1
|
Xu Y, Wang W, Su W, Wang M, Xu H, Zhang X, Li L. A widespread single amino acid mutation in AcrA reduces tigecycline susceptibility in Klebsiella pneumoniae. Microbiol Spectr 2024; 12:e0203023. [PMID: 38032193 PMCID: PMC10782974 DOI: 10.1128/spectrum.02030-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
IMPORTANCE Tigecycline, a glycecycline antibiotic with broad-spectrum activity against almost all Gram-positive and Gram-negative bacteria, is a highly concerned "last-resort" antibiotic. In addition to plasmid-hosted mobile tet(X) conferring high-level resistance to tigecycline, there are many reports suggesting increased expression of AcrAB-TolC efflux pump leads to tigecycline non-susceptibility. However, the role of mutations in AcrAB-TolC on tigecycline resistance has not been identified. This study reports a novel T188A mutation of the AcrA subunit of AcrAB-TolC complex in a clinical tigecycline-resistant Klebsiella pneumoniae strain and reveals the role of AcrA mutation on tigecycline resistance in K. pneumoniae. High prevalence of A188 type AcrA in hypervirulent multidrug-resistant K. pneumoniae indicates that mutations of the AcrAB-TolC complex may play a larger role in determining bacterial pathogenesis and antibiotic susceptibility than previously expected.
Collapse
Affiliation(s)
- Yingchao Xu
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Wenjia Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Wenya Su
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Mingyu Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Hai Xu
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Xuhua Zhang
- Laboratory Medicine Center, The Second Hospital of Shandong University, Jinan, China
| | - Ling Li
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| |
Collapse
|
2
|
Kavanaugh LG, Mahoney AR, Dey D, Wuest WM, Conn GL. Di-berberine conjugates as chemical probes of Pseudomonas aeruginosa MexXY-OprM efflux function and inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.533986. [PMID: 37425949 PMCID: PMC10327050 DOI: 10.1101/2023.03.24.533986] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The Resistance-Nodulation-Division (RND) efflux pump superfamily is pervasive among Gram-negative pathogens and contributes extensively to clinical antibiotic resistance. The opportunistic pathogen Pseudomonas aeruginosa contains 12 RND-type efflux systems, with four contributing to resistance including MexXY-OprM which is uniquely able to export aminoglycosides. At the site of initial substrate recognition, small molecule probes of the inner membrane transporter (e.g., MexY) have potential as important functional tools to understand substrate selectivity and a foundation for developing adjuvant efflux pump inhibitors (EPIs). Here, we optimized the scaffold of berberine, a known but weak MexY EPI, using an in-silico high-throughput screen to identify di-berberine conjugates with enhanced synergistic action with aminoglycosides. Further, docking and molecular dynamics simulations of di-berberine conjugates reveal unique contact residues and thus sensitivities of MexY from distinct P. aeruginosa strains. This work thereby reveals di-berberine conjugates to be useful probes of MexY transporter function and potential leads for EPI development.
Collapse
Affiliation(s)
- Logan G. Kavanaugh
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | | | - Debayan Dey
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - William M. Wuest
- Department of Chemistry, Emory University, Atlanta, GA
- Emory Antibiotic Resistance Center, Emory University, Atlanta, GA
| | - Graeme L. Conn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
- Emory Antibiotic Resistance Center, Emory University, Atlanta, GA
| |
Collapse
|
3
|
Edris SN, Hamad A, Awad DAB, Sabeq II. Prevalence, antibiotic resistance patterns, and biofilm formation ability of Enterobacterales recovered from food of animal origin in Egypt. Vet World 2023; 16:403-413. [PMID: 37042006 PMCID: PMC10082721 DOI: 10.14202/vetworld.2023.403-413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/30/2023] [Indexed: 03/04/2023] Open
Abstract
Background and Aim: The majority of animal-derived food safety studies have focused on foodborne zoonotic agents; however, members of the opportunistic Enterobacteriaceae (Ops) family are increasingly implicated in foodborne and public health crises due to their robust evolution of acquiring antimicrobial resistance and biofilms, consequently require thorough characterization, particularly in the Egyptian food sector. Therefore, this study aimed to determine the distribution and prevalence of Enterobacteriaceae family members in animal-derived foods, as well as their resistance to important antimicrobials and biofilm-forming potential.
Materials and Methods: A total of 274 beef, rabbit meat, chicken meat, egg, butter, and milk samples were investigated for the presence of Enterobacteriaceae. All isolated strains were first recognized using traditional microbiological techniques. Following that, matrix-assisted laser desorption ionization-time of flight mass spectrometry was used to validate the Enterobacteriaceae's identity. The isolated enterobacteria strains were tested on disk diffusion and crystal violet quantitative microtiter plates to determine their antibiotic resistance and capacity to form biofilms.
Results: There have been thirty isolates of Enterobacteriaceae from seven different species and four genera. Out of the three food types, Pseudomonas aeruginosa had the highest prevalence rate (4.1%). With three species, Enterobacter genera had the second-highest prevalence (3.28%) across five different food categories. In four different food types, the Klebsiella genera had the second-highest distribution and third-highest incidence (2.55%). Almost all isolates, except three Proteus mirabilis, showed prominent levels of resistance, particularly to beta-lactam antibiotics. Except for two Enterobacter cloacae and three P. mirabilis isolates, all isolates were classified as multidrug-resistant (MDR) or extensively multidrug-resistant (XDR). The multiple antibiotic resistance index (MARI) of the majority of isolates dropped between 0.273 and 0.727. The highest MARI was conferred by Klebsiella pneumoniae, at 0.727. Overall, 83.33% of the isolates had strong biofilm capacity, while only 16.67% exhibited moderate capacity.
Conclusion: The MDR, XDR, and strong biofilm indicators confirmed in 83.33% of the currently tested Enterobacteriaceae from animal-derived foods suggest that, if not addressed, there may be rising risks to Egypt's economy and public health.
Collapse
Affiliation(s)
- Shimaa N. Edris
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Benha University, Benha 13736, Egypt
| | - Ahmed Hamad
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Benha University, Benha 13736, Egypt
| | - Dina A. B. Awad
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Benha University, Benha 13736, Egypt
| | - Islam I. Sabeq
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Benha University, Benha 13736, Egypt
| |
Collapse
|
4
|
RND Pump-Mediated Efflux of Amotosalen, a Compound Used in Pathogen Inactivation Technology to Enhance Safety of Blood Transfusion Products, May Compromise Its Gram-Negative Anti-Bacterial Activity. mSphere 2023; 8:e0067322. [PMID: 36853056 PMCID: PMC10117049 DOI: 10.1128/msphere.00673-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Pathogen inactivation is a strategy to improve the safety of transfusion products. The only pathogen reduction technology for blood products currently approved in the US utilizes a psoralen compound, called amotosalen, in combination with UVA light to inactivate bacteria, viruses, and protozoa. Psoralens have structural similarity to bacterial multidrug efflux pump substrates. As these efflux pumps are often overexpressed in multidrug-resistant pathogens, we tested whether contemporary drug-resistant pathogens might show resistance to amotosalen and other psoralens based on multidrug efflux mechanisms through genetic, biophysical, and molecular modeling analysis. The main efflux systems in Enterobacterales, Acinetobacter baumannii, and Pseudomonas aeruginosa are tripartite resistance-nodulation-cell division (RND) systems, which span the inner and outer membranes of Gram-negative pathogens, and expel antibiotics from the bacterial cytoplasm into the extracellular space. We provide evidence that amotosalen is an efflux substrate for the E. coli AcrAB, Acinetobacter baumannii AdeABC, and P. aeruginosa MexXY RND efflux pumps. Furthermore, we show that the MICs for contemporary Gram-negative bacterial isolates for these species and others in vitro approached and exceeded the concentration of amotosalen used in the approved platelet and plasma inactivation procedures. These findings suggest that otherwise safe and effective inactivation methods should be further studied to identify possible gaps in their ability to inactivate contemporary, multidrug-resistant bacterial pathogens. IMPORTANCE Pathogen inactivation is a strategy to enhance the safety of transfused blood products. We identify the compound, amotosalen, widely used for pathogen inactivation, as a bacterial multidrug efflux substrate. Specifically, experiments suggest that amotosalen is pumped out of bacteria by major efflux pumps in E. coli, Acinetobacter baumannii, and Pseudomonas aeruginosa. Such efflux pumps are often overexpressed in multidrug-resistant pathogens. Importantly, the MICs for contemporary multidrug-resistant Enterobacterales, Acinetobacter baumannii, Pseudomonas aeruginosa, Burkholderia spp., and Stenotrophomonas maltophilia isolates approached or exceeded the amotosalen concentration used in approved platelet and plasma inactivation procedures, potentially as a result of efflux pump activity. Although there are important differences in methodology between our experiments and blood product pathogen inactivation, these findings suggest that otherwise safe and effective inactivation methods should be further studied to identify possible gaps in their ability to inactivate contemporary, multidrug-resistant bacterial pathogens.
Collapse
|
5
|
Cheng Y, Li Y, Yu R, Ma M, Yang M, Si H. Identification of Novel tet(X3) Variants Resistant To Tigecycline in Acinetobacter Species. Microbiol Spectr 2022; 10:e0133322. [PMID: 36409072 PMCID: PMC9784759 DOI: 10.1128/spectrum.01333-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
The emergence of the tet(X) gene is a severe challenge to global public health security, as clinical tigecycline resistance shows a rapidly rising trend. In this research, we identified two tigecycline-resistant Acinetobacter sp. strains containing seven novel tet(X3) variants recovered from fecal samples from Chinese farms. The seven Tet(X3) variants showed 15.4% to 99.7% amino acid identity with Tet(X3). By expressing tet(X3.7) and tet(X3.9), the tigecycline MIC values for Escherichia coli JM109 increased 64-fold (from 0.13 to 8 mg/L). However, the other tet(X3) variants did not have a significant change in the MIC of tigecycline. We found that the 26th amino acid site of Tet(X3.7) changed from proline to serine, and the 25th amino acid site of Tet(X3.9) changed from glycine to alanine, which reduced the MIC of tigecycline by 2-fold [the MIC of tet(X3) to tigecycline was 16 mg/L] but did not affect its expression to tigecycline. The tet(X3) variants surrounded by mobile genetic elements appeared in the structure of gene clusters with tandem repeat sequences and were adjacent to the site-specific recombinase-encoding gene xerD. Therefore, there is a risk of horizontal transfer of resistant genes. Our study reports seven novel tet(X3) variants; the continuing emergence of tigecycline variants makes continuous monitoring of resistance to tigecycline even more critical. IMPORTANCE Although it is illegal to use tigecycline and carbapenems to treat bacterial infections in animals, we can still isolate bacteria containing both mobile resistance genes from animals, and tet(X) is currently an essential factor in degrading tigecycline. Here, we characterized two multidrug-resistant Acinetobacter sp. strains that contained vital resistance genes, such as sul2, a blaOXA-164-like gene, floR, tetM, and multiple novel tet(X3) variants with different tandem structures. It is of paramount significance that their mechanism may transfer to other Gram-negative pathogens, even if their tandem structures have no cumulative effect on tigecycline resistance.
Collapse
Affiliation(s)
- Yumeng Cheng
- College of Animal Science and Technology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Yakun Li
- College of Life Science and Technology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Runhao Yu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Mingxiang Ma
- College of Animal Science and Technology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Meng Yang
- College of Animal Science and Technology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| | - Hongbin Si
- College of Animal Science and Technology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
| |
Collapse
|
6
|
Transcriptome analysis of sRNA responses to four different antibiotics in Pseudomonas aeruginosa PAO1. Microb Pathog 2022; 173:105865. [DOI: 10.1016/j.micpath.2022.105865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
|
7
|
Kotecka K, Kawalek A, Modrzejewska-Balcerek M, Gawor J, Zuchniewicz K, Gromadka R, Bartosik AA. Functional Characterization of TetR-like Transcriptional Regulator PA3973 from Pseudomonas aeruginosa. Int J Mol Sci 2022; 23:ijms232314584. [PMID: 36498910 PMCID: PMC9736018 DOI: 10.3390/ijms232314584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Pseudomonas aeruginosa, a human opportunistic pathogen, is a common cause of nosocomial infections. Its ability to survive under different conditions relies on a complex regulatory network engaging transcriptional regulators controlling metabolic pathways and capabilities to efficiently use the available resources. P. aeruginosa PA3973 encodes an uncharacterized TetR family transcriptional regulator. In this study, we applied a transcriptome profiling (RNA-seq), genome-wide identification of binding sites using ChIP-seq, as well as the phenotype analyses to unravel the biological role of PA3973. Transcriptional profiling of P. aeruginosa PAO1161 overexpressing PA3973 showed changes in the mRNA level of 648 genes. Concomitantly, ChIP-seq analysis identified more than 300 PA3973 binding sites in the P. aeruginosa genome. A 13 bp sequence motif was indicated as the binding site of PA3973. The PA3973 regulon encompasses the PA3972-PA3971 genes encoding a probable acyl-CoA dehydrogenase and a thioesterase. In vitro analysis showed PA3973 binding to PA3973p. Accordingly, the lack of PA3973 triggered increased expression of PA3972 and PA3971. The ∆PA3972-71 PAO1161 strain demonstrated impaired growth in the presence of stress-inducing agents hydroxylamine or hydroxyurea, thus suggesting the role of PA3972-71 in pathogen survival upon stress. Overall our results showed that TetR-type transcriptional regulator PA3973 has multiple binding sites in the P. aeruginosa genome and influences the expression of diverse genes, including PA3972-PA3971, encoding proteins with a proposed role in stress response.
Collapse
|
8
|
Sung K, Park M, Chon J, Kweon O, Khan SA, Shen A, Paredes A. Concentration-Dependent Global Quantitative Proteome Response of Staphylococcus epidermidis RP62A Biofilms to Subinhibitory Tigecycline. Cells 2022; 11:3488. [PMID: 36359886 PMCID: PMC9655631 DOI: 10.3390/cells11213488] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 07/21/2023] Open
Abstract
Staphylococcus epidermidis is a leading cause of biofilm-associated infections on implanted medical devices. During the treatment of an infection, bacterial cells inside biofilms may be exposed to sublethal concentrations of the antimicrobial agents. In the present study, the effect of subinhibitory concentrations of tigecycline (TC) on biofilms formed by S. epidermidis strain RP62A was investigated using a quantitative global proteomic technique. Sublethal concentrations of TC [1/8 (T1) and 1/4 minimum inhibitory concentration (MIC) (T2)] promoted biofilm production in strain RP62A, but 1/2 MIC TC (T3) significantly inhibited biofilm production. Overall, 413, 429, and 518 proteins were differentially expressed in biofilms grown with 1/8 (T1), 1/4 (T2), and 1/2 (T3) MIC of TC, respectively. As the TC concentration increased, the number of induced proteins in each Cluster of Orthologous Groups (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway increased. The TC concentration dependence of the proteome response highlights the diverse mechanisms of adaptive responses in strain RP62A biofilms. In both COG and KEGG functional analyses, most upregulated proteins belong to the metabolism pathway, suggesting that it may play an important role in the defense of strain RP62A biofilm cells against TC stress. Sub-MIC TC treatment of strain RP62A biofilms led to significant changes of protein expression related to biofilm formation, antimicrobial resistance, virulence, quorum sensing, ABC transporters, protein export, purine/pyrimidine biosynthesis, ribosomes, and essential proteins. Interestingly, in addition to tetracycline resistance, proteins involved in resistance of various antibiotics, including aminoglycosides, antimicrobial peptides, β-lactams, erythromycin, fluoroquinolones, fusidic acid, glycopeptides, lipopeptides, mupirocin, rifampicin and trimethoprim were differentially expressed. Our study demonstrates that global protein expression profiling of biofilm cells to antibiotic pressure may improve our understanding of the mechanisms of antibiotic resistance in biofilms.
Collapse
Affiliation(s)
- Kidon Sung
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Miseon Park
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Jungwhan Chon
- Companion Animal Health, Inje University, Gimhae 50834, Korea
| | - Ohgew Kweon
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Saeed A. Khan
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Andrew Shen
- Division of Neurotoxicology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Angel Paredes
- Office of Scientific Coordination, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| |
Collapse
|
9
|
Grace A, Sahu R, Owen DR, Dennis VA. Pseudomonas aeruginosa reference strains PAO1 and PA14: A genomic, phenotypic, and therapeutic review. Front Microbiol 2022; 13:1023523. [PMID: 36312971 PMCID: PMC9607943 DOI: 10.3389/fmicb.2022.1023523] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous, motile, gram-negative bacterium that has been recently identified as a multi-drug resistant pathogen in critical need of novel therapeutics. Of the approximately 5,000 strains, PAO1 and PA14 are common laboratory reference strains, modeling moderately and hyper-virulent phenotypes, respectively. PAO1 and PA14 have been instrumental in facilitating the discovery of novel drug targets, testing novel therapeutics, and supplying critical genomic information on the bacterium. While the two strains have contributed to a wide breadth of knowledge on the natural behaviors and therapeutic susceptibilities of P. aeruginosa, they have demonstrated significant deviations from observations in human infections. Many of these deviations are related to experimental inconsistencies in laboratory strain environment that complicate and, at times, terminate translation from laboratory results to clinical applications. This review aims to provide a comparative analysis of the two strains and potential methods to improve their clinical relevance.
Collapse
Affiliation(s)
- Amber Grace
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | | | - Vida A. Dennis
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
- *Correspondence: Vida A. Dennis,
| |
Collapse
|
10
|
Okeke ES, Chukwudozie KI, Nyaruaba R, Ita RE, Oladipo A, Ejeromedoghene O, Atakpa EO, Agu CV, Okoye CO. Antibiotic resistance in aquaculture and aquatic organisms: a review of current nanotechnology applications for sustainable management. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:69241-69274. [PMID: 35969340 PMCID: PMC9376131 DOI: 10.1007/s11356-022-22319-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/27/2022] [Indexed: 05/13/2023]
Abstract
Aquaculture has emerged as one of the world's fastest-growing food industries in recent years, helping food security and boosting global economic status. The indiscriminate disposal of untreated or improperly managed waste and effluents from different sources including production plants, food processing sectors, and healthcare sectors release various contaminants such as bioactive compounds and unmetabolized antibiotics, and antibiotic-resistant organisms into the environment. These emerging contaminants (ECs), especially antibiotics, have the potential to pollute the environment, particularly the aquatic ecosystem due to their widespread use in aquaculture, leading to various toxicological effects on aquatic organisms as well as long-term persistence in the environment. However, various forms of nanotechnology-based technologies are now being explored to assist other remediation technologies to boost productivity, efficiency, and sustainability. In this review, we critically highlighted several ecofriendly nanotechnological methods including nanodrug and vaccine delivery, nanoformulations, and nanosensor for their antimicrobial effects in aquaculture and aquatic organisms, potential public health risks associated with nanoparticles, and their mitigation measures for sustainable management.
Collapse
Affiliation(s)
- Emmanuel Sunday Okeke
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 41000, Enugu State, Nigeria
- Natural Science Unit, School of General Studies, University of Nigeria, Nsukka, 41000, Enugu State, Nigeria
- Institute of Environmental Health and Ecological Security, School of Environment & Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
| | - Kingsley Ikechukwu Chukwudozie
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Nigeria
- Department of Clinical Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Raphael Nyaruaba
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Center for Biosafety Megascience, Wuhan Institute of Virology, CAS, Wuhan, China
| | - Richard Ekeng Ita
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Ritman University, Ikot Ekpene, Akwa Ibom State, Nigeria
| | - Abiodun Oladipo
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037, Jiangsu, People's Republic of China
| | - Onome Ejeromedoghene
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- School of Chemistry and Chemical Engineering, Southeast University, Jiangning District, Nanjing, Jiangsu Province, 211189, People's Republic of China
| | - Edidiong Okokon Atakpa
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Institute of Marine Biology & Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
- Department of Animal & Environmental Biology, University of Uyo, Uyo, 1017, Akwa Ibom State, Nigeria
| | | | - Charles Obinwanne Okoye
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya.
- Department of Zoology & Environmental Biology, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Nigeria.
- School of Environment & Safety Engineering, Biofuels Institute, Jiangsu University, Zhenjiang, 212013, China.
- Key Laboratory of Intelligent Agricultural Machinery Equipment, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
11
|
Jadimurthy R, Mayegowda SB, Nayak S, Mohan CD, Rangappa KS. Escaping mechanisms of ESKAPE pathogens from antibiotics and their targeting by natural compounds. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2022; 34:e00728. [PMID: 35686013 PMCID: PMC9171455 DOI: 10.1016/j.btre.2022.e00728] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/10/2022] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
The microorganisms that have developed resistance to available therapeutic agents are threatening the globe and multidrug resistance among the bacterial pathogens is becoming a major concern of public health worldwide. Bacteria develop protective mechanisms to counteract the deleterious effects of antibiotics, which may eventually result in loss of growth-inhibitory potential of antibiotics. ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) pathogens display multidrug resistance and virulence through various mechanisms and it is the need of the hour to discover or design new antibiotics against ESKAPE pathogens. In this article, we have discussed the mechanisms acquired by ESKAPE pathogens to counteract the effect of antibiotics and elaborated on recently discovered secondary metabolites derived from bacteria and plant sources that are endowed with good antibacterial activity towards pathogenic bacteria in general, ESKAPE organisms in particular. Abyssomicin C, allicin, anthracimycin, berberine, biochanin A, caffeic acid, daptomycin, kibdelomycin, piperine, platensimycin, plazomicin, taxifolin, teixobactin, and thymol are the major metabolites whose antibacterial potential have been discussed in this article.
Collapse
Affiliation(s)
- Ragi Jadimurthy
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Shilpa Borehalli Mayegowda
- Dayananda Sagar University, School of Basic and Applied Sciences, Shavige Malleswara Hills, Kumaraswamy layout, Bengaluru 560111, India
| | - S.Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore 570006, India
| | | | | |
Collapse
|
12
|
Synergistic Activity of Imipenem in Combination with Ceftazidime/Avibactam or Avibactam against Non-MBL-Producing Extensively Drug-Resistant Pseudomonas aeruginosa. Microbiol Spectr 2022; 10:e0274021. [PMID: 35315696 PMCID: PMC9045292 DOI: 10.1128/spectrum.02740-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Extensively drug-resistant Pseudomonas aeruginosa (XDRPA) infection is a significant public health threat due to a lack of effective therapeutic options. New β-lactam-β-lactamase inhibitor combinations, including ceftazidime-avibactam (CZA), have shown a high resistance rate to XDRPA. This study was therefore conducted to describe the underlying genomic mechanism of resistance for CZA nonsusceptible XDRPA strains that are non-metallo-β-lactamase (MBL) producers as well as to examine synergism of CZA and other antipseudomonal agents. Furthermore, the synergistic antibacterial activity of the most effective antimicrobial combination against non-MBL-producing XDRPA was evaluated through in vitro experiments. The resistance profiles of 15 CZA-resistant XDRPA strains isolated from clinical specimens in China-Japan Friendship Hospital between January 2017 to December 2020 were obtained by whole-genome sequencing (WGS) analysis. MBL genes blaIMP-1 and blaIMP-45 were found in 2 isolates (2/15, 13.3%); the other underlying CZA-resistance mechanisms involved the decreased OprD porin (13/13), blaAmpC overexpression (8/13) or mutation (13/13), and upregulated efflux pumps (13/13). CZA-imipenem (CZA-IPM) combination was identified to be the most effective against non-MBL-producing XDRPA according to the results of WGS analysis and combined antimicrobial susceptibility tests, with an approximately 16.62-fold reduction in MICs compared to CZA alone. Furthermore, the results of checkerboard analysis and growth curve displayed the synergistic antimicrobial activity of CZA and IPM against non-MBL-producing XDRPA. Electron microscopy also revealed that CZA-IPM combination might lead to more cellular structural alterations than CZA or IPM alone. This study suggested that the CZA-IPM combination has potential for non-MBL-producing XDRPA with blaAmpC overexpression or mutation, decreased OprD porin, and upregulated efflux pumps. IMPORTANCE Handling the infections by extensively drug-resistant Pseudomonas aeruginosa (XDRPA) strains is challenging due to their complicated antibiotic resistance mechanisms in immunosuppressed patients with pulmonary diseases (e.g., cystic fibrosis, chronic obstructive pulmonary disease, and lung transplant), ventilator-associated pneumonia, and bloodstream infections. The current study suggested the potentiality of the ceftazidime-avibactam-imipenem combination against XDRPA with blaAmpC overexpression or mutation, decreased OprD porin, and/or upregulated efflux pumps. Our findings indicate the necessity of combined drug sensitivity tests against XDRPA and also lay a foundation for the development of prevention, control, and treatment strategies in XDRPA infections.
Collapse
|
13
|
Marcoleta AE, Arros P, Varas MA, Costa J, Rojas-Salgado J, Berríos-Pastén C, Tapia-Fuentes S, Silva D, Fierro J, Canales N, Chávez FP, Gaete A, González M, Allende ML, Lagos R. The highly diverse Antarctic Peninsula soil microbiota as a source of novel resistance genes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 810:152003. [PMID: 34856283 DOI: 10.1016/j.scitotenv.2021.152003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
The rise of multiresistant bacterial pathogens is currently one of the most critical threats to global health, encouraging a better understanding of the evolution and spread of antimicrobial resistance. In this regard, the role of the environment as a source of resistance mechanisms remains poorly understood. Moreover, we still know a minimal part of the microbial diversity and resistome present in remote and extreme environments, hosting microbes that evolved to resist harsh conditions and thus a potentially rich source of novel resistance genes. This work demonstrated that the Antarctic Peninsula soils host a remarkable microbial diversity and a widespread presence of autochthonous antibiotic-resistant bacteria and resistance genes. We observed resistance to a wide array of antibiotics among isolates, including Pseudomonas resisting ten or more different compounds, with an overall increased resistance in bacteria from non-intervened areas. In addition, genome analysis of selected isolates showed several genes encoding efflux pumps, as well as a lack of known resistance genes for some of the resisted antibiotics, including colistin, suggesting novel uncharacterized mechanisms. By combining metagenomic approaches based on analyzing raw reads, assembled contigs, and metagenome-assembled genomes, we found hundreds of widely distributed genes potentially conferring resistance to different antibiotics (including an outstanding variety of inactivation enzymes), metals, and biocides, hosted mainly by Polaromonas, Pseudomonas, Streptomyces, Variovorax, and Burkholderia. Furthermore, a proportion of these genes were found inside predicted plasmids and other mobile elements, including a putative OXA-like carbapenemase from Polaromonas harboring conserved key residues and predicted structural features. All this evidence indicates that the Antarctic Peninsula soil microbiota has a broad natural resistome, part of which could be transferred horizontally to pathogenic bacteria, acting as a potential source of novel resistance genes.
Collapse
Affiliation(s)
- Andrés E Marcoleta
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.
| | - Patricio Arros
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Macarena A Varas
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - José Costa
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Johanna Rojas-Salgado
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Camilo Berríos-Pastén
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Sofía Tapia-Fuentes
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Daniel Silva
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - José Fierro
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Nicolás Canales
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Francisco P Chávez
- Laboratorio de Microbiología de Sistemas, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Alexis Gaete
- Laboratorio de Bioinformática y Expresión Génica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile; FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Mauricio González
- Laboratorio de Bioinformática y Expresión Génica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Miguel L Allende
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Rosalba Lagos
- Grupo de Microbiología Integrativa, Laboratorio de Biología Estructural y Molecular BEM, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
14
|
Antimicrobial Treatment Options for Difficult-to-Treat Resistant Gram-Negative Bacteria Causing Cystitis, Pyelonephritis, and Prostatitis: A Narrative Review. Drugs 2022; 82:407-438. [PMID: 35286622 PMCID: PMC9057390 DOI: 10.1007/s40265-022-01676-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2022] [Indexed: 02/06/2023]
Abstract
Urinary tract infections, including cystitis, acute pyelonephritis, and prostatitis, are among the most common diagnoses prompting antibiotic prescribing. The rise in antimicrobial resistance over the past decades has led to the increasing challenge of urinary tract infections because of multidrug-resistant and "difficult-to-treat resistance" among Gram-negative bacteria. Recent advances in pharmacotherapy and medical microbiology are modernizing how these urinary tract infections are treated. Advances in pharmacotherapy have included not only the development and approval of novel antibiotics, such as ceftazidime/avibactam, meropenem/vaborbactam, imipenem/relebactam, ceftolozane/tazobactam, cefiderocol, plazomicin, and glycylcyclines, but also the re-examination of the potential role of legacy antibiotics, including older aminoglycosides and tetracyclines. Recent advances in medical microbiology allow phenotypic and molecular mechanism of resistance testing, and thus antibiotic prescribing can be tailored to the mechanism of resistance in the infecting pathogen. Here, we provide a narrative review on the clinical and pre-clinical studies of drugs that can be used for difficult-to-treat resistant Gram-negative bacteria, with a particular focus on data relevant to the urinary tract. We also offer a pragmatic framework for antibiotic selection when encountering urinary tract infections due to difficult-to-treat resistant Gram-negative bacteria based on the organism and its mechanism of resistance.
Collapse
|
15
|
Jones F, Hu Y, Coates A. The Efficacy of Using Combination Therapy against Multi-Drug and Extensively Drug-Resistant Pseudomonas aeruginosa in Clinical Settings. Antibiotics (Basel) 2022; 11:323. [PMID: 35326786 PMCID: PMC8944682 DOI: 10.3390/antibiotics11030323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative bacterium which is capable of developing a high level of antibiotic resistance. It has been placed on the WHO's critical priority pathogen list and it is commonly found in ventilator-associated pneumonia infections, blood stream infections and other largely hospital-acquired illnesses. These infections are difficult to effectively treat due to their increasing antibiotic resistance and as such patients are often treated with antibiotic combination regimens. METHODS We conducted a systematic search with screening criteria using the Ovid search engine and the Embase, Ovid Medline, and APA PsycInfo databases. RESULTS It was found that in many cases the combination therapies were able to match or outperform the monotherapies and none performed noticeably worse than the monotherapies. However, the clinical studies were mostly small, only a few were prospective randomized clinical trials and statistical significance was lacking. CONCLUSIONS It was concluded that combination therapies have a place in the treatment of these highly resistant bacteria and, in some cases, there is some evidence to suggest that they provide a more effective treatment than monotherapies.
Collapse
Affiliation(s)
| | | | - Anthony Coates
- Institute for Infection and Immunity, St George’s University of London, London SW17 0RE, UK; (F.J.); (Y.H.)
| |
Collapse
|
16
|
Garcia ÍR, de Oliveira Garcia FA, Pereira PS, Coutinho HDM, Siyadatpanah A, Norouzi R, Wilairatana P, de Lourdes Pereira M, Nissapatorn V, Tintino SR, Rodrigues FFG. Microbial resistance: The role of efflux pump superfamilies and their respective substrates. Life Sci 2022; 295:120391. [PMID: 35149116 DOI: 10.1016/j.lfs.2022.120391] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 12/24/2022]
Abstract
The microorganism resistance to antibiotics has become one of the most worrying issues for science due to the difficulties related to clinical treatment and the rapid spread of diseases. Efflux pumps are classified into six groups of carrier proteins that are part of the different types of mechanisms that contribute to resistance in microorganisms, allowing their survival. The present study aimed to carry out a bibliographic review on the superfamilies of carriers in order to understand their compositions, expressions, substrates, and role in intrinsic resistance. At first, a search for manuscripts was carried out in the databases Medline, Pubmed, ScienceDirect, and Scielo, using as descriptors: efflux pump, expression, pump inhibitors and efflux superfamily. For article selection, two criteria were taken into account: for inclusion, those published between 2000 and 2020, including textbooks, and for exclusion, duplicates and academic collections. In this research, 139,615 published articles were obtained, with 312 selected articles and 7 book chapters that best met the aim. From the comprehensive analysis, it was possible to consider that the chromosomes and genetic elements can contain genes encoding efflux pumps and are responsible for multidrug resistance. Even though this is a well-explored topic in the scientific community, understanding the behavior of antibiotics as substrates that increase the expression of pump-encoding genes has challenged medicine. This review study succinctly summarizes the most relevant features of these systems, as well as their contribution to multidrug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Abolghasem Siyadatpanah
- Ferdows School of Paramedical and Health, Birjand University of Medical Sciences, Birjand, Iran
| | - Roghayeh Norouzi
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials & Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences and Research Excellence Center for Innovation and Health, Walailak University, Thailand
| | | | | |
Collapse
|
17
|
Evaluation of Heterocyclic Carboxamides as Potential Efflux Pump Inhibitors in Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 11:antibiotics11010030. [PMID: 35052908 PMCID: PMC8772707 DOI: 10.3390/antibiotics11010030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/19/2022] Open
Abstract
The ability to rescue the activity of antimicrobials that are no longer effective against bacterial pathogens such as Pseudomonas aeruginosa is an attractive strategy to combat antimicrobial drug resistance. Herein, novel efflux pump inhibitors (EPIs) demonstrating strong potentiation in combination with levofloxacin against wild-type P. aeruginosa ATCC 27853 are presented. A structure activity relationship of aryl substituted heterocyclic carboxamides containing a pentane diamine side chain is described. Out of several classes of fused heterocyclic carboxamides, aryl indole carboxamide compound 6j (TXA01182) at 6.25 µg/mL showed 8-fold potentiation of levofloxacin. TXA01182 was found to have equally synergistic activities with other antimicrobial classes (monobactam, fluoroquinolones, sulfonamide and tetracyclines) against P. aeruginosa. Several biophysical and genetic studies rule out membrane disruption and support efflux inhibition as the mechanism of action (MOA) of TXA01182. TXA01182 was determined to lower the frequency of resistance (FoR) of the partner antimicrobials and enhance the killing kinetics of levofloxacin. Furthermore, TXA01182 demonstrated a synergistic effect with levofloxacin against several multidrug resistant P. aeruginosa clinical isolates.
Collapse
|
18
|
The Development of Third-Generation Tetracycline Antibiotics and New Perspectives. Pharmaceutics 2021; 13:pharmaceutics13122085. [PMID: 34959366 PMCID: PMC8707899 DOI: 10.3390/pharmaceutics13122085] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 01/04/2023] Open
Abstract
The tetracycline antibiotic class has acquired new valuable members due to the optimisation of the chemical structure. The first modern tetracycline introduced into therapy was tigecycline, followed by omadacycline, eravacycline, and sarecycline (the third generation). Structural and physicochemical key elements which led to the discovery of modern tetracyclines are approached. Thus, several chemical subgroups are distinguished, such as glycylcyclines, aminomethylcyclines, and fluorocyclines, which have excellent development potential. The antibacterial spectrum comprises several resistant bacteria, including those resistant to old tetracyclines. Sarecycline, a narrow-spectrum tetracycline, is notable for being very effective against Cutinebacterium acnes. The mechanism of antibacterial action from the perspective of the new compound is approached. Several severe bacterial infections are treated with tigecycline, omadacycline, and eravacycline (with parenteral or oral formulations). In addition, sarecycline is very useful in treating acne vulgaris. Tetracyclines also have other non-antibiotic properties that require in-depth studies, such as the anti-inflammatory effect effect of sarecycline. The main side effects of modern tetracyclines are described in accordance with published clinical studies. Undoubtedly, this class of antibiotics continues to arouse the interest of researchers. As a result, new derivatives are developed and studied primarily for the antibiotic effect and other biological effects.
Collapse
|
19
|
Kim DH, Kim JH. Efficacy of Tigecycline and Linezolid Against Pan-Drug-Resistant Bacteria Isolated From Companion Dogs in South Korea. Front Vet Sci 2021; 8:693506. [PMID: 34422945 PMCID: PMC8377367 DOI: 10.3389/fvets.2021.693506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
The emergence of multidrug-resistant bacteria in companion animals is an increasing concern in view of the concept of One Health. The antimicrobials linezolid (LZD) and tigecycline (TGC) are effective against multidrug-resistant bacteria isolated from humans; however, thus far, no previous study has evaluated the efficacy of these drugs against bacteria isolated from companion animals. This study aimed to evaluate the efficacy of LZD and TGC against bacteria that were isolated from companion dogs and showed resistance to all classes of antimicrobial agents. Clinical samples (auditory channel, eye, skin, and urine) were collected from dogs that visited the Veterinary Medical Teaching Hospital of Konkuk University (Seoul, South Korea) from October 2017 to September 2020. In total, 392 bacterial isolates were obtained, of which 85 were resistant to all classes of antimicrobial agents tested and were, therefore, considered potentially pan-drug resistant (PDR). The susceptibility of isolates to LZD and TGC was determined by the disk diffusion method and interpreted using the Clinical Laboratory Standards Institute guidelines. In total, 95.6% (43/45) and 97.8% (44/45) of gram-positive isolates were susceptible to LZD and TGC, respectively, whereas 82.5% (33/40) of gram-negative isolates were sensitive to TGC. In conclusion, both agents showed favorable efficacy, with the susceptibility rates for all potential PDR bacteria, except Pseudomonas spp., ranging from 72.7 to 100%. Thus, these drugs may serve as excellent antimicrobial options for veterinary medicine in the future.
Collapse
Affiliation(s)
| | - Jung-Hyun Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| |
Collapse
|
20
|
Kang D, Revtovich AV, Deyanov AE, Kirienko NV. Pyoverdine Inhibitors and Gallium Nitrate Synergistically Affect Pseudomonas aeruginosa. mSphere 2021; 6:e0040121. [PMID: 34133200 PMCID: PMC8265654 DOI: 10.1128/msphere.00401-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/26/2021] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is a multidrug-resistant, opportunistic pathogen that frequently causes ventilator-associated pneumonia in intensive care units and chronic lung infections in cystic fibrosis patients. The rising prevalence of drug-resistant bacteria demands the exploration of new therapeutic avenues for treating P. aeruginosa infections. Perhaps the most thoroughly explored alternative is to use novel treatments to target pathogen virulence factors, like biofilm or toxin production. Gallium(III) nitrate is one such agent. It has been recognized for its ability to inhibit pathogen growth and biofilm formation in P. aeruginosa by disrupting bacterial iron homeostasis. However, irreversible sequestration by pyoverdine substantially limits its effectiveness. In this report, we show that disrupting pyoverdine production (genetically or chemically) potentiates the efficacy of gallium nitrate. Interestingly, we report that the pyoverdine inhibitor 5-fluorocytosine primarily functions as an antivirulent, even when it indirectly affects bacterial growth in the presence of gallium, and that low selective pressure for resistance occurs. We also demonstrate that the antibiotic tetracycline inhibits pyoverdine at concentrations below those required to prevent bacterial growth, and this activity allows it to synergize with gallium to inhibit bacterial growth and rescue Caenorhabditis elegans during P. aeruginosa pathogenesis. IMPORTANCE P. aeruginosa is one of the most common causative agents for ventilator-associated pneumonia and nosocomial bacteremia and is a leading cause of death in patients with cystic fibrosis. Pandrug-resistant strains of P. aeruginosa are increasingly identified in clinical samples and show resistance to virtually all major classes of antibiotics, including aminoglycosides, cephalosporins, and carbapenems. Gallium(III) nitrate has received considerable attention as an antipseudomonal agent that inhibits P. aeruginosa growth and biofilm formation by disrupting bacterial iron homeostasis. This report demonstrates that biosynthetic inhibitors of pyoverdine, such as 5-fluorocytosine and tetracycline, synergize with gallium nitrate to inhibit P. aeruginosa growth and biofilm formation, rescuing C. elegans hosts during pathogenesis.
Collapse
Affiliation(s)
- Donghoon Kang
- Department of BioSciences, Rice University, Houston, Texas, USA
| | | | | | | |
Collapse
|
21
|
Kakoullis L, Papachristodoulou E, Chra P, Panos G. Mechanisms of Antibiotic Resistance in Important Gram-Positive and Gram-Negative Pathogens and Novel Antibiotic Solutions. Antibiotics (Basel) 2021; 10:415. [PMID: 33920199 PMCID: PMC8069106 DOI: 10.3390/antibiotics10040415] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 01/04/2023] Open
Abstract
Multidrug-resistant bacteria have on overwhelming impact on human health, as they cause over 670,000 infections and 33,000 deaths annually in the European Union alone. Of these, the vast majority of infections and deaths are caused by only a handful of species-multi-drug resistant Escherichia coli, Staphylococcus aureus, Pseudomonas aeruginosa, Enterococcus spp., Acinetobacter spp. and Klebsiella pneumoniae. These pathogens employ a multitude of antibiotic resistance mechanisms, such as the production of antibiotic deactivating enzymes, changes in antibiotic targets, or a reduction of intracellular antibiotic concentration, which render them insusceptible to multiple antibiotics. The purpose of this review is to summarize in a clinical manner the resistance mechanisms of each of these 6 pathogens, as well as the mechanisms of recently developed antibiotics designed to overcome them. Through a basic understanding of the mechanisms of antibiotic resistance, the clinician can better comprehend and predict resistance patterns even to antibiotics not reported on the antibiogram and can subsequently select the most appropriate antibiotic for the pathogen in question.
Collapse
Affiliation(s)
- Loukas Kakoullis
- Department of Respiratory Medicine, University General Hospital of Patras, 26504 Patras, Greece;
| | - Eleni Papachristodoulou
- Department of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece;
| | - Paraskevi Chra
- Department of Microbiology, Evangelismos Hospital, 10676 Athens, Greece;
| | - George Panos
- Department of Internal Medicine, Division of Infectious Diseases, University General Hospital of Patras, 26504 Patras, Greece
| |
Collapse
|
22
|
Hobson C, Chan AN, Wright GD. The Antibiotic Resistome: A Guide for the Discovery of Natural Products as Antimicrobial Agents. Chem Rev 2021; 121:3464-3494. [PMID: 33606500 DOI: 10.1021/acs.chemrev.0c01214] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The use of life-saving antibiotics has long been plagued by the ability of pathogenic bacteria to acquire and develop an array of antibiotic resistance mechanisms. The sum of these resistance mechanisms, the antibiotic resistome, is a formidable threat to antibiotic discovery, development, and use. The study and understanding of the molecular mechanisms in the resistome provide the basis for traditional approaches to combat resistance, including semisynthetic modification of naturally occurring antibiotic scaffolds, the development of adjuvant therapies that overcome resistance mechanisms, and the total synthesis of new antibiotics and their analogues. Using two major classes of antibiotics, the aminoglycosides and tetracyclines as case studies, we review the success and limitations of these strategies when used to combat the many forms of resistance that have emerged toward natural product-based antibiotics specifically. Furthermore, we discuss the use of the resistome as a guide for the genomics-driven discovery of novel antimicrobials, which are essential to combat the growing number of emerging pathogens that are resistant to even the newest approved therapies.
Collapse
Affiliation(s)
- Christian Hobson
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Andrew N Chan
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Gerard D Wright
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
23
|
Wilfred PM, Chandy SJ, Rebecca G, Satyendra S, Jasmine S. Intravenous colistin for the management of multidrug-resistant bacterial infections in Indian patients. Trop Doct 2021; 51:301-306. [PMID: 33491607 DOI: 10.1177/0049475520987777] [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] [Indexed: 11/17/2022]
Abstract
Multidrug-resistant Gram-negative bacterial infection is a serious global concern and especially in low and middle-income countries (LMIC) such as India. Colistin, an antimicrobial once abandoned following reports of organ toxicity, has re-emerged as an essential therapeutic agent in the management of these infections. A retrospective review of 162 inpatients was done, focusing on culture-proven multidrug-resistant infections requiring colistin. The overall clinical outcome in 58% of patients was found to be good, with nephrotoxicity and neurotoxicity occurring only in 8 (5%) and 4 (2.5%) patients, respectively. Multivariate analysis revealed an elevated lactate and raised urea to be independent factors associated with poor clinical response. In conclusion, there appears to be strong evidence supporting the use of colistin in the management of multidrug-resistant Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Premila M Wilfred
- Resident, Department of Pharmacology and Clinical Pharmacology, Christian Medical College, Vellore, India
| | - Sujith J Chandy
- Resident, Department of Pharmacology and Clinical Pharmacology, Christian Medical College, Vellore, India
| | - Grace Rebecca
- Tutor, Department of Biostatistics, Christian Medical College, Vellore, India
| | - Sowmya Satyendra
- Professor, Department of Medicine, Christian Medical College, Vellore, India
| | - Sudha Jasmine
- Professor, Department of Medicine, Christian Medical College, Vellore, India
| |
Collapse
|
24
|
Yaghoubi S, Zekiy AO, Krutova M, Gholami M, Kouhsari E, Sholeh M, Ghafouri Z, Maleki F. Tigecycline antibacterial activity, clinical effectiveness, and mechanisms and epidemiology of resistance: narrative review. Eur J Clin Microbiol Infect Dis 2021; 41:1003-1022. [PMID: 33403565 PMCID: PMC7785128 DOI: 10.1007/s10096-020-04121-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022]
Abstract
Tigecycline is unique glycylcycline class of semisynthetic antimicrobial agents developed for the treatment of polymicrobial infections caused by multidrug-resistant Gram-positive and Gram-negative pathogens. Tigecycline evades the main tetracycline resistance genetic mechanisms, such as tetracycline-specific efflux pump acquisition and ribosomal protection, via the addition of a glycyclamide moiety to the 9-position of minocycline. The use of the parenteral form of tigecycline is approved for complicated skin and skin structure infections (excluding diabetes foot infection), complicated intra-abdominal infections, and community-acquired bacterial pneumonia in adults. New evidence also suggests the effectiveness of tigecycline for the treatment of severe Clostridioides difficile infections. Tigecycline showed in vitro susceptibility to Coxiella spp., Rickettsia spp., and multidrug-resistant Neisseria gonnorrhoeae strains which indicate the possible use of tigecycline in the treatment of infections caused by these pathogens. Except for intrinsic, or often reported resistance in some Gram-negatives, tigecycline is effective against a wide range of multidrug-resistant nosocomial pathogens. Herein, we summarize the currently available data on tigecycline pharmacokinetics and pharmacodynamics, its mechanism of action, the epidemiology of tigecycline resistance, and its clinical effectiveness.
Collapse
Affiliation(s)
- Sajad Yaghoubi
- Department of Clinical Microbiology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Russian Federation, Trubetskaya st., 8-2, 119991, Moscow, Russia
| | - Marcela Krutova
- Department of Medical Microbiology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Mehrdad Gholami
- Department of Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ebrahim Kouhsari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, P.O. Box 6939177143, Gorgan- Sari Road, Golestan Province, Gorgan, Iran. .,Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, P.O. Box 6939177143, Gorgan- Sari Road, Golestan Province, Gorgan, Iran.
| | - Mohammad Sholeh
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Ghafouri
- Department of Biochemistry, Biophysics and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Farajolah Maleki
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ilam University of Medical sciences, Ilam, Iran.
| |
Collapse
|
25
|
Mohseni N, Rad M, Mokhtari AR, Yahyaraeyat R, Zahraie Salehi T. Evaluation of MexAB-OprM efflux pump and determination of antimicrobial susceptibility in Pseudomonas aeruginosa human and veterinary isolates. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2021. [DOI: 10.15547/bjvm.2019-0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pseudomonas aeruginosa could cause serious infections in hospitals and is highlighted as a source of financial problems in farms. The revelation of drug resistant, particularly multi-drug resistant (MDR) P. aeruginosa is important around the world. The efflux pump activity is identified as one of the most important intrinsic resistant mechanisms in P. aeruginosa. A total of 96 P. aeruginosa isolates from inpatient and animal sources were tested for antimicrobial susceptibility and subjected to multiplex PCR (mPCR) assay to detect MexAB-OprM efflux pump system. The human isolates have shown the highest resistance against cefazolin, ampicillin, nalidixic acid, trimethoprim/sulfamethoxazole, cephalothin, oxacillin (100%). All farm animal isolates were resistant to cefazolin, kanamycin, amoxicillin clavulanic acid, and cephalothin (100%). In both isolate groups, the presence of MexA was more common than that of MexB. MexAB-OprM was demonstrated as a valuable mechanism in P. aeruginosa antimicrobial resistant strains.
Collapse
|
26
|
Li L, Hassan KA, Tetu SG, Naidu V, Pokhrel A, Cain AK, Paulsen IT. The Transcriptomic Signature of Tigecycline in Acinetobacter baumannii. Front Microbiol 2020; 11:565438. [PMID: 33193153 PMCID: PMC7652931 DOI: 10.3389/fmicb.2020.565438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/08/2020] [Indexed: 11/14/2022] Open
Abstract
Tigecycline, a protein translation inhibitor, is a treatment of last resort for infections caused by the opportunistic multidrug resistance human pathogen Acinetobacter baumannii. However, strains resistant to tigecycline were reported not long after its clinical introduction. Translation inhibitor antibiotics perturb ribosome function and induce the reduction of (p)ppGpp, an alarmone involved in the stringent response that negatively modulates ribosome production. Through RNA sequencing, this study revealed a significant reduction in the transcription of genes in citric acid cycle and cell respiration, suggesting tigecycline inhibits or slows down bacterial growth. Our results indicated that the drug-induced reduction of (p)ppGpp level promoted the production but diminished the degradation of ribosomes, which mitigates the translational inhibition effect by tigecycline. The reduction of (p)ppGpp also led to a decrease of transcription coupled nucleotide excision repair which likely increases the chances of development of tigecycline resistant mutants. Increased expression of genes linked to horizontal gene transfer were also observed. The most upregulated gene, rtcB, involving in RNA repair, is either a direct tigecycline stress response or is in response to the transcription de-repression of a toxin-antitoxin system. The most down-regulated genes encode two β-lactamases, which is a possible by-product of tigecycline-induced reduction in transcription of genes associated with peptidoglycan biogenesis. This transcriptomics study provides a global genetic view of why A. baumannii is able to rapidly develop tigecycline resistance.
Collapse
Affiliation(s)
- Liping Li
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | - Karl A Hassan
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Sasha G Tetu
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Varsha Naidu
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | - Alaska Pokhrel
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | - Amy K Cain
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Ian T Paulsen
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
27
|
Characterizing the Mechanism of Action of an Ancient Antimicrobial, Manuka Honey, against Pseudomonas aeruginosa Using Modern Transcriptomics. mSystems 2020; 5:5/3/e00106-20. [PMID: 32606022 PMCID: PMC7329319 DOI: 10.1128/msystems.00106-20] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Manuka honey has broad-spectrum antimicrobial activity, and unlike traditional antibiotics, resistance to its killing effects has not been reported. However, its mechanism of action remains unclear. Here, we investigated the mechanism of action of manuka honey and its key antibacterial components using a transcriptomic approach in a model organism, Pseudomonas aeruginosa We show that no single component of honey can account for its total antimicrobial action, and that honey affects the expression of genes in the SOS response, oxidative damage, and quorum sensing. Manuka honey uniquely affects genes involved in the explosive cell lysis process and in maintaining the electron transport chain, causing protons to leak across membranes and collapsing the proton motive force, and it induces membrane depolarization and permeabilization in P. aeruginosa These data indicate that the activity of manuka honey comes from multiple mechanisms of action that do not engender bacterial resistance.IMPORTANCE The threat of antimicrobial resistance to human health has prompted interest in complex, natural products with antimicrobial activity. Honey has been an effective topical wound treatment throughout history, predominantly due to its broad-spectrum antimicrobial activity. Unlike traditional antibiotics, honey-resistant bacteria have not been reported; however, honey remains underutilized in the clinic in part due to a lack of understanding of its mechanism of action. Here, we demonstrate that honey affects multiple processes in bacteria, and this is not explained by its major antibacterial components. Honey also uniquely affects bacterial membranes, and this can be exploited for combination therapy with antibiotics that are otherwise ineffective on their own. We argue that honey should be included as part of the current array of wound treatments due to its effective antibacterial activity that does not promote resistance in bacteria.
Collapse
|
28
|
Gasparrini AJ, Markley JL, Kumar H, Wang B, Fang L, Irum S, Symister CT, Wallace M, Burnham CAD, Andleeb S, Tolia NH, Wencewicz TA, Dantas G. Tetracycline-inactivating enzymes from environmental, human commensal, and pathogenic bacteria cause broad-spectrum tetracycline resistance. Commun Biol 2020; 3:241. [PMID: 32415166 PMCID: PMC7229144 DOI: 10.1038/s42003-020-0966-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/17/2020] [Indexed: 12/19/2022] Open
Abstract
Tetracycline resistance by antibiotic inactivation was first identified in commensal organisms but has since been reported in environmental and pathogenic microbes. Here, we identify and characterize an expanded pool of tet(X)-like genes in environmental and human commensal metagenomes via inactivation by antibiotic selection of metagenomic libraries. These genes formed two distinct clades according to habitat of origin, and resistance phenotypes were similarly correlated. Each gene isolated from the human gut encodes resistance to all tetracyclines tested, including eravacycline and omadacycline. We report a biochemical and structural characterization of one enzyme, Tet(X7). Further, we identify Tet(X7) in a clinical Pseudomonas aeruginosa isolate and demonstrate its contribution to tetracycline resistance. Lastly, we show anhydrotetracycline and semi-synthetic analogues inhibit Tet(X7) to prevent enzymatic tetracycline degradation and increase tetracycline efficacy against strains expressing tet(X7). This work improves our understanding of resistance by tetracycline-inactivation and provides the foundation for an inhibition-based strategy for countering resistance.
Collapse
Affiliation(s)
- Andrew J Gasparrini
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jana L Markley
- Department of Chemistry, Washington University, St. Louis, MO, 63130, USA
| | - Hirdesh Kumar
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Luting Fang
- Department of Chemistry, Washington University, St. Louis, MO, 63130, USA
| | - Sidra Irum
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Chanez T Symister
- Department of Chemistry, Washington University, St. Louis, MO, 63130, USA
| | - Meghan Wallace
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Carey-Ann D Burnham
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Saadia Andleeb
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Niraj H Tolia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | | - Gautam Dantas
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Biomedical Engineering, Washington University, St. Louis, MO, 63130, USA.
| |
Collapse
|
29
|
Emergence of a Plasmid-Encoded Resistance-Nodulation-Division Efflux Pump Conferring Resistance to Multiple Drugs, Including Tigecycline, in Klebsiella pneumoniae. mBio 2020; 11:mBio.02930-19. [PMID: 32127452 PMCID: PMC7064769 DOI: 10.1128/mbio.02930-19] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In an era of increasing concerns about antimicrobial resistance, tigecycline is likely to have a critically important role in the treatment of carbapenem-resistant Enterobacteriaceae, the most problematic pathogens in human clinical settings—especially carbapenem-resistant K.pneumoniae. Here, we identified a new plasmid-borne RND-type tigecycline resistance determinant, TMexCD1-TOprJ1, which is widespread among K. pneumoniae isolates from food animals. tmexCD1-toprJ1 appears to have originated from the chromosome of a Pseudomonas species and may have been transferred onto plasmids by adjacent site-specific integrases. Although tmexCD1-toprJ1 still appears to be rare in human clinical isolates, considering the transferability of the tmexCD1-toprJ1 gene cluster and the broad substrate spectrum of TMexCD1-TOprJ1, further dissemination of this mobile tigecycline resistance determinant is possible. Therefore, from a “One Health” perspective, measures are urgently needed to monitor and control its further spread. The current low prevalence in human clinical isolates provides a precious time window to design and implement measures to tackle this. Transporters belonging to the chromosomally encoded resistance-nodulation-division (RND) superfamily mediate multidrug resistance in Gram-negative bacteria. However, the cotransfer of large gene clusters encoding RND-type pumps from the chromosome to a plasmid appears infrequent, and no plasmid-mediated RND efflux pump gene cluster has yet been found to confer resistance to tigecycline. Here, we identified a novel RND efflux pump gene cluster, designated tmexCD1-toprJ1, on plasmids from five pandrug-resistant Klebsiella pneumoniae isolates of animal origin. TMexCD1-TOprJ1 increased (by 4- to 32-fold) the MICs of tetracyclines (including tigecycline and eravacycline), quinolones, cephalosporins, and aminoglycosides for K.pneumoniae, Escherichia coli, and Salmonella. TMexCD1-TOprJ1 is closely related (64.5% to 77.8% amino acid identity) to the MexCD-OprJ efflux pump encoded on the chromosome of Pseudomonas aeruginosa. In an IncFIA plasmid, pHNAH8I, the tmexCD1-toprJ1 gene cluster lies adjacent to two genes encoding site-specific integrases, which may have been responsible for its acquisition. Expression of TMexCD1-TOprJ1 in E. coli resulted in increased tigecycline efflux and in K. pneumoniae negated the efficacy of tigecycline in an in vivo infection model. Expression of TMexCD1-TOprJ1 reduced the growth of E. coli and Salmonella but not K. pneumoniae. tmexCD1-toprJ1-positive Enterobacteriaceae isolates were rare in humans (0.08%) but more common in chicken fecal (14.3%) and retail meat (3.4%) samples. Plasmid-borne tmexCD1-toprJ1-like gene clusters were identified in sequences in GenBank from Enterobacteriaceae and Pseudomonas strains from multiple continents. The possibility of further global dissemination of the tmexCD1-toprJ1 gene cluster and its analogues in Enterobacteriaceae via plasmids may be an important consideration for public health planning.
Collapse
|
30
|
Yoon EJ, Oh Y, Jeong SH. Development of Tigecycline Resistance in Carbapenemase-Producing Klebsiella pneumoniae Sequence Type 147 via AcrAB Overproduction Mediated by Replacement of the ramA Promoter. Ann Lab Med 2020; 40:15-20. [PMID: 31432634 PMCID: PMC6713659 DOI: 10.3343/alm.2020.40.1.15] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/09/2019] [Accepted: 08/01/2019] [Indexed: 11/24/2022] Open
Abstract
Background Carbapenem-resistant K. pneumoniae 2297, isolated from a patient treated with tigecycline for pneumonia, developed tigecycline resistance, in contrast to carbapenem-resistant isolate 1215, which was collected four months prior to the 2297 isolate. Mechanisms underlying tigecycline resistance were elucidated for the clinical isolates. Methods The tigecycline minimum inhibitory concentration (MIC) was determined using the broth microdilution method, with or without phenylalanine-arginine β-naphthylamide (PABN), and whole-genome sequencing was carried out by single-molecule real-time sequencing. The expression levels of the genes acrA,oqxA,ramA,rarA, and rpoB were determined by reverse-transcription quantitative PCR. Results Both isolates presented identical antibiograms, except for tigecycline, which showed an MIC of 0.5 mg/L in 1215 and 2 mg/L in 2297. The addition of PABN to tigecycline-resistant 2297 caused a four-fold decrease in the tigecycline MIC to 0.5 mg/L, although acrA expression (encoding the AcrAB efflux pump) was upregulated by 2.5 fold and ramA expression (encoding the pump activator RamA) was upregulated by 1.4 fold. We identified a 6,096-bp fragment insertion flanking direct TATAT repeats that disrupted the romA gene located upstream of ramA in the chromosome of K. pneumoniae 2297; the insertion led the ramA gene promoter replacement resulting in stronger activation of the gene. Conclusions The K. pneumoniae isolate developed tigecycline resistance during tigecycline treatment. It was related to the overexpression of the AcrAB resistance-nodulation-cell division efflux system due to promoter replacement.
Collapse
Affiliation(s)
- Eun Jeong Yoon
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, Korea
| | - Yena Oh
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, Korea
| | - Seok Hoon Jeong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
31
|
Veeraraghavan B, Pragasam AK, Bakthavatchalam YD, Anandan S, Swaminathan S, Sundaram B. Colistin-sparing approaches with newer antimicrobials to treat carbapenem-resistant organisms: Current evidence and future prospects. Indian J Med Microbiol 2019; 37:72-90. [PMID: 31424014 DOI: 10.4103/ijmm.ijmm_19_215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Antimicrobial resistance is on the rise across the globe. Increasing incidence of infections due to carbapenem resistance organisms is becoming difficult to treat, due to the limited availability of therapeutic agents. Very few agents such as colistin, fosfomycin, tigecycline and minocycline are widely used, despite its toxicity. However, with the availability of novel antimicrobials, beta-lactam/beta-lactamase inhibitor-based and non-beta-lactam-based agents could be of great relief. This review covers three important aspects which include (i) current management of carbapenem-resistant infections, (ii) determination of specific types of carbapenemases produced by multidrug-resistant and extensively drug-resistant Gram-negative pathogens and (iii) the currently available novel beta-lactam/beta-lactamase inhibitors and non-beta-lactam-based agents' laboratory findings, clinical outcome and implications.
Collapse
Affiliation(s)
- Balaji Veeraraghavan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Agila Kumari Pragasam
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | | | - Shalini Anandan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | | | | |
Collapse
|
32
|
Tian Z, Liu R, Zhang H, Yang M, Zhang Y. Developmental dynamics of antibiotic resistome in aerobic biofilm microbiota treating wastewater under stepwise increasing tigecycline concentrations. ENVIRONMENT INTERNATIONAL 2019; 131:105008. [PMID: 31336253 DOI: 10.1016/j.envint.2019.105008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 06/10/2023]
Abstract
This study aimed to investigate the impact of tigecycline, the third generation tetracycline, on the antibiotic resistance development in environmental microbiota. Two biological contact oxidation reactors containing aerobic biofilm microbiota were constructed, one of which was constantly fed with synthetic wastewater spiked with increasing concentrations of tigecycline (0 to 25 mg/L) under a hydrolytic retention time of 24 h. Over a period of 636 days, chemical oxygen demand removal over 90% and complete nitrification were achieved for both the control and tigecycline-exposed reactors, and effluent tigecycline concentrations in the tigecycline-exposed system were always <0.051 mg/L. Significant increases (p < 0.01) in resistome abundance and resistant bacteria ratio were detected at a tigecycline dose of 10 and 25 mg/L, respectively, revealed by metagenomic sequencing and culture-based method. The increase of resistome in the tigecycline system was mainly attributed to the enrichment of tetX, one cooperative tetracycline degrading gene. Partial canonical correspondence analysis showed that the change of resistome was mainly driven by bacterial community shift (vertical pathway). Network and genome binning analyses further suggested that the proliferation of Flavobacterium harboring tetX contributed to a relatively low community-wide resistance development in the aerobic biofilm microbiota under tigecycline selection by reducing the antibiotic concentration. This work provides scientific bases for the management and evaluation of the resistance risk induced by this novel antibiotic.
Collapse
Affiliation(s)
- Zhe Tian
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; National Engineering Laboratory for Industrial Wastewater Treatment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruyin Liu
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Zhang
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Min Yang
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; National Engineering Laboratory for Industrial Wastewater Treatment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yu Zhang
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; National Engineering Laboratory for Industrial Wastewater Treatment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
33
|
Kawalek A, Modrzejewska M, Zieniuk B, Bartosik AA, Jagura-Burdzy G. Interaction of ArmZ with the DNA-binding domain of MexZ induces expression of mexXY multidrug efflux pump genes and antimicrobial resistance in Pseudomonas aeruginosa. Antimicrob Agents Chemother 2019; 63:AAC.01199-19. [PMID: 31527038 PMCID: PMC6879243 DOI: 10.1128/aac.01199-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/13/2019] [Indexed: 12/17/2022] Open
Abstract
Multidrug efflux pumps play an important role in antibiotic resistance in bacteria. In Pseudomonas aeruginosa, MexXY pump provides intrinsic resistance to many antimicrobials, including aminoglycosides. The expression of mexXY operon is negatively regulated by MexZ repressor. The repression is alleviated in response to the antibiotic-induced ribosome stress, which results in increased synthesis of anti-repressor ArmZ, interacting with MexZ. The molecular mechanism of MexZ inactivation by ArmZ is not known. Here, we showed that the N-terminal part of MexZ, encompassing the DNA-binding domain, is required for interaction with ArmZ. Using the bacterial two hybrid system based mutant screening and pull-down analyses we identified substitutions in MexZ that diminished (R3S, K6E, R13H) or completely impaired (K53E) the interaction with ArmZ without blocking MexZ activity as a transcriptional repressor. Introduction of corresponding mexZ missense mutations into P aeruginosa PAO1161 chromosome impaired (mexZ K6E, mexZ R13H) or blocked (mexZ K53E) tetracycline mediated induction of mexY expression. Concomitantly, PAO1161 mexZ K53E strain was more susceptible to aminoglycosides. The identified residues are highly conserved in MexZ-like transcriptional regulators found in bacterial genomes encoding both MexX/MexY/MexZ and ArmZ/PA5470 orthologs, suggesting that a similar mechanism may contribute to induction of efflux mediated resistance in other bacterial species. Overall, our data shed light on the molecular mechanism of ArmZ mediated induction of intrinsic antimicrobial resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Adam Kawalek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Department of Microbial Biochemistry, Warsaw, Poland
| | - Magdalena Modrzejewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Department of Microbial Biochemistry, Warsaw, Poland
| | - Bartlomiej Zieniuk
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, Warsaw, Poland
| | - Aneta Agnieszka Bartosik
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Department of Microbial Biochemistry, Warsaw, Poland
| | - Grazyna Jagura-Burdzy
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Department of Microbial Biochemistry, Warsaw, Poland
| |
Collapse
|
34
|
Benzalkonium Chlorides: Uses, Regulatory Status, and Microbial Resistance. Appl Environ Microbiol 2019; 85:AEM.00377-19. [PMID: 31028024 DOI: 10.1128/aem.00377-19] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Benzalkonium chlorides (BACs) are chemicals with widespread applications due to their broad-spectrum antimicrobial properties against bacteria, fungi, and viruses. This review provides an overview of the market for BACs, as well as regulatory measures and available data on safety, toxicity, and environmental contamination. We focus on the effect of frequent exposure of microbial communities to BACs and the potential for cross-resistant phenotypes to emerge. Toward this goal, we review BAC concentrations in consumer products, their correlation with the emergence of tolerance in microbial populations, and the associated risk potential. Our analysis suggests that the ubiquitous and frequent use of BACs in commercial products can generate selective environments that favor microbial phenotypes potentially cross-resistant to a variety of compounds. An analysis of benefits versus risks should be the guidepost for regulatory actions regarding compounds such as BACs.
Collapse
|
35
|
Pachori P, Gothalwal R, Gandhi P. Emergence of antibiotic resistance Pseudomonas aeruginosa in intensive care unit; a critical review. Genes Dis 2019; 6:109-119. [PMID: 31194018 PMCID: PMC6545445 DOI: 10.1016/j.gendis.2019.04.001] [Citation(s) in RCA: 274] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/09/2019] [Indexed: 12/29/2022] Open
Abstract
The emergence of antibiotic resistant bacteria in the healthcare is a serious concern. In the Healthcare premises precisely intensive care unit are major sources of microbial diversity. Recent findings have demonstrated not only microbial diversity but also drug resistant microbes largely habitat in ICU. Pseudomonas aeruginosa found as a part of normal intestinal flora and a significant pathogen responsible for wide range of ICU acquired infection in critically ill patients. Nosocomial infection associated with this organism including gastrointestinal infection, urinary tract infections and blood stream infection. Infection caused by this organism are difficult to treat because of the presence of its innate resistance to many antibiotics (β-lactam and penem group of antibiotics), and its ability to acquire further resistance mechanism to multiple class of antibiotics, including Beta-lactams, aminoglycosides and fluoroquinolones. In the molecular evolution microbes adopted several mechanism to maintain genomic plasticity. The tool microbe use for its survival is mainly biofilm formation, quorum sensing, and horizontal gene transfer and enzyme promiscuity. Such genomic plasticity provide an ideal habitat to grow and survive in hearse environment mainly antibiotics pressure. This review focus on infection caused by Pseudomonas aeruginosa, its mechanisms of resistance and available treatment options. The present study provides a systemic review on major source of Pseudomonas aeruginosa in ICU. Further, study also emphasizes virulence gene/s associated with Pseudomonas aeruginosa genome for extended drug resistance. Study gives detailed overview of antibiotic drug resistance mechanism.
Collapse
Affiliation(s)
- Preeti Pachori
- Department of Biotechnology, Barkatullah University, Bhopal 462026, Madhya Pradesh, India
| | - Ragini Gothalwal
- Department of Biotechnology, Barkatullah University, Bhopal 462026, Madhya Pradesh, India
| | - Puneet Gandhi
- Department of Research, Bhopal Memorial Hospital and Research Centre (BMHRC), Bhopal 462037, Madhya Pradesh, India
| |
Collapse
|
36
|
Idowu T, Arthur G, Zhanel GG, Schweizer F. Heterodimeric Rifampicin-Tobramycin conjugates break intrinsic resistance of Pseudomonas aeruginosa to doxycycline and chloramphenicol in vitro and in a Galleria mellonella in vivo model. Eur J Med Chem 2019; 174:16-32. [PMID: 31022550 DOI: 10.1016/j.ejmech.2019.04.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/01/2019] [Accepted: 04/13/2019] [Indexed: 11/25/2022]
Abstract
Intrinsic resistance in Pseudomonas aeruginosa, defined by chromosomally encoded low outer membrane permeability and constitutively over-expressed efflux pumps, is a major reason why the pathogen is refractory to many antibiotics. Herein, we report that heterodimeric rifampicin-tobramycin conjugates break this intrinsic resistance and sensitize multidrug and extensively drug-resistant P. aeruginosa to doxycycline and chloramphenicol in vitro and in vivo. Tetracyclines and chloramphenicol are model compounds for bacteriostatic effects, but when combined with rifampicin-tobramycin adjuvants, their effects became bactericidal at sub MIC levels. Potentiation of tetracyclines correlates with the SAR of this class of drugs and is consistent with outer membrane permeabilization and efflux pump inhibition. Overall, this strategy finds new uses for old drugs and presents an avenue to expand the therapeutic utility of legacy antibiotics to recalcitrant pathogens such as P. aeruginosa.
Collapse
Affiliation(s)
- Temilolu Idowu
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 1R9, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada; Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 1R9, Canada.
| |
Collapse
|
37
|
Hawkey PM, Warren RE, Livermore DM, McNulty CAM, Enoch DA, Otter JA, Wilson APR. Treatment of infections caused by multidrug-resistant Gram-negative bacteria: report of the British Society for Antimicrobial Chemotherapy/Healthcare Infection Society/British Infection Association Joint Working Party. J Antimicrob Chemother 2019. [PMID: 29514274 DOI: 10.1093/jac/dky027] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The Working Party makes more than 100 tabulated recommendations in antimicrobial prescribing for the treatment of infections caused by multidrug-resistant (MDR) Gram-negative bacteria (GNB) and suggest further research, and algorithms for hospital and community antimicrobial usage in urinary infection. The international definition of MDR is complex, unsatisfactory and hinders the setting and monitoring of improvement programmes. We give a new definition of multiresistance. The background information on the mechanisms, global spread and UK prevalence of antibiotic prescribing and resistance has been systematically reviewed. The treatment options available in hospitals using intravenous antibiotics and in primary care using oral agents have been reviewed, ending with a consideration of antibiotic stewardship and recommendations. The guidance has been derived from current peer-reviewed publications and expert opinion with open consultation. Methods for systematic review were NICE compliant and in accordance with the SIGN 50 Handbook; critical appraisal was applied using AGREE II. Published guidelines were used as part of the evidence base and to support expert consensus. The guidance includes recommendations for stakeholders (including prescribers) and antibiotic-specific recommendations. The clinical efficacy of different agents is critically reviewed. We found there are very few good-quality comparative randomized clinical trials to support treatment regimens, particularly for licensed older agents. Susceptibility testing of MDR GNB causing infection to guide treatment needs critical enhancements. Meropenem- or imipenem-resistant Enterobacteriaceae should have their carbapenem MICs tested urgently, and any carbapenemase class should be identified: mandatory reporting of these isolates from all anatomical sites and specimens would improve risk assessments. Broth microdilution methods should be adopted for colistin susceptibility testing. Antimicrobial stewardship programmes should be instituted in all care settings, based on resistance rates and audit of compliance with guidelines, but should be augmented by improved surveillance of outcome in Gram-negative bacteraemia, and feedback to prescribers. Local and national surveillance of antibiotic use, resistance and outcomes should be supported and antibiotic prescribing guidelines should be informed by these data. The diagnosis and treatment of both presumptive and confirmed cases of infection by GNB should be improved. This guidance, with infection control to arrest increases in MDR, should be used to improve the outcome of infections with such strains. Anticipated users include medical, scientific, nursing, antimicrobial pharmacy and paramedical staff where they can be adapted for local use.
Collapse
Affiliation(s)
- Peter M Hawkey
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | | | - Cliodna A M McNulty
- Microbiology Department, Gloucestershire Royal Hospital, Great Western Road, Gloucester GL1 3NN, UK
| | - David A Enoch
- Public Health England, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - A Peter R Wilson
- Department of Microbiology and Virology, University College London Hospitals, London, UK
| |
Collapse
|
38
|
Li J, Zhang H, Ning J, Sajid A, Cheng G, Yuan Z, Hao H. The nature and epidemiology of OqxAB, a multidrug efflux pump. Antimicrob Resist Infect Control 2019; 8:44. [PMID: 30834112 PMCID: PMC6387526 DOI: 10.1186/s13756-019-0489-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 02/03/2019] [Indexed: 01/03/2023] Open
Abstract
Background OqxAB efflux pump has been found to mediate multidrug resistance (MDR) in various bacteria over the past decades. The updates on the nature and epidemiology of OqxAB efflux pump need to be fully reviewed to broaden our understanding of this MDR determinant. Methods A literature search using the keyword of "oqxAB" was conducted in the online databases of Pubmed and ISI Web of Science with no restriction on the date of publication. The 87 publications were included into this review as references due to their close relevance to the nature and/or epidemiology of OqxAB efflux pump. Results The oqxAB gene generally locates on chromosome and/or plasmids flanked by IS26-like elements in clinical isolates of Enterobacteriaceae and Klebsiella pneumoniae, conferring low to intermediated resistance to quinoxalines, quinolones tigecycline, nitrofurantoin, several detergents and disinfectants (benzalkonium chloride, triclosan and SDS). It could co-spread with other antimicrobial resistance genes (bla CTX-M, rmtB and aac(6')-Ib etc.), virulence genes and heavy metal resistance genes (pco and sil operons). Both RarA (activator) and OqxR (repressor) play important roles on regulation of the expression of OqxAB. Conclusions The dissemination of oqxAB gene may pose a great risk on food safety and public health. Further investigation and understanding of the natural functions, horizontal transfer, and regulation mechanism of the OqxAB efflux pump will aid in future strategies of antimicrobial usage.
Collapse
Affiliation(s)
- Jun Li
- 1National Reference Laboratory of Veterinary Drug Residues (HZAU) and Key Laboratory of the Detection for Veterinary Drug Residues, Wuhan, 430070 Hubei China.,2Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014 Jiangsu China
| | - Heying Zhang
- 1National Reference Laboratory of Veterinary Drug Residues (HZAU) and Key Laboratory of the Detection for Veterinary Drug Residues, Wuhan, 430070 Hubei China
| | - Jianan Ning
- 1National Reference Laboratory of Veterinary Drug Residues (HZAU) and Key Laboratory of the Detection for Veterinary Drug Residues, Wuhan, 430070 Hubei China
| | - Abdul Sajid
- 1National Reference Laboratory of Veterinary Drug Residues (HZAU) and Key Laboratory of the Detection for Veterinary Drug Residues, Wuhan, 430070 Hubei China.,4College of Veterinary Sciences and Animal Husbandry, Abdul Wali Khan University Mardan, Mardan, KP Pakistan
| | - Guyue Cheng
- 3Laboratory of Quality & Safety Risk Assessment for Livestock and Poultry Products (Wuhan), Ministry of Agriculture, P.R China, Wuhan, 430070 Hubei China
| | - Zonghui Yuan
- 1National Reference Laboratory of Veterinary Drug Residues (HZAU) and Key Laboratory of the Detection for Veterinary Drug Residues, Wuhan, 430070 Hubei China.,3Laboratory of Quality & Safety Risk Assessment for Livestock and Poultry Products (Wuhan), Ministry of Agriculture, P.R China, Wuhan, 430070 Hubei China
| | - Haihong Hao
- 1National Reference Laboratory of Veterinary Drug Residues (HZAU) and Key Laboratory of the Detection for Veterinary Drug Residues, Wuhan, 430070 Hubei China.,3Laboratory of Quality & Safety Risk Assessment for Livestock and Poultry Products (Wuhan), Ministry of Agriculture, P.R China, Wuhan, 430070 Hubei China
| |
Collapse
|
39
|
Sanz-García F, Hernando-Amado S, Martínez JL. Mutational Evolution of Pseudomonas aeruginosa Resistance to Ribosome-Targeting Antibiotics. Front Genet 2018; 9:451. [PMID: 30405685 PMCID: PMC6200844 DOI: 10.3389/fgene.2018.00451] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/18/2018] [Indexed: 01/21/2023] Open
Abstract
The present work examines the evolutionary trajectories of replicate Pseudomonas aeruginosa cultures in presence of the ribosome-targeting antibiotics tobramycin and tigecycline. It is known that large number of mutations across different genes - and therefore a large number of potential pathways - may be involved in resistance to any single antibiotic. Thus, evolution toward resistance might, to a large degree, rely on stochasticity, which might preclude the use of predictive strategies for fighting antibiotic resistance. However, the present results show that P. aeruginosa populations evolving in parallel in the presence of antibiotics (either tobramycin or tigecycline) follow a set of trajectories that present common elements. In addition, the pattern of resistance mutations involved include common elements for these two ribosome-targeting antimicrobials. This indicates that mutational evolution toward resistance (and perhaps other properties) is to a certain degree deterministic and, consequently, predictable. These findings are of interest, not just for P. aeruginosa, but in understanding the general rules involved in the evolution of antibiotic resistance also. In addition, the results indicate that bacteria can evolve toward higher levels of resistance to antibiotics against which they are considered to be intrinsically resistant, as tigecycline in the case of P. aeruginosa and that this may confer cross-resistance to other antibiotics of therapeutic value. Our results are particularly relevant in the case of patients under empiric treatment with tigecycline, which frequently suffer P. aeruginosa superinfections.
Collapse
Affiliation(s)
| | - Sara Hernando-Amado
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - José L. Martínez
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
40
|
Ravithej Singh L, Tripathi VC, Raj S, Kumar A, Gupta S, Horam S, Upadhyay A, Kushwaha P, Arockiaraj J, Sashidhara KV, Pasupuleti M. In-house chemical library repurposing: A case example for Pseudomonas aeruginosa antibiofilm activity and quorum sensing inhibition. Drug Dev Res 2018; 79:383-390. [PMID: 30291767 DOI: 10.1002/ddr.21458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/03/2018] [Accepted: 08/05/2018] [Indexed: 02/01/2023]
Abstract
Hit, Lead & Candidate Discovery Drug repurposing has become a recent trend in drug development programs, where previously developed drugs are explored for hit and redeveloped into potential therapeutic agents for new diseases. Globally, in any drug development program, a series of molecules are synthesized and evaluated for the hypothesized activity. Hits are developed into lead molecules or drugs, whereas the negative ones are shelved in the lab with no immediate use. We in this project took the previously sidelined small chemical molecules to the next level of utility, where previously developed in-house small molecules library are tested for the unexplored biological relevant activity. As biofilm formation and quorum sensing play a vital role in bacterial pathogenesis, we believe that they could be one of the most effective targets for antimicrobial agents. In this study, we report the evaluation of 50 different compounds for anti-biofilm and anti-quorum sensing activity against Pseudomonas aeruginosa. Out of the screened compounds, three hydrazine-carboxamide hybrid derivatives showed promising anti-biofilm property and inhibition of pyocyanin production without any direct antimicrobial activity and cytotoxicity issues. Hydrazine-carboxamide hybrids can be a new class and promising leads for further anti-biofilm and anti-virulence development against microbial infections.
Collapse
Affiliation(s)
- L Ravithej Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vikash C Tripathi
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sneha Raj
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Anoop Kumar
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sampa Gupta
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Soyar Horam
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Akanksha Upadhyay
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Pragati Kushwaha
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Jesu Arockiaraj
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Chennai, Tamil Nadu, India
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mukesh Pasupuleti
- Microbiology Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
41
|
Update on prevalence and mechanisms of resistance to linezolid, tigecycline and daptomycin in enterococci in Europe: Towards a common nomenclature. Drug Resist Updat 2018; 40:25-39. [DOI: 10.1016/j.drup.2018.10.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 08/10/2018] [Accepted: 10/30/2018] [Indexed: 01/04/2023]
|
42
|
Zhao J, Liu Y, Liu Y, Wang D, Ni W, Wang R, Liu Y, Zhang B. Frequency and Genetic Determinants of Tigecycline Resistance in Clinically Isolated Stenotrophomonas maltophilia in Beijing, China. Front Microbiol 2018; 9:549. [PMID: 29632524 PMCID: PMC5879106 DOI: 10.3389/fmicb.2018.00549] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/12/2018] [Indexed: 01/27/2023] Open
Abstract
Stenotrophomonas maltophilia is an emerging nosocomial pathogen with high resistance to most clinically used antimicrobials. Tigecycline is a potential alternative antimicrobial for S. maltophilia infection treatment, but its resistance mechanism in clinical isolates is not fully elucidated. We investigated the antimicrobial susceptibility of 450 S. maltophilia isolated during 2012–2015 from three university hospitals in Beijing, China. These strains exhibited high susceptibility to minocycline (98.44%), sulfamethoxazole/trimethoprim (87.56%), tigecycline (77.78 %), doxycycline (81.33%), levofloxacin (67.56%), and ticarcillin/clavulanate (73.00%). The susceptibility of tigecycline-nonsusceptible strains (TNS) to doxycycline and levofloxacin was much lower than that of tigecycline-susceptible strains (TSS) (25.00% vs. 97.71% for doxycycline, P < 0.001; 17.00% vs. 82.00% for levofloxacin, P < 0.001). We further selected 48 TNS and TSS and compared the detection rate of eight tetracycline-specific genes by PCR and the expression level of six intrinsic multidrug resistance efflux pumps by real-time PCR. Only one tetB and two tetH genes in TNS and three tetH genes in TSS were detected, and the detection rate had no difference. The average expression level of smeD in TNS was higher than that in TSS [20.59 (11.53, 112.54) vs. 2.07 (0.80, 4.96), P < 0.001], while the average expression levels of smeA, smeI, smeO, smeV, and smrA were not significantly different, indicating that smeDEF was the predominant resistance genetic determinant in clinical S. maltophilia. Higher smeD expression was also observed in levofloxacin- and doxycycline-nonsusceptible isolates than in their corresponding susceptible isolates [16.46 (5.83, 102.24) vs. 2.72 (0.80, 6.25) for doxycycline, P < 0.001; 19.69 (8.07, 115.10) vs. 3.01(1.00, 6.03), P < 0.001], indicating that smeDEF was also the resistance genetic determinant to levofloxacin and doxycycline. The consistent resistance profile and common resistance genetic determinant highlight the importance of rational use of tigecycline for preventing the occurrence and spread of multidrug resistance.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Pulmonary and Critical Care Medicine, Air Force General Hospital of PLA, Beijing, China
| | - Yunxi Liu
- Department of Infection Management and Disease Control, Chinese PLA General Hospital, Beijing, China
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Air Force General Hospital of PLA, Beijing, China
| | - Dong Wang
- Department of Pulmonary and Critical Care Medicine, Air Force General Hospital of PLA, Beijing, China
| | - Wentao Ni
- Department of Respiratory Medicine, Chinese PLA General Hospital, Beijing, China
| | - Rui Wang
- Department of Clinical Pharmacology, Chinese PLA General Hospital, Beijing, China
| | - Youning Liu
- Department of Respiratory Medicine, Chinese PLA General Hospital, Beijing, China
| | - Bo Zhang
- Department of Pulmonary and Critical Care Medicine, Air Force General Hospital of PLA, Beijing, China
| |
Collapse
|
43
|
Genetic characterisation of tigecycline-resistant Enterobacter spp. in blood isolates causing bacteraemia. J Glob Antimicrob Resist 2018; 13:115-118. [PMID: 29307862 DOI: 10.1016/j.jgar.2017.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/21/2017] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES Tigecycline (TIG) is one of the most important antimicrobial agents used to treat infections by multidrug-resistant bacteria. However, rates of TIG-resistant pathogens have increased recently. This study was conducted to identify the antimicrobial susceptibility profiles and to investigate the role of efflux pumps in high-level TIG-resistant Enterobacter spp. isolates causing bacteraemia. METHODS A total of 323 Enterobacter spp. causing bacteraemia were collected from eight hospitals in various regions of South Korea. Minimum inhibitory concentrations (MICs) were determined by the broth microdilution method and Etest. Expression levels of the efflux pump gene acrA and its regulators (ramA and rarA) were examined by quantitative real-time PCR. Isolate relatedness was determined by multilocus sequence typing (MLST). RESULTS Among the 323 clinical isolates included in this study, 37 (11.5%) were TIG-non-susceptible, of which 8 isolates were highly resistant to TIG with MICs of 8mg/L (4 isolates) or 16mg/L (4 isolates). All high-level TIG-resistant isolates showed increased expression of acrA (0.93-13.3-fold) and ramA (1.4-8.2-fold). Isolates with a tigecycline MIC of 16mg/L also showed overexpression of rarA compared with TIG-susceptible isolates. CONCLUSIONS In this study, overexpression of acrA, ramA and rarA was observed in high-level TIG-resistant Enterobacter spp. isolates. We suggest that rarA might be involved in the regulation of acrA overexpression in high-level TIG-resistant Enterobacter spp. isolates. Efflux pump-mediated resistance should be closely monitored because it could be indirectly attributed to the use of other antibiotics transported by the same efflux pump.
Collapse
|
44
|
Deitchman AN, Singh RSP, Rand KH, Derendorf H. Enhanced in vitro activity of tigecycline in the presence of chelating agents. Int J Antimicrob Agents 2018; 51:799-802. [PMID: 29305959 DOI: 10.1016/j.ijantimicag.2017.12.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 12/14/2017] [Accepted: 12/23/2017] [Indexed: 10/18/2022]
Abstract
The lack of availability of novel antibiotic agents and the rise of resistance to existing therapies has led clinicians to utilise combination therapy to adequately treat bacterial infections. Here we examined how chelators may impact the in vitro activity of tigecycline (TIG) against Pseudomonas aeruginosa, Escherichia coli and Klebsiella pneumoniae. Minimum inhibitory concentrations (MICs) were determined by broth dilution with and without various combinations of chelators (EDTA and other tetracyclines) and metal ions (i.e. calcium, magnesium). Trimethoprim (TMP) was used as a non-chelating control. Addition of metal ions led to increases in MICs, whilst addition of EDTA led to decreases in MICs. The chelating effects of EDTA were reversed by addition of magnesium and most profoundly calcium. Similar effects of EDTA and calcium were observed for tetracycline (TET) and TMP. When other tetracyclines (TET, oxytetracycline (OXY) and chlortetracycline (CHL)) were used as chelators at concentrations below their MICs, TIG MICs decreased for P. aeruginosa but not for E. coli. Some decreases in TIG MICs were observed for K. pneumoniae when TET and CHL were added. A dose-dependent decrease in TIG MIC was observed for TET and was reversed by the addition of calcium. The presence of effects of EDTA and calcium on TMP MICs indicates that mechanisms outside of TIG chelation likely play a role in enhanced activity. Full characterisation of an unexpected interaction such as TIG-TET with different microorganisms could provide valuable insights into the underlying mechanisms and design of physiologically viable chelators as candidates for future combinations regimens.
Collapse
Affiliation(s)
- Amelia N Deitchman
- University of Florida, Department of Pharmaceutics, P.O. Box 100494, Gainesville, FL 32610, USA
| | - Ravi Shankar Prasad Singh
- University of Florida, Department of Pharmaceutics, P.O. Box 100494, Gainesville, FL 32610, USA; Pfizer Inc., 1 Portland Street, Cambridge, MA 02139, USA
| | - Kenneth H Rand
- University of Florida, Department of Pathology, Immunology and Laboratory Medicine, Box 100275, Gainesville, FL 32610, USA
| | - Hartmut Derendorf
- University of Florida, Department of Pharmaceutics, P.O. Box 100494, Gainesville, FL 32610, USA.
| |
Collapse
|
45
|
Dabul ANG, Avaca-Crusca JS, Van Tyne D, Gilmore MS, Camargo ILBC. Resistance in In Vitro Selected Tigecycline-Resistant Methicillin-Resistant Staphylococcus aureus Sequence Type 5 Is Driven by Mutations in mepR and mepA Genes. Microb Drug Resist 2017; 24:519-526. [PMID: 29039719 DOI: 10.1089/mdr.2017.0279] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A tigecycline-susceptible (TGC-S) Sequence Type (ST) 5 clinical methicillin-resistant Staphylococcus aureus (MRSA) strain was cultured in escalating levels of tigecycline, yielding mutants eightfold more resistant. Their genomes were sequenced to identify genetic alterations, resulting in resistance. Alterations in rpsJ, commonly related to tigecycline resistance, were also investigated. Tigecycline resistance was mediated by loss-of-function mutations in the transcriptional repressor mepR, resulting in derepression of the efflux pump mepA. Increased levels of resistance were obtained by successive mutations in mepA itself. No alterations in RpsJ were observed in selected strains, but we observed a K57M substitution, previously correlated with resistance, among TGC-S clinical strains. Thus, the pathway to tigecycline resistance in CC5 MRSA in vitro appears to be derepression of mep operon as the result of mepR loss-of-function mutation, followed by alterations in MepA efflux pump. This shows that other evolutionary pathways, besides mutation of rpsJ, are available for evolving tigecycline resistance in CC5 MRSA.
Collapse
Affiliation(s)
- Andrei Nicoli Gebieluca Dabul
- 1 Department of Physics and Interdisciplinary Science, São Carlos Institute of Physics, University of São Paulo , São Carlos, Brazil
| | - Juliana Sposto Avaca-Crusca
- 1 Department of Physics and Interdisciplinary Science, São Carlos Institute of Physics, University of São Paulo , São Carlos, Brazil
| | - Daria Van Tyne
- 2 Department of Ophthalmology, Harvard Medical School , Massachusetts Eye and Ear Infirmary, Boston, Massachusetts.,3 Department of Microbiology and Immunobiology, Harvard Medical School , Boston, Massachusetts
| | - Michael S Gilmore
- 2 Department of Ophthalmology, Harvard Medical School , Massachusetts Eye and Ear Infirmary, Boston, Massachusetts.,3 Department of Microbiology and Immunobiology, Harvard Medical School , Boston, Massachusetts
| | | |
Collapse
|
46
|
Jacqueline C, Howland K, Chesnel L. In vitro activity of ceftolozane/tazobactam in combination with other classes of antibacterial agents. J Glob Antimicrob Resist 2017; 10:326-329. [DOI: 10.1016/j.jgar.2017.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/30/2017] [Accepted: 04/06/2017] [Indexed: 11/26/2022] Open
|
47
|
Abstract
Each month, subscribers to The Formulary Monograph Service receive five to six well-documented monographs on drugs that are newly released or are in late phase 3 trials. The monographs are targeted to your Pharmacy and Therapeutics Committee. Subscribers also receive monthly one-page summary monographs on the agents that are useful for agendas and pharmacy/nursing in-services. A comprehensive target drug utilization evaluation (DUE) is also provided each month. With a subscription, the monographs are sent to you in print and CD ROM forms and are available online. Monographs can be customized to meet the needs of your facility. Subscribers to the The Formulary Monograph Service also receive access to a pharmacy bulletin board, The Formulary Information Exchange (The F.I.X.). All topics pertinent to clinical and hospital pharmacy are discussed on The F.I.X. Through the cooperation of The Formulary, Hospital Pharmacy publishes selected reviews in this column. If you would like information about The Formulary Monograph Service or The F.I.X., call The Formulary at 800-322-4349. The October 2005 monograph topics are ramelteon, lidocaine/tetracaine patch, dapsone gel, altrasentan, and deferasirox. The DUE is on ramelteon.
Collapse
Affiliation(s)
| | - Terri Levien
- Drug Information Pharmacist, Drug Information Center, Washington State University Spokane
| | - Danial E. Baker
- Drug Information Center and Pharmacy Practice, Washington State University Spokane, PO Box 1495, Spokane, WA 99210-1495
| |
Collapse
|
48
|
Abstract
Emergence of resistance among the most important bacterial pathogens is recognized as a major public health threat affecting humans worldwide. Multidrug-resistant organisms have not only emerged in the hospital environment but are now often identified in community settings, suggesting that reservoirs of antibiotic-resistant bacteria are present outside the hospital. The bacterial response to the antibiotic "attack" is the prime example of bacterial adaptation and the pinnacle of evolution. "Survival of the fittest" is a consequence of an immense genetic plasticity of bacterial pathogens that trigger specific responses that result in mutational adaptations, acquisition of genetic material, or alteration of gene expression producing resistance to virtually all antibiotics currently available in clinical practice. Therefore, understanding the biochemical and genetic basis of resistance is of paramount importance to design strategies to curtail the emergence and spread of resistance and to devise innovative therapeutic approaches against multidrug-resistant organisms. In this chapter, we will describe in detail the major mechanisms of antibiotic resistance encountered in clinical practice, providing specific examples in relevant bacterial pathogens.
Collapse
|
49
|
Lyu Y, Yang X, Goswami S, Gorityala BK, Idowu T, Domalaon R, Zhanel GG, Shan A, Schweizer F. Amphiphilic Tobramycin-Lysine Conjugates Sensitize Multidrug Resistant Gram-Negative Bacteria to Rifampicin and Minocycline. J Med Chem 2017; 60:3684-3702. [PMID: 28409644 DOI: 10.1021/acs.jmedchem.6b01742] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chromosomally encoded low membrane permeability and highly efficient efflux systems are major mechanisms by which Pseudomonas aeruginosa evades antibiotic actions. Our previous reports have shown that amphiphilic tobramycin-fluoroquinolone hybrids can enhance efficacy of fluoroquinolone antibiotics against multidrug-resistant (MDR) P. aeruginosa isolates. Herein, we report on a novel class of tobramycin-lysine conjugates containing an optimized amphiphilic tobramycin-C12 tether that sensitize Gram-negative bacteria to legacy antibiotics. Combination studies indicate the ability of these conjugates to synergize rifampicin and minocycline against MDR and extensively drug resistant (XDR) P. aeruginosa isolates and enhance efficacy of both antibiotics in the Galleria mellonella larvae in vivo infection model. Mode of action studies indicate that the amphiphilic tobramycin-lysine adjuvants enhance outer membrane cell penetration and affect the proton motive force, which energizes efflux pumps. Overall, this study provides a strategy for generating effective antibiotic adjuvants that overcome resistance of rifampicin and minocycline in MDR and XDR Gram-negative bacteria including P. aeruginosa.
Collapse
Affiliation(s)
- Yinfeng Lyu
- Institute of Animal Nutrition, Northeast Agricultural University , Harbin, Heilongjiang 150030, P.R. China.,Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - Xuan Yang
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - Sudeep Goswami
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | | | - Temilolu Idowu
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - Ronald Domalaon
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, University of Manitoba , Winnipeg, MB R3T 1R9, Canada
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University , Harbin, Heilongjiang 150030, P.R. China
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada.,Department of Medical Microbiology/Infectious Diseases, University of Manitoba , Winnipeg, MB R3T 1R9, Canada
| |
Collapse
|
50
|
Mastrolia MV, Galli L, De Martino M, Chiappini E. Use of tigecycline in pediatric clinical practice. Expert Rev Anti Infect Ther 2017; 15:605-612. [PMID: 28395551 DOI: 10.1080/14787210.2017.1318064] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Tigecycline, a derivative of minocycline, is an extended-spectrum antimicrobial agent. It has a restricted approval field in children and the experience of its adoption in clinical practice is reserved for cases of challenging infections. The aim of this review was to summarize evidence regarding the use of tigecycline in infants and children, focusing on the drug's clinical efficacy data and tolerability profile. Areas covered: We have conducted a literature search of the Cochrane Library, EMBASE, and MEDLINE databases, from their inception through 5 January 2017, using the following terms: tigecycline, newborn, infant, child, pediatrics, adolescent, human, clinical trial, and case report. Articles were excluded if they were redundant or not pertinent. Bibliographies of all relevant articles were also evaluated. Seventeen publications were included: 1 pharmacokinetic study, 16 case reports. In the selected publications, the patients' mean age was 4.45 years, 38.7% of children was <3 years old and favorable clinical response was achieved in 74.2% of cases. Expert commentary: Tigecycline may be a considerable option in life-threatening infections in pediatric patients. Its administration is well tolerated and has demonstrated a good clinical response in nonbacteremic patients. However, the available clinical records are limited and more studies are needed.
Collapse
Affiliation(s)
- Maria Vincenza Mastrolia
- a Pediatric Infectious Disease Unit , Anna Meyer Children's University Hospital , Florence , Italy
| | - Luisa Galli
- a Pediatric Infectious Disease Unit , Anna Meyer Children's University Hospital , Florence , Italy
| | - Maurizio De Martino
- a Pediatric Infectious Disease Unit , Anna Meyer Children's University Hospital , Florence , Italy
| | - Elena Chiappini
- a Pediatric Infectious Disease Unit , Anna Meyer Children's University Hospital , Florence , Italy
| |
Collapse
|