidium iodide (PI). A fluorescent probe (H2DCFDA) was used for the ROS analyses; (3) Results: No reduction in the count colony was detected after O3 exposure compared to the control group. However, the cell viability of E. coli (30%), P. aeruginosa (25%), and A. baumannii (15%) was reduced considerably. The bacterial membrane of all strains was not affected by O3 but presented a significant increase of ROS in E. coli (90 ± 14%), P. aeruginosa (62.5 ± 19%), and A. baumanni (52.6 ± 5%); (4) Conclusion: Low doses of ozone were able to interfere in the cell viability of most strains studied, and although it does not cause damage to the bacterial membrane, increased levels of reactive ROS are responsible for causing a detrimental effect in the lipids, proteins, and DNA metabolism.
Collapse
|
13
|
Mohsin S, Abdul-Elah Bakir W, Arsheed M. Determination of the prevalence of blaoxa-like gene and ISAba1 elements among extensiveــdrug resistant (XDR) Acinetobacter boumannii isolates. BIONATURA 2021. [DOI: 10.21931/rb/2021.06.04.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The capacity of Multiـdrug resistant (MDR) Acinetobacter baumannii to survive in any state of affairs concerning the gaining of various gene types of virulence and antimicrobial agent resistance are the main anxiety in the hospital’s environments. So, it is very crucial to determine the prevalence of insertion sequences in A. baumannii. In the hospitals. Detecting the blaoxa-51 gene through the polymerase chain reaction (PCR) was performed to confirm Acinetobacter baumannii and the search for ISAba1 element. Between October 2020 and February 2021, 540 distinct clinical specimens were gathered from five hospitals in Baghdad. Thirty-eight A. baumannii isolates were obtained from various clinical specimens. The isolates were initially identified phenotypically using standard microbiological techniques and by the Vitek2 compact automated machine. Isolates of A. baumannii were identified genotypically by amplification of the blaoxa-51-like gene. Antimicrobials are studied by Kirby-Bauer (disc diffusion) technique on Muller-Hinton agar as specified by the recent clinical and laboratory standard institute (CLSI) guidelines (2020). The actual results of the current study indicated that from total isolated (38) A.baumannii isolates, 23 isolates (61%) were resistant to meropenem and 25 isolates (66%) were resistant to imipenem. The blaoxa-51 gene was identified in all strains examined, ISAba1 was also present in all A. baumannii isolates. ISAba1 has a high predominance between drug-resistant A. baumannii. Identifying these parameters can assist in the control of infection and decreasing the microorganism’s prevalence rate.
Collapse
Affiliation(s)
- Salah Mohsin
- Department of Microbiology, College of Medicine, Mustansiriyah University. Baghdad. Iraq
| | - Wasan Abdul-Elah Bakir
- Department of Microbiology, College of Medicine, Mustansiriyah University. Baghdad. Iraq
| | - Majeed Arsheed
- Gene bank Department/ Forensic DNA for research and training Centre/ Al-nahrain University, Baghdad. Iraq
| |
Collapse
|
14
|
Edache EI, Uzairu A, Shallangwa GA, Mamza PA. Virtual screening, pharmacokinetics, and molecular dynamics simulations studies to identify potent approved drugs for Chlamydia trachomatis treatment. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00367-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The most frequent bacterial sexually transmitted disease is Chlamydia trachomatis (STD). In 2010, the Centers for Disease Control and Prevention (CDC) received 1.3 million reports of cases (CDC). Human chlamydial infections are linked to a variety of clinical symptoms. Inclusion (IncA) membranes are a promising drug target for the treatment of Chlamydia trachomatis. In the present study, molecular docking, ADMET, golden triangle, and molecular dynamics (MD) simulation studies were performed on a series of salicylidene acylhydrazides derivatives against Chlamydia trachomatis. Three types of docking software with different algorithms were used to screen the potential candidate against Chlamydia trachomatis.
Results
The results obtained from the docking analysis succeeded in screening nine novel hit compounds with high affinity to IncA membranes. Then, pharmacokinetics properties were calculated to spot out the drug-likeness of the selected compounds. Also, golden triangles were performed on the selected compounds. Compounds outside the golden triangle indicate that they would have clearance problems. Out of the nine novel hits drugs, four compounds pass the golden triangle screening and virtually all the quality assurance tests proposed by the model and were used for further analysis. One-ns molecular dynamics simulations on the docked complex of compound 44 (one of the highly active selected compounds of the dataset) aided in the further exploration of the binding interactions. Some crucial residues such as Ser111, Gln114, Asn107, Leu142, Gly144, Gln143, Lys104, Tyr149, Phe108, Phe145, and Arg146 were identified. Conventional and carbon–hydrogen bond interactions with amino residues Arg146, Asn107, Phe145, and Ser111 were critical for the binding of inclusion (IncA) membranes inhibitors.
Conclusion
Outcomes of the study can further be exploited to develop potent inclusion (IncA) membranes inhibitors.
Collapse
|
15
|
Pinkert L, Lai YH, Peukert C, Hotop SK, Karge B, Schulze LM, Grunenberg J, Brönstrup M. Antibiotic Conjugates with an Artificial MECAM-Based Siderophore Are Potent Agents against Gram-Positive and Gram-Negative Bacterial Pathogens. J Med Chem 2021; 64:15440-15460. [PMID: 34619959 DOI: 10.1021/acs.jmedchem.1c01482] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The development of novel drugs against Gram-negative bacteria represents an urgent medical need. To overcome their outer cell membrane, we synthesized conjugates of antibiotics and artificial siderophores based on the MECAM core, which are imported by bacterial iron uptake systems. Structures, spin states, and iron binding properties were predicted in silico using density functional theory. The capability of MECAM to function as an effective artificial siderophore in Escherichia coli was proven in microbiological growth recovery and bioanalytical assays. Following a linker optimization focused on transport efficiency, five β-lactam and one daptomycin conjugates were prepared. The most potent conjugate 27 showed growth inhibition of Gram-positive and Gram-negative multidrug-resistant pathogens at nanomolar concentrations. The uptake pathway of MECAMs was deciphered by knockout mutants and highlighted the relevance of FepA, CirA, and Fiu. Resistance against 27 was mediated by a mutation in the gene encoding ExbB, which is involved in siderophore transport.
Collapse
Affiliation(s)
- Lukas Pinkert
- Department of Chemical Biology Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Yi-Hui Lai
- Department of Chemical Biology Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Carsten Peukert
- Department of Chemical Biology Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Sven-Kevin Hotop
- Department of Chemical Biology Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Bianka Karge
- Department of Chemical Biology Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Lara Marie Schulze
- Institute for Organic Chemistry, Technical University of Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| | - Jörg Grunenberg
- Institute for Organic Chemistry, Technical University of Braunschweig, Hagenring 30, 38106 Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, 38124 Braunschweig, Germany.,Center of Biomolecular Drug Research (BMWZ), Leibniz Universität, 30159 Hannover, Germany
| |
Collapse
|
16
|
Screening of compound library identifies novel inhibitors against the MurA enzyme of Escherichia coli. Appl Microbiol Biotechnol 2021; 105:3611-3623. [PMID: 33860835 DOI: 10.1007/s00253-021-11272-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/27/2021] [Accepted: 04/05/2021] [Indexed: 11/27/2022]
Abstract
Bacterial cell has always been an attractive target for anti-infective drug discovery. MurA (UDP-N-acetylglucosamine enolpyruvyl transferase) enzyme of Escherichia coli (E.coli) is crucial for peptidoglycan biosynthetic pathway, as it is involved in the early stages of bacterial cell wall biosynthesis. In the present study we aim to identify novel chemical structures targeting the MurA enzyme. For screening purpose, we used in silico approach (pharmacophore based strategy) for 52,026 library compounds (Chembridge, Chemdiv and in house synthetics) which resulted in identification of 50 compounds. These compounds were screened in vitro against MurA enzyme and release of inorganic phosphate (Pi) was estimated. Two compounds (IN00152 and IN00156) were found to inhibit MurA enzyme > 70% in primary screening and IC50 of 14.03 to 32.30 μM respectively. These two hits were further evaluated for their mode of inhibition studies and whole-cell activity where we observed 2-4 folds increase in activity in presence of Permeabilizer EDTA (Ethylenediaminetetraacetic acid). Combination studies were also performed with known antibiotics in presence of EDTA. Hits are reported for the first time against this target and our report also support the use of OM permeabilizer in combination with antibacterial compounds to address the permeability and efficacy issue. These lead hits can be further optimized for drug discovery. KEY POINTS: • Emerging Gram negative resistant strains is a matter of concern. • Need for new screening strategies to cope with drying up antibiotics pipeline. • Outer membrane permeabilizers could be useful to improve potency of molecules to reach its target.
Collapse
|
17
|
Verma P, Tiwari M, Tiwari V. Efflux pumps in multidrug-resistant Acinetobacter baumannii: Current status and challenges in the discovery of efflux pumps inhibitors. Microb Pathog 2021; 152:104766. [PMID: 33545327 DOI: 10.1016/j.micpath.2021.104766] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/13/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
Acinetobacter baumannii is an ESKAPE pathogen known to cause fatal nosocomial infections. With the surge of multidrug resistance (MDR) in the bacterial system, effective treatment measures have become very limited. The MDR in A. baumannii is contributed by various factors out of which efflux pumps have gained major attention due to their broad substrate specificity and wide distribution among bacterial species. The efflux pumps are involved in the MDR as well as contribute to other physiological processes in bacteria, therefore, it is critically important to inhibit efflux pumps in order to combat emerging resistance. The present review provides insight about the different efflux pump systems in A. baumannii and their role in multidrug resistance. A major focus has been put on the different strategies and alternate therapeutics to inhibit the efflux system. This includes use of different efflux pump inhibitors-natural, synthetic or combinatorial therapy. The use of phage therapy and nanoparticles for inhibiting efflux pumps have also been discussed here. Moreover, the present review provides the knowledge of barriers in development of efflux pump inhibitors (EPIs) and their approval for commercialization. Here, different prospectives have been discussed to improve the therapeutic development process and make it more compatible for clinical use.
Collapse
Affiliation(s)
- Privita Verma
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India.
| |
Collapse
|
18
|
Ismail MM, Samir R, Saber FR, Ahmed SR, Farag MA. Pimenta Oil as A Potential Treatment for Acinetobacter Baumannii Wound Infection: In Vitro and In Vivo Bioassays in Relation to Its Chemical Composition. Antibiotics (Basel) 2020; 9:antibiotics9100679. [PMID: 33036456 PMCID: PMC7600634 DOI: 10.3390/antibiotics9100679] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/04/2020] [Accepted: 10/04/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial biofilm contributes to antibiotic resistance. Developing antibiofilm agents, more favored from natural origin, is a potential method for treatment of highly virulent multidrug resistant (MDR) bacterial strains; The potential of Pimenta dioica and Pimenta racemosa essential oils (E.Os) antibacterial and antibiofilm activities in relation to their chemical composition, in addition to their ability to treat Acinetobacter baumannii wound infection in mice model were investigated; P. dioica leaf E.O at 0.05 µg·mL−1 efficiently inhibited and eradicated biofilm formed by A. baumannii by 85% and 34%, respectively. Both P. diocia and P. racemosa leaf E.Os showed a bactericidal action against A. baumanii within 6h at 2.08 µg·mL−1. In addition, a significant reduction of A. baumannii microbial load in mice wound infection model was found. Furthermore, gas chromatography mass spectrometry analysis revealed qualitative and quantitative differences among P. racemosa and P. dioica leaf and berry E.Os. Monoterpene hydrocarbons, oxygenated monoterpenes, and phenolics were the major detected classes. β-Myrcene, limonene, 1,8-cineole, and eugenol were the most abundant volatiles. While, sesquiterpenes were found as minor components in Pimenta berries E.O; Our finding suggests the potential antimicrobial activity of Pimenta leaf E.O against MDR A. baumannii wound infections and their underlying mechanism and to be further tested clinically as treatment for MDR A. baumannii infections.
Collapse
Affiliation(s)
- Maha M. Ismail
- Microbiology and Immunology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt;
- Correspondence: (M.M.I.); (F.R.S.); Tel./Fax: +20-3628426 (ext. 00202) (F.R.S.)
| | - Reham Samir
- Microbiology and Immunology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt;
| | - Fatema R. Saber
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr el-Aini Street, Cairo 11562, Egypt; (S.R.A.); (M.A.F.)
- Correspondence: (M.M.I.); (F.R.S.); Tel./Fax: +20-3628426 (ext. 00202) (F.R.S.)
| | - Shaimaa R. Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr el-Aini Street, Cairo 11562, Egypt; (S.R.A.); (M.A.F.)
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka 2014, Saudi Arabia
| | - Mohamed A. Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr el-Aini Street, Cairo 11562, Egypt; (S.R.A.); (M.A.F.)
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
19
|
Ha Y, Jang M, Lee S, Lee JY, Lee WC, Bae S, Kang J, Han M, Kim Y. Identification of inhibitor binding hotspots in Acinetobacter baumannii β-ketoacyl acyl carrier protein synthase III using molecular dynamics simulation. J Mol Graph Model 2020; 100:107669. [PMID: 32659632 DOI: 10.1016/j.jmgm.2020.107669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/01/2020] [Accepted: 06/13/2020] [Indexed: 10/23/2022]
Abstract
Acinetobacter baumannii is a gram-negative bacterium that is rapidly developing drug resistance due to the abuse of antibiotics. The emergence of multidrug-resistant A. baumannii has greatly contributed to the urgency of developing new antibiotics. Previously, we had discovered two potent inhibitors of A. baumannii β-ketoacyl acyl carrier protein synthase III (abKAS III), YKab-4 and YKab-6, which showed potent activity against A. baumannii. In addition, we have reported the crystal structure of abKAS III. In the present study, we investigated the binding between abKAS III and its inhibitors by docking simulation. Molecular dynamics (MD) simulations were performed using docked inhibitor models to identify the hotspot residues related to inhibitor binding. The binding free energies estimated using the MD simulations suggest that residues I198 and F260 of abKAS III serve as the inhibitor binding hotspots. I198, found to be responsible for mediating hydrophobic interactions with inhibitors, had the strongest residual binding energy among all abKAS III residues. We modeled glutamine substitutions of residues I198 and F260 and estimated the relative binding energies of the I198Q and F260Q variants. The results confirmed that I198 and F260 are the key inhibitor binding residues. The roles of the key residues in inhibitor binding, i.e. F260 in the α9 helix and the I198 in the β6β7 loop region, were investigated using principal component analysis (PCA). PCA revealed the structural changes resulting from the abKAS III I198Q and F260Q mutations and described the essential dynamics of the α9 helix. In addition, the results suggest that the β6β7 loop region may act as a gate keeper for ligand binding. Hydrophobic interactions involving I198 and F260 in abKAS III appear to be essential for the binding of the inhibitors YKab-4 and YKab-6. In conclusion, this study provides valuable information for the rational design of antibiotics via the inhibition of abKAS III.
Collapse
Affiliation(s)
- Yuna Ha
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, South Korea
| | - Mihee Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, South Korea
| | - Sehan Lee
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea
| | - Jee-Young Lee
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea
| | - Woo Cheol Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, South Korea
| | - Seri Bae
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea
| | - Jihee Kang
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea
| | - Minwoo Han
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea.
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
20
|
Tan L, Zhou Z, Liu X, Li J, Zheng Y, Cui Z, Yang X, Liang Y, Li Z, Feng X, Zhu S, Yeung KWK, Yang C, Wang X, Wu S. Overcoming Multidrug-Resistant MRSA Using Conventional Aminoglycoside Antibiotics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902070. [PMID: 32382474 PMCID: PMC7201259 DOI: 10.1002/advs.201902070] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/04/2019] [Indexed: 05/20/2023]
Abstract
Global multidrug-resistant (MDR) bacteria are spreading rapidly and causing a great threat to human health due to the abuse of antibiotics. Determining how to resensitize MDR bacteria to conventional inefficient antibiotics is of extreme urgency. Here, a low-temperature photothermal treatment (PTT, 45 °C) is utilized with red phosphorus nanoparticles to resensitize methicillin-resistant Staphylococcus aureus (MRSA) to conventional aminoglycoside antibiotics. The antibacterial mechanism is studied by the proteomic technique and molecular dynamics (MD) simulation, which proves that the aminoglycoside antibiotics against MRSA can be selectively potentiated by low-temperature PTT. The catalytic activity of 2-aminoglycoside phosphotransferase (APH (2″))-a modifying enzyme-is demonstrated to be obviously inhibited via detecting the consumption of adenosine triphosphate (ATP) in the catalytic reaction. It is also found that the active site of aspartic acid (ASP) residues in APH (2″) is thermally unstable from the results of molecular dynamics simulation. Its catalytic ability is inhibited by preventing the deprotonating procedure for the target -OH of gentamycin. The combined therapy also exhibits great biocompatibility and successfully treats MRSA infections in vivo. This low-temperature PTT strategy has the potential to be an exogenous-modifying enzyme inhibitor for the treatment of MDR bacterial infection.
Collapse
Affiliation(s)
- Lei Tan
- Hubei Key Laboratory of Polymer MaterialsMinistry‐of‐Education Key Laboratory for the Green Preparation and Application of Functional MaterialsSchool of Materials Science & EngineeringHubei UniversityWuhan430062China
| | - Ziao Zhou
- Hubei Key Laboratory of Polymer MaterialsMinistry‐of‐Education Key Laboratory for the Green Preparation and Application of Functional MaterialsSchool of Materials Science & EngineeringHubei UniversityWuhan430062China
| | - Xiangmei Liu
- Hubei Key Laboratory of Polymer MaterialsMinistry‐of‐Education Key Laboratory for the Green Preparation and Application of Functional MaterialsSchool of Materials Science & EngineeringHubei UniversityWuhan430062China
| | - Jun Li
- School of Materials Science & EngineeringThe Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of ChinaTianjin UniversityTianjin300072China
| | - Yufeng Zheng
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and EngineeringCollege of EngineeringPeking UniversityBeijing100871China
| | - Zhenduo Cui
- School of Materials Science & EngineeringThe Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of ChinaTianjin UniversityTianjin300072China
| | - Xianjin Yang
- School of Materials Science & EngineeringThe Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of ChinaTianjin UniversityTianjin300072China
| | - Yanqin Liang
- School of Materials Science & EngineeringThe Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of ChinaTianjin UniversityTianjin300072China
| | - Zhaoyang Li
- School of Materials Science & EngineeringThe Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of ChinaTianjin UniversityTianjin300072China
| | - Xiaobo Feng
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Shengli Zhu
- School of Materials Science & EngineeringThe Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of ChinaTianjin UniversityTianjin300072China
| | - Kelvin Wai Kwok Yeung
- Department of Orthopaedics & TraumatologyLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong KongChina
| | - Cao Yang
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xianbao Wang
- Hubei Key Laboratory of Polymer MaterialsMinistry‐of‐Education Key Laboratory for the Green Preparation and Application of Functional MaterialsSchool of Materials Science & EngineeringHubei UniversityWuhan430062China
| | - Shuilin Wu
- Hubei Key Laboratory of Polymer MaterialsMinistry‐of‐Education Key Laboratory for the Green Preparation and Application of Functional MaterialsSchool of Materials Science & EngineeringHubei UniversityWuhan430062China
- School of Materials Science & EngineeringThe Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of ChinaTianjin UniversityTianjin300072China
| |
Collapse
|
21
|
Garza-González E, Franco-Cendejas R, Morfín-Otero R, Echaniz-Aviles G, Rojas-Larios F, Bocanegra-Ibarias P, Flores-Treviño S, Ponce-de-León A, Rodríguez-Noriega E, Alavez-Ramírez N, Mena-Ramirez JP, Rincón-Zuno J, Fong-Camargo MG, Morales-De-la-Peña CT, Huerta-Baltazar CR, López-Jacome LE, Carnalla-Barajas MN, Soto-Noguerón A, Sanchez-Francia D, Moncada-Barrón D, Ortíz-Brizuela E, García-Mendoza L, Newton-Sánchez OA, Choy-Chang EV, Aviles-Benitez LK, Martínez-Miranda R, Feliciano-Guzmán JM, Peña-Lopez CD, Couoh-May CA, López-Gutiérrez E, Gil-Veloz M, Armenta-Rodríguez LC, Manriquez-Reyes M, Gutierrez-Brito M, López-Ovilla I, Adame-Álvarez C, Barajas-Magallón JM, Aguirre-Burciaga E, Coronado-Ramírez AM, Rosales-García AA, Sida-Rodríguez S, Urbina-Rodríguez RE, López-Moreno LI, Juárez-Velázquez GE, Martínez-Villarreal RT, Canizales-Oviedo JL, Cetina-Umaña CM, Perez-Juárez MM, González-Moreno A, Romero-Romero D, Bello-Pazos FD, Aguilar-Orozco G, Barlandas-Rendón NRE, Maldonado-Anicacio JY, Valadez-Quiroz A, Camacho-Ortiz A. The Evolution of Antimicrobial Resistance in Mexico During the Last Decade: Results from the INVIFAR Group. Microb Drug Resist 2020; 26:1372-1382. [PMID: 32027229 DOI: 10.1089/mdr.2019.0354] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: Surveillance of antimicrobial resistance (AMR) requires an international approach with national and local strategies. Our aim was to summarize a retrospective 10-year report of antibiotic resistance of gram-positive and gram-negative bacteria in Mexico. Methods: A total of 46 centers from 22 states of Mexico participated. Databases of AMR from January 2009 to December 2018 were included for most species. The 10-year period was divided into five 2-year periods. Results: For Staphylococcus aureus, a decrease in resistance in all specimens was observed for erythromycin and oxacillin (p < 0.0001 for each). For Enterobacter spp., resistance to meropenem increased for urine specimens (p = 0.0042). For Klebsiella spp., increased drug resistance in specimens collected from blood was observed for trimethoprim/sulfamethoxazole, gentamicin, tobramycin (p < 0.0001 for each), meropenem (p = 0.0014), and aztreonam (p = 0.0030). For Acinetobacter baumannii complex, high drug resistance was detected for almost all antibiotics, including carbapenems, except for tobramycin, which showed decreased resistance for urine, respiratory, and blood isolates (p < 0.0001 for each), and for amikacin, which showed a decrease in resistance in urine specimens (p = 0.0002). An increase in resistance to cefepime was found for urine, respiratory, and blood specimens (p < 0.0001 for each). For Pseudomonas aeruginosa, aztreonam resistance increased for isolates recovered from blood (p = 0.0001). Conclusion: This laboratory-based surveillance of antibiotic resistance shows that resistance is increasing for some antibiotics in different bacterial species in Mexico and highlights the need for continuous monitoring of antibiotic resistance.
Collapse
Affiliation(s)
- Elvira Garza-González
- Hospital Universitario Dr. José E. González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Rafael Franco-Cendejas
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | - Rayo Morfín-Otero
- Hospital Civil de Guadalajara e Instituto de Patología Infecciosa, Universidad de Guadalajara, Guadalajara, Mexico
| | | | | | - Paola Bocanegra-Ibarias
- Hospital Universitario Dr. José E. González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Samantha Flores-Treviño
- Hospital Universitario Dr. José E. González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Alfredo Ponce-de-León
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Eduardo Rodríguez-Noriega
- Hospital Civil de Guadalajara e Instituto de Patología Infecciosa, Universidad de Guadalajara, Guadalajara, Mexico
| | - Norma Alavez-Ramírez
- Hospital Regional Tipo B, de Alta Especialidad Bicentenario de La Independencia, Tultitlán de Mariano Escobedo, Mexico
| | - Juan Pablo Mena-Ramirez
- Hospital General de Zona No. 21 IMSS, Centro Universitario de los Altos (CUALTOS), Universidad de Guadalajara, Tepatitlán de Morelos, Mexico
| | - Joaquín Rincón-Zuno
- Hospital Para el Niño de Toluca, Instituto Materno Infantil del Estado De México, Toluca, Mexico
| | | | | | | | - Luis Esau López-Jacome
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | | | | | | | | | - Edgar Ortíz-Brizuela
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jorge Luis Canizales-Oviedo
- Centro Universitario De Salud, Universidad Autónoma de Nuevo León, Laboratorio Pueblo Nuevo, Monterrey, Mexico
| | | | | | | | | | | | | | | | | | | | - Adrián Camacho-Ortiz
- Hospital Universitario Dr. José E. González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| |
Collapse
|
22
|
Siebert DCB, Sommer R, Pogorevc D, Hoffmann M, Wenzel SC, Müller R, Titz A. Chemical synthesis of tripeptide thioesters for the biotechnological incorporation into the myxobacterial secondary metabolite argyrin via mutasynthesis. Beilstein J Org Chem 2019; 15:2922-2929. [PMID: 31839838 PMCID: PMC6902895 DOI: 10.3762/bjoc.15.286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/20/2019] [Indexed: 11/23/2022] Open
Abstract
The argyrins are secondary metabolites from myxobacteria with antibiotic activity against Pseudomonas aeruginosa. Studying their structure–activity relationship is hampered by the complexity of the chemical total synthesis. Mutasynthesis is a promising approach where simpler and fully synthetic intermediates of the natural product’s biosynthesis can be biotechnologically incorporated. Here, we report the synthesis of a series of tripeptide thioesters as mutasynthons containing the native sequence with a dehydroalanine (Dha) Michael acceptor attached to a sarcosine (Sar) and derivatives. Chemical synthesis of the native sequence ᴅ-Ala-Dha-Sar thioester required revision of the sequential peptide synthesis into a convergent strategy where the thioester with sarcosine was formed before coupling to the Dha-containing dipeptide.
Collapse
Affiliation(s)
- David C B Siebert
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Roman Sommer
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Domen Pogorevc
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany.,Microbial Natural Substances, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | - Michael Hoffmann
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany.,Microbial Natural Substances, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | - Silke C Wenzel
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany.,Microbial Natural Substances, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | - Rolf Müller
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany.,Microbial Natural Substances, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany.,Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| |
Collapse
|
23
|
Bitew A. High Prevalence of Multi-Drug Resistance and Extended Spectrum Beta Lactamase Production in Non-Fermenting Gram-Negative Bacilli in Ethiopia. Infect Dis (Lond) 2019; 12:1178633719884951. [PMID: 31723320 PMCID: PMC6836305 DOI: 10.1177/1178633719884951] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/06/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Emergence of resistance to multiple antimicrobial agents in Non-Fermenting Gram-Negative Bacilli is a major problem to public health, as it limits drug treatment options against infections. The aim of this study was to determine the prevalence of multi-drug resistance and extended spectrum beta lactamase production in Non-Fermenting Gram-Negative Bacilli. MATERIALS AND METHODS Different clinical samples were collected and processed following standard procedures. Each sample was then inoculated onto culture media. Identification, drug susceptibility testing, and extended spectrum beta lactamase production of the isolates were carried out by using the VITEK 2 compact system. RESULTS Among 996 clinical samples, 135 samples yielded Non-Fermenting Gram-Negative Bacilli of which Pseudomonas and Acinetobacter species were the commonest isolates. The overall drug resistance rates of Non-Fermenting Gram-Negative Bacilli were above 80% against ampicillin (89.6%), cefuroxime axetil (88.9%), nitrofurantoin (85.9%), cefalotin (84.4%), cefoxitin (83.7%), cefazolin (83.0%), and cefuroxime (83.0%). Tobramycin with a resistance rate of 19.3% was the most active antimicrobial agent. Out of 135 isolates, 81.5% were multi-drug resistant of which 13.3% were extensively drug resistant and 10.4% were pandrug resistant. Extended spectrum beta lactamase production was detected in 48.9% of the isolates. CONCLUSIONS The spectrum of bacterial species isolated was diverse. The isolates demonstrated high level of drug resistance in different classes of antibiotics. The magnitude of multi-drug resistance and the level of extended spectrum beta lactamase production were high. Hence, further studies on multi-drug resistant and extended spectrum beta lactamase producing Non-Fermenting Gram-Negative Bacilli both in the community and in hospital setting are essential.
Collapse
Affiliation(s)
- Adane Bitew
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
24
|
Beyene D, Bitew A, Fantew S, Mihret A, Evans M. Multidrug-resistant profile and prevalence of extended spectrum β-lactamase and carbapenemase production in fermentative Gram-negative bacilli recovered from patients and specimens referred to National Reference Laboratory, Addis Ababa, Ethiopia. PLoS One 2019; 14:e0222911. [PMID: 31553773 PMCID: PMC6760794 DOI: 10.1371/journal.pone.0222911] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 09/10/2019] [Indexed: 12/17/2022] Open
Abstract
Background The emergence of multidrug-resistance (MDR), production of extended-spectrum β-lactamases, and carbapenemase in members of fermentative gram-negative bacilli are a serious threat to public health. Objective The aim of this study was to determine the burden of multi-drug resistance, the production of extended-spectrum β-lactamases (ESBLs), and carbapenemase in fermentative Gram-negative bacilli in Ethiopian Public Health Institute. Materials and methods A cross-sectional study was carried out from December 2017 to June 2018. Different clinical samples were collected, inoculated, and incubated according to standard protocols related to each sample. Bacterial identification was performed by using the VITEKR 2 compact system using the GNR card. Antimicrobial susceptibility testing was carried out by the Kirby-Bauer disc diffusion method. Production of ESBL and carbapenemase were confirmed by combination disc and modified Hodge Test method respectively. Results A total of 238 fermentative Gram-negative bacilli were recovered during the study period, among which E.coli were the predominant isolates followed by K. pneumoniae. The highest percentage of antibiotic resistance was noted against ampicillin (100%) followed by trimethoprim/sulfamethoxazole (81.9%). The isolates showed better sensitivity towards carbapenem drugs. Out of 238 isolates, 94.5% were MDR and of which 8.8% and 0.8% were extensively and pan drug resistant, respectively. Nearly 67% and 2% of isolates were producers of ESBL and carbapenemase, respectively. The isolation rates of MDR, ESBL, and carbapenemase producing stains of the isolates were ≥70% in intensive care unit while the isolation rates in other wards were ≤25%. Conclusions The findings of this study revealed that the burden of MDR and ESBL was high and carbapenemase producing isolates were also identified which is concerning. This situation warrants a consistent surveillance of antimicrobial resistance of fermentative Gram-negative bacilli and implementation of an efficient infection control program.
Collapse
Affiliation(s)
- Degefu Beyene
- Ethiopian Public Health Institute, Clinical Bacteriology and Mycology Research Case Team, Addis Ababa, Ethiopia
- * E-mail:
| | - Adane Bitew
- Department of Medical Laboratory Sciences, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Surafel Fantew
- Ethiopian Public Health Institute, Clinical Bacteriology and Mycology Research Case Team, Addis Ababa, Ethiopia
| | - Amete Mihret
- Ethiopian Public Health Institute, Clinical Bacteriology and Mycology Research Case Team, Addis Ababa, Ethiopia
| | - Martin Evans
- American Society for Microbiology, New York, New York, United States of America
| |
Collapse
|
25
|
Codjoe FS, Brown CA, Smith TJ, Miller K, Donkor ES. Genetic relatedness in carbapenem-resistant isolates from clinical specimens in Ghana using ERIC-PCR technique. PLoS One 2019; 14:e0222168. [PMID: 31513633 PMCID: PMC6742460 DOI: 10.1371/journal.pone.0222168] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/19/2019] [Indexed: 11/19/2022] Open
Abstract
AIM Enterobacterial repetitive intergenic consensus (ERIC) sequence analysis is a powerful tool for epidemiological analysis of bacterial species. This study aimed to determine the genetic relatedness or variability in carbapenem-resistant isolates by species using this technique. METHODS A total of 111 non-duplicated carbapenem-resistant (CR) Gram-negative bacilli isolates from a three-year collection period (2012-2014) were investigated by enterobacterial repetitive intergenic consensus-polymerase chain reaction (ERIC-PCR) in four selected hospital laboratories in Ghana. The isolates were also screened for carbapenemase and extended spectrum β-lactamase genes by PCR. RESULTS A proportion of 23.4% (26/111) of the genomic DNA extracts were carriers of PCR-positive carbapenemase genes, including 14.4% blaNDM-1, 7.2% blaVIM-1 and 1.8% blaOXA-48. The highest prevalence of carbapenemase genes was from non-fermenters, Acinetobacter baumannii and Pseudomonas aeruginosa. For the ESBL genes tested, 96.4% (107/111) of the CR isolates co-harboured both TEM-1 and SHV-1 genes. The ERIC-PCR gel analysis exhibited 1 to 8 bands ranging from 50 to 800 bp. Band patterns of 93 complex dissimilarities were visually distinguished from the 111 CR isolates studied, while the remaining 18 showed band similarities in pairs. CONCLUSION Overall, ERIC-PCR fingerprints have shown a high level of diversity among the species of Gram-negative bacterial pathogens and specimen collection sites in this study. ERIC-PCR optimisation assays may serve as a suitable genotyping tool for the assessment of genetic diversity or close relatedness of isolates that are found in clinical settings.
Collapse
Affiliation(s)
- Francis S. Codjoe
- Department of Medical Laboratory Sciences, School of Biomedical & Allied Health Sciences, College of Health Sciences, University of Ghana, Ghana
- Biomolecular Science Research Centre, Sheffield Hallam University, Sheffield, England, United Kingdom
| | - Charles A. Brown
- Department of Medical Laboratory Sciences, School of Biomedical & Allied Health Sciences, College of Health Sciences, University of Ghana, Ghana
| | - Thomas J. Smith
- Biomolecular Science Research Centre, Sheffield Hallam University, Sheffield, England, United Kingdom
| | - Keith Miller
- Biomolecular Science Research Centre, Sheffield Hallam University, Sheffield, England, United Kingdom
- * E-mail: (KM); (ESD)
| | - Eric S. Donkor
- Department of Medical Microbiology, School of Biomedical & Allied Health Sciences, College of Health Sciences, University of Ghana, Ghana
- * E-mail: (KM); (ESD)
| |
Collapse
|
26
|
Amarh V, Arthur PK. DNA double-strand break formation and repair as targets for novel antibiotic combination chemotherapy. Future Sci OA 2019; 5:FSO411. [PMID: 31534779 PMCID: PMC6745595 DOI: 10.2144/fsoa-2019-0034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/02/2019] [Indexed: 11/23/2022] Open
Abstract
An unrepaired DNA double-strand break (DSB) is lethal to cells. In bacteria, DSBs are usually repaired either via an error-prone pathway, which ligates the ends of the break or an accurate recombination pathway. Due to this lethality, drugs that induce persistent DSBs have been successful in bacterial infection treatment. However, recurrent usage of these drugs has led to emergence of resistant strains. Several articles have thoroughly reviewed the causes, mechanisms and effects of bacterial drug resistance while others have also discussed approaches for facilitating drug discovery and development. Here, we focus on a hypothetical chemotherapeutic strategy that can be explored for minimizing development of resistance to novel DSB-inducing compounds. We also highlight the possibility of utilizing bacterial DSB repair pathways as targets for the discovery and development of novel antibiotics.
Collapse
Affiliation(s)
- Vincent Amarh
- Department of Biochemistry, Cell & Molecular Biology, West African Center for Cell Biology of Infectious Pathogens, University of Ghana, PO Box LG54, Legon, Accra, Ghana
| | - Patrick K Arthur
- Department of Biochemistry, Cell & Molecular Biology, West African Center for Cell Biology of Infectious Pathogens, University of Ghana, PO Box LG54, Legon, Accra, Ghana
| |
Collapse
|
27
|
Ugwuja CG, Adelowo OO, Ogunlaja A, Omorogie MO, Olukanni OD, Ikhimiukor OO, Iermak I, Kolawole GA, Guenter C, Taubert A, Bodede O, Moodley R, Inada NM, de Camargo ASS, Unuabonah EI. Visible-Light-Mediated Photodynamic Water Disinfection @ Bimetallic-Doped Hybrid Clay Nanocomposites. ACS APPLIED MATERIALS & INTERFACES 2019; 11:25483-25494. [PMID: 31268651 DOI: 10.1021/acsami.9b01212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This study reports a new class of photocatalytic hybrid clay nanocomposites prepared from low-cost sources (kaolinite clay and Carica papaya seeds) doped with Zn and Cu salts via a solvothermal process. X-ray diffraction analysis suggests that Cu-doping and Cu/Zn-doping introduce new phases into the crystalline structure of Kaolinite clay, which is linked to the reduced band gap of kaolinite from typically between 4.9 and 8.2 eV to 2.69 eV for Cu-doped and 1.5 eV for Cu/Zn hybrid clay nanocomposites (Nisar, J.; Århammar, C.; Jämstorp, E.; Ahuja, R. Phys. Rev. B 2011, 84, 075120). In the presence of solar light irradiation, Cu- and Cu/Zn-doped nanocomposites facilitate the electron-hole pair separation. This promotes the generation of singlet oxygen which in turn improves the water disinfection efficiencies of these novel nanocomposite materials. The nanocomposite materials were further characterized using high-resolution scanning electron microscopy, fluorimetry, thermogravimetric analysis, and Raman spectroscopy. The breakthrough times of the nanocomposites for a fixed bed mode of disinfection of water contaminated with 2.32 × 107 cfu/mL E. coli ATCC 25922 under solar light irradiation are 25 h for Zn-doped, 30 h for Cu-doped, and 35 h for Cu/Zn-doped nanocomposites. In the presence of multidrug and multimetal resistant strains of E. coli, the breakthrough time decreases significantly. Zn-only doped nanocomposites are not photocatalytically active. In the absence of light, the nanocomposites are still effective in decontaminating water, although less efficient than under solar light irradiation. Electrostatic interaction, metal toxicity, and release of singlet oxygen (only in the Cu-doped and Cu/Zn-doped nanocomposites) are the three disinfection mechanisms by which these nanocomposites disinfect water. A regrowth study indicates the absence of any living E. coli cells in treated water even after 4 days. These data and the long hydraulic times (under gravity) exhibited by these nanocomposites during photodisinfection of water indicate an unusually high potential of these nanocomposites as efficient, affordable, and sustainable point-of-use systems for the disinfection of water in developing countries.
Collapse
Affiliation(s)
| | - Olawale O Adelowo
- Department of Microbiology , University of Ibadan , PMB 5116 , Ibadan , Oyo State 200284 , Nigeria
| | | | | | | | - Odion O Ikhimiukor
- Department of Microbiology , University of Ibadan , PMB 5116 , Ibadan , Oyo State 200284 , Nigeria
| | - Ievgeniia Iermak
- São Carlos Institute of Physics , University of São Paulo , Avenida Trabalhador Sãocarlense 400 , São Carlos 13566-590 , Brazil
| | - Gabriel A Kolawole
- Department of Chemistry , University of Zululand , Kwadlangezwa , 3886 , Republic of South Africa
| | | | | | - Olusola Bodede
- School of Chemistry and Physics , University of KwaZulu-Natal , Westville Campus , Durban , 3630 , South Africa
| | - Roshila Moodley
- School of Chemistry and Physics , University of KwaZulu-Natal , Westville Campus , Durban , 3630 , South Africa
| | - Natalia M Inada
- São Carlos Institute of Physics , University of São Paulo , Avenida Trabalhador Sãocarlense 400 , São Carlos 13566-590 , Brazil
| | - Andrea S S de Camargo
- São Carlos Institute of Physics , University of São Paulo , Avenida Trabalhador Sãocarlense 400 , São Carlos 13566-590 , Brazil
| | - Emmanuel I Unuabonah
- São Carlos Institute of Physics , University of São Paulo , Avenida Trabalhador Sãocarlense 400 , São Carlos 13566-590 , Brazil
| |
Collapse
|
28
|
|
29
|
Garza-González E, Morfín-Otero R, Mendoza-Olazarán S, Bocanegra-Ibarias P, Flores-Treviño S, Rodríguez-Noriega E, Ponce-de-León A, Sanchez-Francia D, Franco-Cendejas R, Arroyo-Escalante S, Velázquez-Acosta C, Rojas-Larios F, Quintanilla LJ, Maldonado-Anicacio JY, Martínez-Miranda R, Ostos-Cantú HL, Gomez-Choel A, Jaime-Sanchez JL, Avilés-Benítez LK, Feliciano-Guzmán JM, Peña-López CD, Couoh-May CA, Molina-Jaimes A, Vázquez -Narvaez EG, Rincón-Zuno J, Rivera-Garay R, Galindo-Espinoza A, Martínez-Ramirez A, Mora JP, Corte- Rojas RE, López-Ovilla I, Monroy-Colin VA, Barajas-Magallón JM, Morales-De-la-Peña CT, Aguirre-Burciaga E, Coronado-Ramírez M, Rosales-García AA, Ayala-Tarín MDJ, Sida-Rodríguez S, Pérez-Vega BA, Navarro-Rodríguez A, Juárez-Velázquez GE, Cetina-Umaña CM, Mena-Ramírez JP, Canizales-Oviedo J, Moreno-Méndez MI, Romero-Romero D, Arévalo-Mejía A, Cobos-Canul DI, Aguilar-Orozco G, Silva-Sánchez J, Camacho-Ortiz A. A snapshot of antimicrobial resistance in Mexico. Results from 47 centers from 20 states during a six-month period. PLoS One 2019; 14:e0209865. [PMID: 30913243 PMCID: PMC6435111 DOI: 10.1371/journal.pone.0209865] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/12/2018] [Indexed: 01/22/2023] Open
Abstract
Aim We aimed to assess the resistance rates of antimicrobial-resistant, in bacterial pathogens of epidemiological importance in 47 Mexican centers. Material and methods In this retrospective study, we included a stratified sample of 47 centers, covering 20 Mexican states. Selected isolates considered as potential causatives of disease collected over a 6-month period were included. Laboratories employed their usual methods to perform microbiological studies. The results were deposited into a database and analyzed with the WHONET 5.6 software. Results In this 6-month study, a total of 22,943 strains were included. Regarding Gram-negatives, carbapenem resistance was detected in ≤ 3% in Escherichia coli, 12.5% in Klebsiella sp. and Enterobacter sp., and up to 40% in Pseudomonas aeruginosa; in the latter, the resistance rate for piperacillin-tazobactam (TZP) was as high as 19.1%. In Acinetobacter sp., resistance rates for cefepime, ciprofloxacin, meropenem, and TZP were higher than 50%. Regarding Gram-positives, methicillin resistance in Staphylococcus aureus (MRSA) was as high as 21.4%, and vancomycin (VAN) resistance reached up to 21% in Enterococcus faecium. Acinetobacter sp. presented the highest multidrug resistance (53%) followed by Klebsiella sp. (22.6%) and E. coli (19.4%). Conclusion The multidrug resistance of Acinetobacter sp., Klebsiella sp. and E. coli and the carbapenem resistance in specific groups of enterobacteria deserve special attention in Mexico. Vancomycin-resistant enterococci (VRE) and MRSA are common in our hospitals. Our results present valuable information for the implementation of measures to control drug resistance.
Collapse
Affiliation(s)
- Elvira Garza-González
- Hospital Universitario Dr. José Eleuterio González, Monterrey, Nuevo León, Mexico
- * E-mail:
| | - Rayo Morfín-Otero
- Hospital Civil de Guadalajara e Instituto de Patología Infecciosa, Guadalajara, Jalisco, Mexico
| | | | | | | | | | - Alfredo Ponce-de-León
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | | | - Rafael Franco-Cendejas
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | | | | | - Fabián Rojas-Larios
- Hospital Regional Universitario de los Servicios de Salud del Estado de Colima y Facultad de Medicina, Universidad de Colima, Colima, Colima, Mexico
| | | | | | - Rafael Martínez-Miranda
- Hospital General de Mexicali/Facultad de Medicina Mexicali UABC, Mexicali, Baja California, Mexico
| | | | | | | | | | | | | | - Carlos A. Couoh-May
- Hospital General de Mérida Yucatán “Dr. Agustín O ‘Horan”, Mérida, Yucatán, Mexico
| | - Aaron Molina-Jaimes
- Hospital Regional de Alta Especialidad Bicentenario de la Independencia, Tultitlán de Mariano Escobedo, Estado de México, Mexico
| | | | | | - Raúl Rivera-Garay
- Hospital Regional de Alta Especialidad del Bajío, León, Guanajuato, Mexico
| | | | | | - Javier P. Mora
- Hospital de Alta Especialidad de Veracruz, Veracruz, Veracruz, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Juan P. Mena-Ramírez
- Hospital General de zona 21 Tepatitlán de Morelos, Tepatitlán de Morelos, Jalisco, Mexico
| | - Jorge Canizales-Oviedo
- Centro Universitario de Salud, UANL Pueblo Nuevo, Monterrey, Nuevo León, Mexico
- Centro Universitario de Salud, UANL Vicente Guerrero, Monterrey, Nuevo León, Mexico
| | | | - Daniel Romero-Romero
- Laboratorio de Análisis Bioquímico Clínicos "Louis Pasteur", Toluca, Estado de México, Mexico
| | | | | | | | | | - Adrián Camacho-Ortiz
- Hospital Universitario Dr. José Eleuterio González, Monterrey, Nuevo León, Mexico
| |
Collapse
|
30
|
Bozkurt Guzel C, Oyardi O, B. Savage P. Comparative in vitro antimicrobial activities of CSA-142 and CSA-192, second-generation ceragenins, with CSA-13 against various microorganisms. J Chemother 2019; 30:332-337. [DOI: 10.1080/1120009x.2018.1534567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Cagla Bozkurt Guzel
- Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Istanbul University, Istanbul, Turkey,
| | - Ozlem Oyardi
- Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Istanbul University, Istanbul, Turkey,
| | - Paul B. Savage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| |
Collapse
|
31
|
Rossitto M, Fiscarelli EV, Rosati P. Challenges and Promises for Planning Future Clinical Research Into Bacteriophage Therapy Against Pseudomonas aeruginosa in Cystic Fibrosis. An Argumentative Review. Front Microbiol 2018; 9:775. [PMID: 29780361 PMCID: PMC5945972 DOI: 10.3389/fmicb.2018.00775] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 04/05/2018] [Indexed: 01/16/2023] Open
Abstract
Although early aggressive and prolonged treatment with specific antibiotics can extend survival in patients with cystic fibrosis (CF) colonized by opportunistic Pseudomonas aeruginosa (PA), antibiotics fail to eradicate the infecting multidrug-resistant (MDR) PA strains in CF. Century-long research has suggested treating patients with bacteriophages (phages, prokaryotic viruses) naturally hosted by bacteria. Although the only phage types used in therapy, lytic phages, lyse PA aggregated in biofilm matrix by depolymerase degrading enzymes, how they can effectively, safely, and persistently do so in patients with CF is unclear. Even though advanced techniques for formulating phage cocktails, training phages and collecting phage libraries have improved efficacy in vitro, whether personalized or ready-to-use therapeutic approaches or phages and antibiotics combined are effective and safe in vivo, and can reduce PA biofilms, remains debatable. Hence, to advance clinical research on phage therapy in clinical trials, also involving mucoid and non-mucoid multidrug-resistant PA in CF, and overcome problems in Western international regulations, we need reliable and repeatable information from experiments in vitro and in vivo on phage characterization, cocktail selection, personalized approaches, and phages combined with antibiotics. These findings, challenges, and promises prompted us to undertake this argumentative review to seek up-to-date information from papers describing lytic phage activity tested in vitro on PA laboratory strains, and PA strains from chronic infections including CF. We also reviewed in vivo studies on phage activity on pulmonary and non-pulmonary animal host models infected by laboratory or CF PA strains. Our argumentative review provides essential information showing that future phage clinical research in CF should use well-characterized and selected phages isolated against CF PA, tested in vitro under dynamic conditions in cocktails or combined with antibiotics, and in vivo on non-pulmonary and pulmonary host models infected with mucoid and non-mucoid CF MDR PA. Our findings should encourage pharmaceutical industries to conduct clinical trials in vitro and in vivo testing patented genomic engineered phages from phage libraries combined with antibiotics to treat or even prevent multidrug-resistant PA in CF, thus helping international regulatory agencies to plan future clinical research on phage therapy in CF.
Collapse
Affiliation(s)
- Martina Rossitto
- Cystic Fibrosis Microbiology, Laboratory Department, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Ersilia V. Fiscarelli
- Cystic Fibrosis Microbiology, Laboratory Department, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Paola Rosati
- Unit of Clinical Epidemiology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| |
Collapse
|
32
|
Zheng W, Sun W, Simeonov A. Drug repurposing screens and synergistic drug-combinations for infectious diseases. Br J Pharmacol 2018; 175:181-191. [PMID: 28685814 PMCID: PMC5758396 DOI: 10.1111/bph.13895] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022] Open
Abstract
Infectious diseases account for nearly one fifth of the worldwide death toll every year. The continuous increase of drug-resistant pathogens is a big challenge for treatment of infectious diseases. In addition, outbreaks of infections and new pathogens are potential threats to public health. Lack of effective treatments for drug-resistant bacteria and recent outbreaks of Ebola and Zika viral infections have become a global public health concern. The number of newly approved antibiotics has decreased significantly in the last two decades compared with previous decades. In parallel with this, is an increase in the number of drug-resistant bacteria. For these threats and challenges to be countered, new strategies and technology platforms are critically needed. Drug repurposing has emerged as an alternative approach for rapid identification of effective therapeutics to treat the infectious diseases. For treatment of severe infections, synergistic drug combinations using approved drugs identified from drug repurposing screens is a useful option which may overcome the problem of weak activity of individual drugs. Collaborative efforts including government, academic researchers and private drug industry can facilitate the translational research to produce more effective new therapeutic agents such as narrow spectrum antibiotics against drug-resistant bacteria for these global challenges. LINKED ARTICLES This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.
Collapse
Affiliation(s)
- Wei Zheng
- National Center for Advancing Translational SciencesNational Institutes of HealthBethesdaMDUSA
| | - Wei Sun
- National Center for Advancing Translational SciencesNational Institutes of HealthBethesdaMDUSA
| | - Anton Simeonov
- National Center for Advancing Translational SciencesNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
33
|
Codjoe FS, Donkor ES. Carbapenem Resistance: A Review. Med Sci (Basel) 2017; 6:medsci6010001. [PMID: 29267233 PMCID: PMC5872158 DOI: 10.3390/medsci6010001] [Citation(s) in RCA: 286] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 11/25/2017] [Accepted: 12/05/2017] [Indexed: 12/16/2022] Open
Abstract
Carbapenem resistance is a major and an on-going public health problem globally. It occurs mainly among Gram-negative pathogens such as Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii, and may be intrinsic or mediated by transferable carbapenemase-encoding genes. This type of resistance genes are already widespread in certain parts of the world, particularly Europe, Asia and South America, while the situation in other places such as sub-Saharan Africa is not well documented. In this paper, we provide an in-depth review of carbapenem resistance providing up-to-date information on the subject.
Collapse
Affiliation(s)
- Francis S Codjoe
- Department of Medical Laboratory Sciences (Microbiology Division), School of Biomedical & Allied Health Sciences, College of Health Sciences, University of Ghana, Korle Bu KB 143 Accra, Ghana.
- Biomolecular Science Research Centre, Sheffield Hallam University, Sheffield S1 1WB, UK.
| | - Eric S Donkor
- Department of Medical Microbiology, School of Biomedical & Allied Health Sciences, College of Health Sciences, University of Ghana, Korle Bu KB 143 Accra, Ghana.
| |
Collapse
|
34
|
Activity of Meropenem-Vaborbactam in Mouse Models of Infection Due to KPC-Producing Carbapenem-Resistant Enterobacteriaceae. Antimicrob Agents Chemother 2017; 62:AAC.01446-17. [PMID: 29109160 DOI: 10.1128/aac.01446-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/26/2017] [Indexed: 12/16/2022] Open
Abstract
Meropenem-vaborbactam (Vabomere) is highly active against Gram-negative pathogens, especially Klebsiella pneumoniae carbapenemase (KPC)-producing, carbapenem-resistant Enterobacteriaceae The objective of these studies was to evaluate the efficacy of meropenem alone and in combination with vaborbactam in mouse thigh and lung infection models. Thighs or lungs of neutropenic mice were infected with KPC-producing carbapenem-resistant Enterobacteriaceae, with meropenem MICs ranging from ≤0.06 to 8 mg/liter in the presence of 8 mg/liter vaborbactam. Mice were treated with meropenem alone or meropenem in combination with vaborbactam every 2 h for 24 h to provide exposures comparable to 2-g doses of each component in humans. Meropenem administered in combination with vaborbactam produced bacterial killing in all strains tested, while treatment with meropenem alone either produced less than 0.5 log CFU/tissue of bacterial killing or none at all. In the thigh model, 11 strains were treated with the combination of meropenem plus vaborbactam (300 plus 50 mg/kg of body weight). This combination produced from 0.8 to 2.89 logs of bacterial killing compared to untreated controls at the start of treatment. In the lung infection model, two strains were treated with the same dosage regimen of meropenem and vaborbactam. The combination produced more than 1.83 logs of bacterial killing against both strains tested compared to untreated controls at the start of treatment. Overall, these data suggest that meropenem-vaborbactam may have utility in the treatment of infections due to KPC-producing carbapenem-resistant Enterobacteriaceae.
Collapse
|
35
|
Sarah MA, Lobna SEH. Combined efficacy of thymol and silver nanoparticles against Staphylococcus aureus. ACTA ACUST UNITED AC 2017. [DOI: 10.5897/ajmr2016-8387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
36
|
Cephem Potentiation by Inactivation of Nonessential Genes Involved in Cell Wall Biogenesis of β-Lactamase-Producing Escherichia coli. Antimicrob Agents Chemother 2017; 61:AAC.01773-16. [PMID: 27956425 DOI: 10.1128/aac.01773-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/07/2016] [Indexed: 11/20/2022] Open
Abstract
Reversal of antimicrobial resistance is an appealing and largely unexplored strategy in drug discovery. The objective of this study was to identify potential targets for "helper" drugs reversing cephem resistance in Escherichia coli strains producing β-lactamases. A CMY-2-encoding plasmid was transferred by conjugation to seven isogenic deletion mutants exhibiting cephem hypersusceptibility. The effect of each mutation was evaluated by comparing the MICs in the wild type and the mutant harboring the same plasmid. Mutation of two genes encoding proteins involved in cell wall biosynthesis, dapF and mrcB, restored susceptibility to cefoxitin (FOX) and reduced the MICs of cefotaxime and ceftazidime, respectively, from the resistant to the intermediate category according to clinical breakpoints. The same mutants harboring a CTX-M-1-encoding plasmid fell into the intermediate or susceptible category for all three drugs. Individual deletion of dapF and mrcB in a clinical isolate of CTX-M-15-producing E. coli sequence type 131 (ST131) resulted in partial reversal of ceftazidime and cefepime resistance but did not reduce MICs below susceptibility breakpoints. Growth curve analysis indicated no fitness cost in a ΔmrcB mutant, whereas a ΔdapF mutant had a 3-fold longer lag phase than the wild type, suggesting that drugs targeting DapF may display antimicrobial activity, in addition to synergizing with selected cephems. DapF appeared to be a potential FOX helper drug target candidate, since dapF inactivation resulted in synergistic potentiation of FOX in the genetic backgrounds tested. The study showed that individual inactivation of two nonessential genes involved in cell wall biogenesis potentiates cephem activity according to drug- and strain-specific patterns.
Collapse
|
37
|
Bagherifard S. Mediating bone regeneration by means of drug eluting implants: From passive to smart strategies. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 71:1241-1252. [PMID: 27987680 DOI: 10.1016/j.msec.2016.11.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/06/2016] [Accepted: 11/02/2016] [Indexed: 02/03/2023]
Abstract
In addition to excellent biocompatibility and mechanical performance, the new generation of bone and craniofacial implants are expected to proactively contribute to the regeneration process and dynamically interact with the host tissue. To this end, integration and sustained delivery of therapeutic agents has become a rapidly expanding area. The incorporated active molecules can offer supplementary features including promoting oteoconduction and angiogenesis, impeding bacterial infection and modulating host body reaction. Major limitations of the current practices consist of low drug stability overtime, poor control of release profile and kinetics as well as complexity of finding clinically appropriate drug dosage. In consideration of the multifaceted cascade of bone regeneration process, this research is moving towards dual/multiple drug delivery, where precise control on simultaneous or sequential delivery, considering the possible synergetic interaction of the incorporated bioactive factors is of utmost importance. Herein, recent advancements in fabrication of synthetic load bearing implants equipped with various drug delivery systems are reviewed. Smart drug delivery solutions, newly developed to provide higher tempo-spatial control on the delivery of the pharmaceutical agents for targeted and stimuli responsive delivery are highlighted. The future trend of implants with bone drug delivery mechanisms and the most common challenges hindering commercialization and the bench to bedside progress of the developed technologies are covered.
Collapse
Affiliation(s)
- Sara Bagherifard
- Politecnico di Milano, Department of Mechanical Engineering, Milan, Italy.
| |
Collapse
|
38
|
Lee JY, Jeong MC, Jeon D, Lee Y, Lee WC, Kim Y. Structure-activity relationship-based screening of antibiotics against Gram-negative Acinetobacter baumannii. Bioorg Med Chem 2016; 25:372-380. [PMID: 27840136 DOI: 10.1016/j.bmc.2016.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/31/2016] [Accepted: 11/01/2016] [Indexed: 12/25/2022]
Abstract
To discover potent antibiotics against the Gram-negative bacteria, we performed a structure-activity relationship (SAR) study of YKsa-6, which was the most potent inhibitor of Staphylococcus aureus β-ketoacyl acyl carrier protein III in our previous study. We identified and selected 11 candidates, and finally screened two active compounds, YKab-4 (4-[(3-chloro-4-methylphenyl)aminoiminomethyl]benzene-1,3-diol) and YKab-6 (4-[[3-(trifluoromethyl)phenyl]aminoiminomethyl]phenol) as inhibitors of Acinetobacter baumannii KAS III (abKAS III). They showed potent antimicrobial activities at 2 or 8 μg/mL, specifically against Acinetobacter baumannii and a strong binding affinity for abKAS III. From the homology modeling, we defined the three-dimensional (3D) structure of abKAS III for the first time and found that it had an extra loop region compared with common Gram-negative bacteria derived KAS IIIs. The docking study revealed that the hydroxyl groups of inhibitors formed extensive hydrogen bonds and the complicated hydrophobic and cation-stacking interactions are important to binding with abKAS III. We confirmed that the hydrophobicity of these compounds might be the essential factor for their antimicrobial activities against Gram-negative bacteria as well as their structural rigidity, a cooperative feature for retaining the hydrophobic interactions between abKAS III and its inhibitors. This study may provide an insight developing strategies for potent antibiotics against A. baumannii.
Collapse
Affiliation(s)
- Jee-Young Lee
- In silico Molecular Design Team, Chemical Occasion by Modeling Alchemy (CheOMA), Anyang-si 14067, Republic of Korea
| | - Min-Cheol Jeong
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Dasom Jeon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yeongjun Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Woo Cheol Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
39
|
Hughes SJ, Barnard L, Mottaghi K, Tempel W, Antoshchenko T, Hong BS, Allali-Hassani A, Smil D, Vedadi M, Strauss E, Park HW. Discovery of Potent Pantothenamide Inhibitors of Staphylococcus aureus Pantothenate Kinase through a Minimal SAR Study: Inhibition Is Due to Trapping of the Product. ACS Infect Dis 2016; 2:627-641. [PMID: 27759386 DOI: 10.1021/acsinfecdis.6b00090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potent antistaphylococcal activity of N-substituted pantothenamides (PanAms) has been shown to at least partially be due to the inhibition of Staphylococcus aureus's atypical type II pantothenate kinase (SaPanKII), the first enzyme of coenzyme A biosynthesis. This mechanism of action follows from SaPanKII having a binding mode for PanAms that is distinct from those of other PanKs. To dissect the molecular interactions responsible for PanAm inhibitory activity, we conducted a mini SAR study in tandem with the cocrystallization of SaPanKII with two classic PanAms (N5-Pan and N7-Pan), culminating in the synthesis and characterization of two new PanAms, N-Pip-PanAm and MeO-N5-PanAm. The cocrystal structures showed that all of the PanAms are phosphorylated by SaPanKII but remain bound at the active site; this occurs primarily through interactions with Tyr240' and Thr172'. Kinetic analysis showed a strong correlation between kcat (slow PanAm turnover) and IC50 (inhibition of pantothenate phosphorylation) values, suggesting that SaPanKII inhibition occurs via a delay in product release. In-depth analysis of the PanAm-bound structures showed that the capacity for accepting a hydrogen bond from the amide of Thr172' was a stronger determinant for PanAm potency than the capacity to π-stack with Tyr240'. The two new PanAms, N-Pip-PanAm and MeO-N5-PanAm, effectively combine both hydrogen bonding and hydrophobic interactions, resulting in the most potent SaPanKII inhibition described to date. Taken together, our results are consistent with an inhibition mechanism wherein PanAms act as SaPanKII substrates that remain bound upon phosphorylation. The phospho-PanAm-SaPanKII interactions described herein may help future antistaphylococcal drug development.
Collapse
Affiliation(s)
| | - Leanne Barnard
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| | | | | | - Tetyana Antoshchenko
- Department
of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, Louisiana 70112, United States
| | | | | | | | | | - Erick Strauss
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Hee-Won Park
- Department
of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
40
|
Kim H, Jang JH, Kim SC, Cho JH. Enhancement of the antimicrobial activity and selectivity of GNU7 against Gram-negative bacteria by fusion with LPS-targeting peptide. Peptides 2016; 82:60-66. [PMID: 27242337 DOI: 10.1016/j.peptides.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 01/18/2023]
Abstract
Antimicrobial peptides (AMPs) provide a potential source of new antimicrobial therapeutics for the treatment of multidrug-resistant pathogens. To develop Gram-negative selective AMPs that can inhibit the effects of lipopolysaccharide (LPS)-induced sepsis, we added various rationally designed LPS-targeting peptides [amino acids 28-34 of lactoferrin (Lf28-34), amino acids 84-99 of bactericidal/permeability increasing protein (BPI84-99), and de novo peptide (Syn)] to the potent AMP, GNU7 (RLLRPLLQLLKQKLR). Compared to our original starting peptide GNU7, hybrid peptides had an 8- to 32-fold improvement in antimicrobial activity against Gram-negative bacteria, such as Escherichia coli and Salmonella typhimurium. Among them, Syn-GNU7 showed the strongest LPS-binding and -neutralizing activities, thus allowing it to selectively eliminate Gram-negative bacteria from within mixed cultures. Our results suggest that LPS-targeting peptides would be useful to increase the antimicrobial activity and selectivity of other AMPs against Gram-negative bacteria.
Collapse
Affiliation(s)
- Hyun Kim
- Division of Life Science, Gyeongsang National University, Jinju 52828, South Korea
| | - Ju Hye Jang
- Research Institute of Life Science, Gyeongsang National University, Jinju 52828, South Korea
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Ju Hyun Cho
- Division of Life Science, Gyeongsang National University, Jinju 52828, South Korea; Research Institute of Life Science, Gyeongsang National University, Jinju 52828, South Korea.
| |
Collapse
|
41
|
Forschner-Dancause S, Poulin E, Meschwitz S. Quorum Sensing Inhibition and Structure-Activity Relationships of β-Keto Esters. Molecules 2016; 21:molecules21080971. [PMID: 27463706 PMCID: PMC5771494 DOI: 10.3390/molecules21080971] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 11/16/2022] Open
Abstract
Traditional therapeutics to treat bacterial infections have given rise to multi-drug resistant pathogens, which pose a major threat to human and animal health. In several pathogens, quorum sensing (QS)-a cell-cell communication system in bacteria-controls the expression of genes responsible for pathogenesis, thus representing a novel target in the fight against bacterial infections. Based on the structure of the autoinducers responsible for QS activity and other QS inhibitors, we hypothesize that β-keto esters with aryl functionality could possess anti-QS activity. A panel of nineteen β-keto ester analogs was tested for the inhibition of bioluminescence (a QS-controlled phenotype) in the marine pathogen Vibrio harveyi. Initial screening demonstrated the need of a phenyl ring at the C-3 position for antagonistic activity. Further additions to the phenyl ring with 4-substituted halo groups or a 3- or 4-substituted methoxy group resulted in the most active compounds with IC50 values ranging from 23 µM to 53 µM. The compounds additionally inhibit green fluorescent protein production by E. coli JB525. Evidence is presented that aryl β-keto esters may act as antagonists of bacterial quorum sensing by competing with N-acyl homoserine lactones for receptor binding. Expansion of the β-keto ester panel will enable us to obtain more insight into the structure-activity relationships needed to allow for the development of novel anti-virulence agents.
Collapse
Affiliation(s)
| | - Emily Poulin
- Department of Chemistry, Salve Regina University, 100 Ochre Point Ave, Newport, RI 02840, USA.
| | - Susan Meschwitz
- Department of Chemistry, Salve Regina University, 100 Ochre Point Ave, Newport, RI 02840, USA.
| |
Collapse
|
42
|
Place of Colistin-Rifampicin Association in the Treatment of Multidrug-Resistant Acinetobacter Baumannii Meningitis: A Case Study. Case Rep Infect Dis 2016; 2016:8794696. [PMID: 27064923 PMCID: PMC4811080 DOI: 10.1155/2016/8794696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/10/2016] [Accepted: 02/15/2016] [Indexed: 11/17/2022] Open
Abstract
Treatment of Acinetobacter baumannii meningitis is an important challenge due to the accumulation of resistance of this bacteria and low meningeal diffusion of several antimicrobial requiring use of an antimicrobial effective combination to eradicate these species. We report a case of Acinetobacter baumannii multidrug-resistant nosocomial meningitis which was successfully treated with intravenous and intrathecal colistin associated with rifampicin.
Collapse
|
43
|
Wagner S, Sommer R, Hinsberger S, Lu C, Hartmann RW, Empting M, Titz A. Novel Strategies for the Treatment of Pseudomonas aeruginosa Infections. J Med Chem 2016; 59:5929-69. [DOI: 10.1021/acs.jmedchem.5b01698] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Stefanie Wagner
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| | - Roman Sommer
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| | - Stefan Hinsberger
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Cenbin Lu
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Martin Empting
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Alexander Titz
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| |
Collapse
|
44
|
Liebens V, Defraine V, Fauvart M. A Whole-Cell-Based High-Throughput Screening Method to Identify Molecules Targeting Pseudomonas aeruginosa Persister Cells. Methods Mol Biol 2016; 1333:113-120. [PMID: 26468104 DOI: 10.1007/978-1-4939-2854-5_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Despite its clinical relevance and the fact that the phenomenon of persistence was discovered in the 1940s, little is known about the mechanisms behind persister cell formation. Research in this field has mainly focused on the model organism Escherichia coli and few genetic determinants of persistence have been described in other bacterial species, impairing the development of target-based strategies to combat these antibiotic-tolerant cells. In this chapter we describe a top-down large-scale screening method capable of specifically identifying small molecule compounds that, in combination with conventional antibiotics, significantly reduce the persister fraction in Pseudomonas aeruginosa. The method is readily adaptable for other species. Further characterization and analysis of the mode of action of the identified compounds can provide additional insight into the mechanisms behind persister formation and can guide the development of future anti-persister therapies.
Collapse
Affiliation(s)
- Veerle Liebens
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven - University of Leuven, Kasteelpark Arenberg 20, Box 2460, Leuven, 3001, Belgium
| | - Valerie Defraine
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven - University of Leuven, Kasteelpark Arenberg 20, Box 2460, Leuven, 3001, Belgium
| | - Maarten Fauvart
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven - University of Leuven, Kasteelpark Arenberg 20, Box 2460, Leuven, 3001, Belgium.
| |
Collapse
|
45
|
The effect of silver or gallium doped titanium against the multidrug resistant Acinetobacter baumannii. Biomaterials 2015; 80:80-95. [PMID: 26708086 DOI: 10.1016/j.biomaterials.2015.11.042] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/12/2015] [Accepted: 11/29/2015] [Indexed: 12/26/2022]
Abstract
Implant-related infection of biomaterials is one of the main causes of arthroplasty and osteosynthesis failure. Bacteria, such as the rapidly-emerging Multi Drug Resistant (MDR) pathogen Acinetobacter Baumannii, initiate the infection by adhering to biomaterials and forming a biofilm. Since the implant surface plays a crucial role in early bacterial adhesion phases, titanium was electrochemically modified by an Anodic Spark Deposition (ASD) treatment, developed previously and thought to provide osseo-integrative properties. In this study, the treatment was modified to insert gallium or silver onto the titanium surface, to provide antibacterial properties. The material was characterized morphologically, chemically, and mechanically; biological properties were investigated by direct cytocompatibility assay, Alkaline Phosphatase (ALP) activity, Scanning Electron Microscopy (SEM), and Immunofluorescent (IF) analysis; antibacterial activity was determined by counting Colony Forming Units, and viability assay. The various ASD-treated surfaces showed similar morphology, micrometric pore size, and uniform pore distribution. Of the treatments studied, gallium-doped specimens showed the best ALP synthesis and antibacterial properties. This study demonstrates the possibility of successfully doping the surface of titanium with gallium or silver, using the ASD technique; this approach can provide antibacterial properties and maintain high osseo-integrative potential.
Collapse
|
46
|
|
47
|
Sharma R, Rani C, Mehra R, Nargotra A, Chib R, Rajput VS, Kumar S, Singh S, Sharma PR, Khan IA. Identification and characterization of novel small molecule inhibitors of the acetyltransferase activity of Escherichia coli N-acetylglucosamine-1-phosphate-uridyltransferase/glucosamine-1-phosphate-acetyltransferase (GlmU). Appl Microbiol Biotechnol 2015; 100:3071-85. [PMID: 26563552 DOI: 10.1007/s00253-015-7123-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/19/2015] [Accepted: 10/26/2015] [Indexed: 10/22/2022]
Abstract
This study aims at identifying novel chemical scaffolds as inhibitors specific to the acetyltransferase domain of a bifunctional enzyme, Escherichia coli GlmU, involved in the cell wall biosynthesis of Gram-negative organisms. A two-pronged approach was used to screen a 50,000 small-molecule library. Using the first approach, the library was in silico screened by docking the library against acetyltransferase domain of E. coli GlmU studies. In the second approach, complete library was screened against Escherichia coli ATCC 25922 to identify the whole cell active compounds. Active compounds from both the screens were screened in a colorimetric absorbance-based assay to identify inhibitors of acetyltransferase domain of E. coli GlmU which resulted in the identification of 1 inhibitor out of 56 hits identified by in silico screening and 4 inhibitors out of 35 whole cell active compounds on Gram-negative bacteria with the most potent inhibitor showing IC50 of 1.40 ± 0.69 μM. Mode of inhibition studies revealed these inhibitors to be competitive with AcCoA and uncompetitive with GlcN-1-P. These selected inhibitors were also tested for their antibacterial and cytotoxic activities. Compounds 5175178 and 5215319 exhibited antibacterial activity that co-related with GlmU inhibition. These compounds, therefore, represent novel chemical scaffolds targeting acetyltransferase activity of E. coli GlmU.
Collapse
Affiliation(s)
- Rashmi Sharma
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Chitra Rani
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Rukmankesh Mehra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu, 180001, India
| | - Amit Nargotra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Reena Chib
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India
| | - Vikrant S Rajput
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Sunil Kumar
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Samsher Singh
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Parduman R Sharma
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Inshad A Khan
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India. .,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India.
| |
Collapse
|
48
|
Liebens V, Gerits E, Knapen WJ, Swings T, Beullens S, Steenackers HP, Robijns S, Lippell A, O'Neill AJ, Veber M, Fröhlich M, Krona A, Lövenklev M, Corbau R, Marchand A, Chaltin P, De Brucker K, Thevissen K, Cammue BP, Fauvart M, Verstraeten N, Michiels J. Identification and characterization of an anti-pseudomonal dichlorocarbazol derivative displaying anti-biofilm activity. Bioorg Med Chem Lett 2015; 24:5404-8. [PMID: 25453797 DOI: 10.1016/j.bmcl.2014.10.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/08/2014] [Accepted: 10/14/2014] [Indexed: 12/31/2022]
Abstract
Pseudomonas aeruginosa strains resistant towards all currently available antibiotics are increasingly encountered, raising the need for new anti-pseudomonal drugs. We therefore conducted a medium-throughput screen of a small-molecule collection resulting in the identification of the N-alkylated 3,6-dihalogenocarbazol 1-(sec-butylamino)-3-(3,6-dichloro-9H-carbazol-9-yl)propan-2-ol (MIC = 18.5 μg mL⁻¹). This compound, compound 1, is bacteriostatic towards a broad spectrum of Gram-positive and Gram-negative pathogens, including P. aeruginosa. Importantly, 1 also eradicates mature biofilms of P. aeruginosa. 1 displays no cytotoxicity against various human cell types, pointing to its potential for further development as a novel antibacterial drug.
Collapse
|
49
|
Hraiech S, Brégeon F, Rolain JM. Bacteriophage-based therapy in cystic fibrosis-associated Pseudomonas aeruginosa infections: rationale and current status. DRUG DESIGN DEVELOPMENT AND THERAPY 2015. [PMID: 26213462 PMCID: PMC4509528 DOI: 10.2147/dddt.s53123] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pulmonary infections involving Pseudomonas aeruginosa are among the leading causes of the deterioration of the respiratory status of cystic fibrosis (CF) patients. The emergence of multidrug-resistant strains in such populations, favored by iterative antibiotic cures, has led to the urgent need for new therapies. Among them, bacteriophage-based therapies deserve a focus. One century of empiric use in the ex-USSR countries suggests that bacteriophages may have beneficial effects against a large range of bacterial infections. Interest in bacteriophages has recently renewed in Western countries, and the in vitro data available suggest that bacteriophage-based therapy may be of significant interest for the treatment of pulmonary infections in CF patients. Although the clinical data concerning this specific population are relatively scarce, the beginning of the first large randomized study evaluating bacteriophage-based therapy in burn infections suggests that the time has come to assess the effectiveness of this new therapy in CF P. aeruginosa pneumonia. Consequently, the aim of this review is, after a brief history, to summarize the evidence concerning bacteriophage efficacy against P. aeruginosa and, more specifically, the in vitro studies, animal models, and clinical trials targeting CF.
Collapse
Affiliation(s)
- Sami Hraiech
- Institut Hospitalo-Universitaire Méditerranée Infection, URMITE CNRS IRD INSERM UMR 7278, Marseille, France ; Réanimation Médicale - Détresses Respiratoires et Infections Sévères, APHM, CHU Nord, Marseille, France
| | - Fabienne Brégeon
- Institut Hospitalo-Universitaire Méditerranée Infection, URMITE CNRS IRD INSERM UMR 7278, Marseille, France ; Service d'Explorations Fonctionnelles Respiratoires, APHM, CHU Nord, Marseille, France
| | - Jean-Marc Rolain
- Institut Hospitalo-Universitaire Méditerranée Infection, URMITE CNRS IRD INSERM UMR 7278, Marseille, France
| |
Collapse
|
50
|
Hraiech S, Papazian L, Rolain JM, Bregeon F. Animal models of polymicrobial pneumonia. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3279-92. [PMID: 26170617 PMCID: PMC4492661 DOI: 10.2147/dddt.s70993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pneumonia is one of the leading causes of severe and occasionally life-threatening infections. The physiopathology of pneumonia has been extensively studied, providing information for the development of new treatments for this condition. In addition to in vitro research, animal models have been largely used in the field of pneumonia. Several models have been described and have provided a better understanding of pneumonia under different settings and with various pathogens. However, the concept of one pathogen leading to one infection has been challenged, and recent flu epidemics suggest that some pathogens exhibit highly virulent potential. Although "two hits" animal models have been used to study infectious diseases, few of these models have been described in pneumonia. Therefore the aims of this review were to provide an overview of the available literature in this field, to describe well-studied and uncommon pathogen associations, and to summarize the major insights obtained from this information.
Collapse
Affiliation(s)
- Sami Hraiech
- IHU Méditerranée infection, URMITE CNRS IRD INSERM UMR 7278, Marseille, France ; Réanimation - Détresses Respiratoires et infections Sévères, APHM, CHU Nord, Marseille, France
| | - Laurent Papazian
- IHU Méditerranée infection, URMITE CNRS IRD INSERM UMR 7278, Marseille, France ; Réanimation - Détresses Respiratoires et infections Sévères, APHM, CHU Nord, Marseille, France
| | - Jean-Marc Rolain
- IHU Méditerranée infection, URMITE CNRS IRD INSERM UMR 7278, Marseille, France
| | - Fabienne Bregeon
- IHU Méditerranée infection, URMITE CNRS IRD INSERM UMR 7278, Marseille, France ; Service d'explorations Fonctionnelles Respiratoires, APHM, CHU Nord, Marseille, France
| |
Collapse
|