1
|
Karampatakis T, Tsergouli K, Behzadi P. Pan-Genome Plasticity and Virulence Factors: A Natural Treasure Trove for Acinetobacter baumannii. Antibiotics (Basel) 2024; 13:257. [PMID: 38534692 DOI: 10.3390/antibiotics13030257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/17/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Acinetobacter baumannii is a Gram-negative pathogen responsible for a variety of community- and hospital-acquired infections. It is recognized as a life-threatening pathogen among hospitalized individuals and, in particular, immunocompromised patients in many countries. A. baumannii, as a member of the ESKAPE group, encompasses high genomic plasticity and simultaneously is predisposed to receive and exchange the mobile genetic elements (MGEs) through horizontal genetic transfer (HGT). Indeed, A. baumannii is a treasure trove that contains a high number of virulence factors. In accordance with these unique pathogenic characteristics of A. baumannii, the authors aim to discuss the natural treasure trove of pan-genome and virulence factors pertaining to this bacterial monster and try to highlight the reasons why this bacterium is a great concern in the global public health system.
Collapse
Affiliation(s)
| | - Katerina Tsergouli
- Microbiology Department, Agios Pavlos General Hospital, 55134 Thessaloniki, Greece
| | - Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran 37541-374, Iran
| |
Collapse
|
2
|
Bostanghadiri N, Narimisa N, Mirshekar M, Dadgar-Zankbar L, Taki E, Navidifar T, Darban-Sarokhalil D. Prevalence of colistin resistance in clinical isolates of Acinetobacter baumannii: a systematic review and meta-analysis. Antimicrob Resist Infect Control 2024; 13:24. [PMID: 38419112 PMCID: PMC10902961 DOI: 10.1186/s13756-024-01376-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/04/2024] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION The development of colistin resistance in Acinetobacter baumannii during treatment has been identified in certain patients, often leading to prolonged or recurrent infections. As colistin, is the last line of therapy for A. baumannii infections that are resistant to almost all other antibiotics, colistin-resistant A. baumannii strains currently represent a significant public health threat, particularly in healthcare settings where there is significant selective pressure. AIM The aim of this study was to comprehensively determine the prevalence of colistin resistance in A. baumannii from clinical samples. Regional differences in these rates were also investigated using subgroup analyses. METHOD The comprehensive search was conducted using "Acinetobacter baumannii", "Colistin resistant" and all relevant keywords. A systematic literature search was performed after searching in PubMed, Embase, Web of Science, and Scopus databases up to April 25, 2023. Statistical analysis was performed using Stata software version 17 and sources of heterogeneity were evaluated using I2. The potential for publication bias was explored using Egger's tests. A total of 30,307 articles were retrieved. After a thorough evaluation, 734 studies were finally eligible for inclusion in the present systematic review and meta-analysis. RESULT According to the results, the prevalence of resistance to colistin among A. baumannii isolates was 4% (95% CI 3-5%), which has increased significantly from 2% before 2011 to 5% after 2012. South America had the highest resistance rate to this antibiotic. The broth microdilution method had the highest level of resistance, while the agar dilution showed the lowest level. CONCLUSIONS This meta-analysis found a low prevalence of colistin resistance among A. baumannii isolates responsible for infections worldwide from 2000 to 2023. However, there is a high prevalence of colistin-resistant isolates in certain countries. This implies an urgent public health threat, as colistin is one of the last antibiotics available for the treatment of infections caused by XDR strains of A. baumannii.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Negar Narimisa
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Mirshekar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Dadgar-Zankbar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Taki
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Tahereh Navidifar
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Ouyang Z, He W, Jiao M, Yu Q, Guo Y, Refat M, Qin Q, Zhang J, Shi Q, Zheng F, Wen Y. Mechanistic and biophysical characterization of polymyxin resistance response regulator PmrA in Acinetobacter baumannii. Front Microbiol 2024; 15:1293990. [PMID: 38476937 PMCID: PMC10927774 DOI: 10.3389/fmicb.2024.1293990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Introduction Acinetobacter baumannii PmrAB is a crucial two-component regulatory system (TCS) that plays a vital role in conferring resistance to polymyxin. PmrA, a response regulator belonging to the OmpR/PhoB family, is composed of a C-terminal DNA-binding effector domain and an N-terminal receiver domain. The receiver domain can be phosphorylated by PmrB, a transmembrane sensor histidine kinase that interacts with PmrA. Once phosphorylated, PmrA undergoes a conformational change, resulting in the formation of a symmetric dimer in the receiver domain. This conformational change facilitates the recognition of promoter DNA by the DNA-binding domain of PmrA, leading to the activation of adaptive responses. Methods X-ray crystallography was carried out to solve the structure of PmrA receiver domain. Electrophoretic mobility shift assay and Isothermal titration calorimetry were recruited to validate the interaction between the recombinant PmrA protein and target DNA. Field-emission scanning electron microscopy (FE-SEM) was employed to characterize the surface morphology of A. baumannii in both the PmrA knockout and mutation strains. Results The receiver domain of PmrA follows the canonical α5β5 response regulator assembly, which undergoes dimerization upon phosphorylation and activation. Beryllium trifluoride is utilized as an aspartate phosphorylation mimic in this process. Mutations involved in phosphorylation and dimerization significantly affected the expression of downstream pmrC and naxD genes. This impact resulted in an enhanced cell surface smoothness with fewer modifications, ultimately contributing to a decrease in colistin (polymyxin E) and polymyxin B resistance. Additionally, a conservative direct-repeat DNA PmrA binding sequence TTTAAGNNNNNTTTAAG was identified at the promoter region of the pmrC and naxD gene. These findings provide structural insights into the PmrA receiver domain and reveal the mechanism of polymyxin resistance, suggesting that PmrA could be a potential drug target to reverse polymyxin resistance in Acinetobacter baumannii.
Collapse
Affiliation(s)
- Zhenlin Ouyang
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wenbo He
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Min Jiao
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qinyue Yu
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yucheng Guo
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Moath Refat
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Qian Qin
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jiaxin Zhang
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qindong Shi
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Fang Zheng
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yurong Wen
- Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
4
|
Erdmann MB, Gardner PP, Lamont IL. The PitA protein contributes to colistin susceptibility in Pseudomonas aeruginosa. PLoS One 2023; 18:e0292818. [PMID: 37824582 PMCID: PMC10569645 DOI: 10.1371/journal.pone.0292818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes a wide range of problematic infections in individuals with predisposing conditions. Infections can be treated with colistin but some isolates are resistant to this antibiotic. To better understand the genetic basis of resistance, we experimentally evolved 19 independent resistant mutants from the susceptible laboratory strain PAO1. Whole genome sequencing identified mutations in multiple genes including phoQ and pmrB that have previously been associated with resistance, pitA that encodes a phosphate transporter, and carB and eno that encode enzymes of metabolism. Individual mutations were engineered into the genome of strain PAO1. Mutations in pitA, pmrB and phoQ increased the minimum inhibitory concentration (MIC) for colistin 8-fold, making the bacteria resistant. Engineered pitA/phoQ and pitA/pmrB double mutants had higher MICs than single mutants, demonstrating additive effects on colistin susceptibility. Single carB and eno mutations did not increase the MIC suggesting that their effect is dependent on the presence of other mutations. Many of the resistant mutants had increased susceptibility to β-lactams and lower growth rates than the parental strain demonstrating that colistin resistance can impose a fitness cost. Two hundred and fourteen P. aeruginosa isolates from a range of sources were tested and 18 (7.8%) were colistin resistant. Sequence variants in genes identified by experimental evolution were present in the 18 resistant isolates and may contribute to resistance. Overall our results identify pitA mutations as novel contributors to colistin resistance and demonstrate that resistance can reduce fitness of the bacteria.
Collapse
Affiliation(s)
| | - Paul P. Gardner
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Iain L. Lamont
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
5
|
Qu Q, Chen T, He P, Geng H, Zeng P, Luan G. Isolation and characterization of a novel lytic bacteriophage vB_Efm_LG62 infecting Enterococcus faecium. Virus Genes 2023; 59:763-774. [PMID: 37422898 DOI: 10.1007/s11262-023-02016-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/11/2023] [Indexed: 07/11/2023]
Abstract
Enterococcus faecium has been classified as a "high priority" pathogen by the World Health Organization. Enterococcus faecium has rapidly evolved as a global nosocomial pathogen with adaptation to the nosocomial environment and the accumulation of resistance to multiple antibiotics. Phage therapy is considered a promising strategy against difficult-to-treat infections and antimicrobial resistance. In this study, we isolated and characterized a novel virulent bacteriophage, vB_Efm_LG62, that specifically infects multidrug-resistant E. faecium. Morphological observations suggested that the phage has siphovirus morphology, with an optimal multiplicity of infection of 0.001. One-step growth tests revealed that its latent growth was at 20 min, with a burst size of 101 PFU/cell. Phage vB_Efm_LG62 was verified to have a double-stranded genome of 42,236 bp (35.21% GC content), containing 66 predicted coding sequences as determined by whole genomic sequencing. No genes were predicted to have functions associated with virulence factors or antibiotic resistance, indicating that the phage vB_Efm_LG62 has good therapeutic potential. Our isolation and characterization of this highly efficient phage aids in expanding our knowledge of E. faecium-targeting phages, and provides additional options for phage cocktail therapy.
Collapse
Affiliation(s)
- Qianyu Qu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tao Chen
- Medical Laboratory, Xindu District People's Hospital of Chengdu, Chengdu, China
| | - Penggang He
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huaixin Geng
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China
| | - Peibin Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Guangxin Luan
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China.
| |
Collapse
|
6
|
Deilamani MO, Nikkhahi F, Bakht M, Ali Alizadeh S, Fardsanei F, Javadi A, Marashi SMA, Aslanimehr M, Peymani A. Evaluation of ethanol and EDTA concentrations in the expression of biofilm-producing smf-1, rpfF genes in XDR clinical isolates of Stenotrophomonas maltophilia. BMC Microbiol 2023; 23:277. [PMID: 37775770 PMCID: PMC10542227 DOI: 10.1186/s12866-023-03008-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Stenotrophomonas maltophilia is able to cause infections in immunocompromised patients, and the treatment of this opportunistic pathogen is complicated due to its virulence factors, antibiotic resistance, and the ability of the bacteria to produce biofilm. The main goals of this study were to assess the susceptibility of extensively drug-resistant (XDR) isolates to ethanol and EDTA, and evaluating the synergistic effect of these disinfectants, and also survey the effect of exposure to sub-inhibitory concentrations of ethanol and EDTA on the expression of biofilm-producing smf-1, rpfF genes. RESULTS The results showed that EDTA significantly increased the effectiveness of the ethanol and have a synergistic effect. All of the 10 XDR isolates included in the current study harbored smf-1 and rpfF genes and produced biofilm. After exposure to MIC, sub-MIC, synergism, and sub-synergism of ethanol and EDTA, the expression of smf-1 and rpfF genes was repressed significantly. CONCLUSION In the current study, it was indicated that the expression of biofilm-producing genes was repressed when bacteria are exposed to different concentrations of ethanol and EDTA. Future studies should include more complex microbial communities residing in the hospitals, and more disinfectants use in hospitals. Expression of other virulence genes in different conditions is suggested.
Collapse
Affiliation(s)
- Mohadeseh Ostovari Deilamani
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
- Student research committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farhad Nikkhahi
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
| | - Mehdi Bakht
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
- Student research committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Safar Ali Alizadeh
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
| | - Fatemeh Fardsanei
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran.
| | - Amir Javadi
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
| | - Seyed Mahmoud Amin Marashi
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
| | - Masoumeh Aslanimehr
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
| | - Amir Peymani
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, PO Box: 34199-15315, Qazvin, Iran
| |
Collapse
|
7
|
Gadar K, de Dios R, Kadeřábková N, Prescott TAK, Mavridou DAI, McCarthy RR. Disrupting iron homeostasis can potentiate colistin activity and overcome colistin resistance mechanisms in Gram-Negative Bacteria. Commun Biol 2023; 6:937. [PMID: 37704838 PMCID: PMC10499790 DOI: 10.1038/s42003-023-05302-2] [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: 01/26/2023] [Accepted: 08/29/2023] [Indexed: 09/15/2023] Open
Abstract
Acinetobacter baumannii is a Gram-negative priority pathogen that can readily overcome antibiotic treatment through a range of intrinsic and acquired resistance mechanisms. Treatment of carbapenem-resistant A. baumannii largely relies on the use of colistin in cases where other treatment options have been exhausted. However, the emergence of resistance against this last-line drug has significantly increased amongst clinical strains. In this study, we identify the phytochemical kaempferol as a potentiator of colistin activity. When administered singularly, kaempferol has no effect on growth but does impact biofilm formation. Nonetheless, co-administration of kaempferol with sub-inhibitory concentrations of colistin exposes bacteria to a metabolic Achilles heel, whereby kaempferol-induced dysregulation of iron homeostasis leads to bacterial killing. We demonstrate that this effect is due to the disruption of Fenton's reaction, and therefore to a lethal build-up of toxic reactive oxygen species in the cell. Furthermore, we show that this vulnerability can be exploited to overcome both intrinsic and acquired colistin resistance in clinical strains of A. baumannii and E. coli in vitro and in the Galleria mellonella model of infection. Overall, our findings provide a proof-of-principle demonstration that targeting iron homeostasis is a promising strategy for enhancing the efficacy of colistin and overcoming colistin-resistant infections.
Collapse
Affiliation(s)
- Kavita Gadar
- Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Rubén de Dios
- Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Nikol Kadeřábková
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Despoina A I Mavridou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
- John Ring LaMontagne Centre for Infectious Diseases, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronan R McCarthy
- Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK.
| |
Collapse
|
8
|
Khoshbakht R, Panahi S, Neshani A, Ghavidel M, Ghazvini K. Novel approaches to overcome Colistin resistance in Acinetobacter baumannii: Exploring quorum quenching as a potential solution. Microb Pathog 2023; 182:106264. [PMID: 37474078 DOI: 10.1016/j.micpath.2023.106264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023]
Abstract
Acinetobacter baumannii is responsible for a variety of infections, such as nosocomial infections. In recent years, this pathogen has gained resistance to many antibiotics, and thus, carbapenems were used to treat infections with MDR A. baumannii strains in clinical settings. However, as carbapenem-resistant isolates are becoming increasingly prevalent, Colistin is now used as the last line of defense against resistant A. baumannii strains. Unfortunately, reports are increasing on the presence of Colistin-resistant phenotypes in infections caused by A. baumannii, creating an urgent need to find a substitute way to combat these resistant isolates. Quorum sensing inhibition, also known as quorum quenching, is an efficient alternative way of reversing resistance in different Gram-negative bacteria. Quorum sensing is a mechanism used by bacteria to communicate with each other by secreting signal molecules. When the population of bacteria increases and the concentration of signal molecules reaches a certain threshold, bacteria can implement mechanisms to adapt to a hostile environment, such as biofilm formation. Biofilms have many advantages for pathogens, such as antibiotic resistance. Different studies have revealed that disrupting the biofilm of A. baumannii makes it more susceptible to antibiotics. Although very few studies have been conducted on the biofilm disruption through quorum quenching in Colistin-resistant A. baumannii, these studies and similar studies bring hope in finding an alternative way of treating the Colistin-resistant isolates. In conclusion, quorum quenching has the potential to be used against Colistin-resistant A. baumannii.
Collapse
Affiliation(s)
- Reza Khoshbakht
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Susan Panahi
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Neshani
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdis Ghavidel
- Shahid Hasheminejad Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kiarash Ghazvini
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Jiang H, Jiao X, Yu T, Wang W, Cheng H, Huang G, Fang J. Contribution of different class 2 integron elements to fitness costs in multi-drug resistant Escherichia coli and evaluation of their adaptability in “farm-to-table” environments. Food Microbiol 2023; 113:104279. [PMID: 37098435 DOI: 10.1016/j.fm.2023.104279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 03/13/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Integrons play a pivotal role in the dissemination of antimicrobial resistance, because they can capture and express exogenous antimicrobial resistance genes. This study aimed to elucidate the structure and contribution of different elements of class 2 integrons to fitness costs in their host bacteria and evaluate their adaptability to the "farm-to-table" process. We mapped 27 typical class 2 integrons of Escherichia coli isolated from aquatic foods and pork products, each harboring an inactive truncated class 2 integrase gene and the gene cassette (GC) array dfrA1-sat2-aadA1 with strong Pc2A/Pc2B promoters. Notably, the fitness costs associated with class 2 integrons depended on the Pc promoter strength and quantity and content of GCs in the array. Additionally, the costs of integrases were activity-dependent, and a balance was identified between GC capture ability and integron stability, which could explain the inactive truncated integrase identified. Although typical class 2 integrons exhibited low-cost structures in E. coli, the bacteria incurred biological costs, including decreasing growth rates and biofilm formation, in farm-to-table environments, especially under low-nutrient conditions. Nevertheless, sub-inhibitory antibiotic concentrations led to the selection of class 2 integron-carrying bacteria. This study provides important insights into how integrons may travel from preharvest to consumer goods.
Collapse
|
10
|
Zeng L, Lin F, Ling B. Effect of traditional Chinese medicine monomers interfering with quorum-sensing on virulence factors of extensively drug-resistant Acinetobacter baumannii. Front Pharmacol 2023; 14:1135180. [PMID: 37063277 PMCID: PMC10097947 DOI: 10.3389/fphar.2023.1135180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
The antimicrobial resistance of Acinetobacter baumannii (A. baumannii) clinical isolates has emerged as a great threat to public health. Quorum sensing (QS) is one of the resistance mechanisms for drug-resistant A. baumannii. Interfering with QS is a promising strategy to combat infections caused by drug-resistant bacteria. This study explored the QS inhibition ability of thirty-four traditional Chinese medicine monomers (TCMMs) and assessed the effect of QS inhibitors (QSIs) on the virulence factors of twelve extensively drug-resistant A. baumannii (XDRAB) strains. Nine traditional Chinese medicine monomers, such as caffeic acid, cinnamic acid, and myricetin, were found to be able to inhibit the bacterial QS. Then, at 1/8 of the minimal inhibitory concentration, we found that these QSIs inhibited extensively drug-resistant A. baumannii adhesion and biofilm formation and downregulated the expression levels of virulence-associated genes (abaI, abaR, csuE, pgaA, and bap). In conclusion, nine traditional Chinese medicine monomers have QS inhibitory activity and may downregulate QS genes to interfere with the QS system, which could inhibit the expression of extensively drug-resistant A. baumannii virulence factors. These results suggest that traditional Chinese medicine monomers could develop as novel anti-virulence compounds to control extensively drug-resistant A. baumannii infections.
Collapse
Affiliation(s)
- Li Zeng
- School of Pharmacy, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The Third People’s Hospital of Yibin, Yibin, China
| | - Fei Lin
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
| | - Baodong Ling
- School of Pharmacy, Chengdu Medical College, Chengdu, China
- *Correspondence: Baodong Ling,
| |
Collapse
|
11
|
Shamkani F, Barzi SM, Badmasti F, Chiani M, Mirabzadeh E, Zafari M, Shafiei M. Enhanced anti-biofilm activity of the minocycline-and-gallium-nitrate using niosome wrapping against Acinetobacter baumannii in C57/BL6 mouse pneumonia model. Int Immunopharmacol 2023; 115:109551. [PMID: 36621329 DOI: 10.1016/j.intimp.2022.109551] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/24/2022] [Accepted: 12/02/2022] [Indexed: 01/09/2023]
Abstract
Acinetobacter baumannii is a worldwide health issue in terms of its high antibiotic resistance and ability to form biofilms. Nanoparticles (NPs) with high biocompatibility, high penetrating ability, and low medication dose can successfully treat the antibiotic-resistant infections. In this research, the anti-biofilm activity of niosomes containing minocycline and gallium nitrate (GaN) against A. baumannii biofilm was determined. In order to improve their anti-biofilm properties, minocycline and GaN were encapsulated in niosomes as biocompatible drug carriers. The niosomes' size, zeta potential, shape, stability, drug entrapment efficacy, drug release pattern and antibacterial activity were assessed. Several clinical samples were isolated from the lungs of patients hospitalized at Loghman hospital, Tehran, Iran. The biofilm formation of most lethal clinical isolates of A. baumannii was analyzed. The pneumonia model was generated by intranasally administering A. baumannii suspension to anesthetized mice whose immune systems was compromised twice by cyclophosphamide. Lung infection of the mouse with A. baumannii was confirmed using PCR. After treatment, the lungs were excised under sterile conditions and stained with hematoxylin and eosin (H&E) to determine histological symptoms, inflammation and intercellular secretions. The niosomes contained minocycline and GaN had an average size of 230 nm and a zeta potential of -40 mV, respectively. The percentage of drug entrapment and delayed drug release was both high in niosomal formulations. Niosomes containing minocycline and GaN dispersed 1, 3 and 5 day old biofilms. The mice given the combination of two compounds required less time to be treated than the animals given the single medication (minocycline). The minocycline& GaN-loaded niosomes could be considered as promising candidates to treat the infections caused by A. baumannii biofilm.
Collapse
Affiliation(s)
- Farnaz Shamkani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Mohsen Chiani
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Esmat Mirabzadeh
- Department of Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Zafari
- Department of Biology, The University of Akron, Akron, OH 44325, United States
| | - Morvarid Shafiei
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
12
|
Identification of novel genes involved in the biofilm formation process of Avian Pathogenic Escherichia coli (APEC). PLoS One 2022; 17:e0279206. [PMID: 36534660 PMCID: PMC9762606 DOI: 10.1371/journal.pone.0279206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Avian pathogenic Escherichia coli (APEC) is the etiological agent of avian colibacillosis, a leading cause of economic loss to the poultry industry worldwide. APEC causes disease using a diverse repertoire of virulence factors and has the ability to form biofilms, which contributes to the survival and persistence of APEC in harsh environments. The objective of this study was to identify genes most widespread and important in APEC that contribute to APEC biofilm formation. Using the characterized APEC O18 as the template strain, a total of 15,660 mutants were randomly generated using signature tagged mutagenesis and evaluated for decreased biofilm formation ability using the crystal violet assay. Biofilm deficient mutants were sequenced, and a total of 547 putative biofilm formation genes were identified. Thirty of these genes were analyzed by PCR for prevalence among 109 APEC isolates and 104 avian fecal E. coli (AFEC) isolates, resulting in nine genes with significantly greater prevalence in APEC than AFEC. The expression of these genes was evaluated in the wild-type APEC O18 strain using quantitative real-time PCR (qPCR) in both the exponential growth phase and the mature biofilm phase. To investigate the role of these genes in biofilm formation, isogenic mutants were constructed and evaluated for their biofilm production and planktonic growth abilities. Four of the mutants (rfaY, rfaI, and two uncharacterized genes) displayed significantly decreased biofilm formation, and of those four, one (rfaI) displayed significantly decreased growth compared to the wild type. Overall, this study identified novel genes that may be important in APEC and its biofilm formation. The data generated from this study will benefit further investigation into the mechanisms of APEC biofilm formation.
Collapse
|
13
|
Specific egg yolk antibody raised to biofilm associated protein (Bap) is protective against murine pneumonia caused by Acinetobacter baumannii. Sci Rep 2022; 12:12576. [PMID: 35869264 PMCID: PMC9307575 DOI: 10.1038/s41598-022-16894-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
Acinetobacter baumannii easily turns into pan drug-resistant (PDR) with a high mortality rate. No effective commercial antibiotic or approved vaccine is available against drug-resistant strains of this pathogen. Egg yolk immunoglobulin (IgY) could be used as a simple and low-cost biotherapeutic against its infections. This study evaluates the prophylactic potential of IgY against A. baumannii in a murine pneumonia model. White Leghorn hens were immunized with intramuscular injection of the recombinant biofilm-associated protein (Bap) from A. baumannii on days 0, 21, 42, and 63. The reactivity and antibiofilm activity of specific IgYs raised against the Bap was evaluated by indirect ELISA and a microtiter plate assay for biofilm formation. The IgYs against Bap were able to decrease the biofilm formation ability of A. baumannii and protect the mice against the challenge of A. baumannii. IgYs antibody raised here shows a good antigen-specificity and protectivity which can be used in passive immunotherapy against A. baumannii. In conclusion, the IgY against biofilm-associated protein proves prophylactic in a murine pneumonia model.
Collapse
|
14
|
Kamoshida G, Yamada N, Nakamura T, Yamaguchi D, Kai D, Yamashita M, Hayashi C, Kanda N, Sakaguchi M, Morimoto H, Sawada T, Okada T, Kaya Y, Takemoto N, Yahiro K. Preferential Selection of Low-Frequency, Lipopolysaccharide-Modified, Colistin-Resistant Mutants with a Combination of Antimicrobials in Acinetobacter baumannii. Microbiol Spectr 2022; 10:e0192822. [PMID: 36173297 PMCID: PMC9602988 DOI: 10.1128/spectrum.01928-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/13/2022] [Indexed: 12/31/2022] Open
Abstract
Colistin, which targets lipopolysaccharide (LPS), is used as a last-resort drug against severe infections caused by drug-resistant Acinetobacter baumannii. However, A. baumannii possesses two colistin-resistance mechanisms. LPS modification caused by mutations in pmrAB genes is often observed in clinical isolates of multidrug-resistant Gram-negative pathogens. In addition to LPS modification, A. baumannii has a unique colistin resistance mechanism, a complete loss of LPS due to mutations in the lpxACD genes, which are involved in LPS biosynthesis. This study aimed to elucidate the detailed mechanism of the emergence of colistin-resistant A. baumannii using strains with the same genetic background. Various colistin-resistant strains were generated experimentally using colistin alone and in combination with other antimicrobials, such as meropenem and ciprofloxacin, and the mutation spectrum was analyzed. In vitro selection of A. baumannii in the presence of colistin led to the emergence of strains harboring mutations in lpxACD genes, resulting in LPS-deficient colistin-resistant strains. However, combination of colistin with other antimicrobials led to the selection of pmrAB mutant strains, resulting in strains with modified LPS (LPS-modified strains). Further, the LPS-deficient strains showed decreased fitness and increased susceptibility to many antibiotics and disinfectants. As LPS-deficient strains have a higher biological cost than LPS-modified strains, our findings suggested that pmrAB mutants are more likely to be isolated in clinical settings. We provide novel insights into the mechanisms of resistance to colistin and provide substantial solutions along with precautions for facilitating current research and treatment of colistin-resistant A. baumannii infections. IMPORTANCE Acinetobacter baumannii has developed resistance to various antimicrobial drugs, and its drug-resistant strains cause nosocomial infections. Controlling these infections has become a global clinical challenge. Carbapenem antibiotics are the frontline treatment drugs for infectious diseases caused by A. baumannii. For patients with infections caused by carbapenem-resistant A. baumannii, colistin-based therapy is often the only treatment option. However, A. baumannii readily acquires resistance to colistin. Many patients infected with colistin-resistant A. baumannii undergo colistin treatment before isolation of the colistin-resistant strain, and it is hypothesized that colistin resistance predominantly emerges under selective pressure during colistin therapy. Although the concomitant use of colistin and carbapenems has been reported to have a synergistic effect in vitro against carbapenem-resistant A. baumannii strains, our observations strongly suggest the need for attention to the emergence of strains with a modified lipopolysaccharide during treatment.
Collapse
Affiliation(s)
- Go Kamoshida
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Noriteru Yamada
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tomoka Nakamura
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Daiki Yamaguchi
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Daichi Kai
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Maho Yamashita
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Chiaki Hayashi
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Nana Kanda
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Moe Sakaguchi
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hitoshi Morimoto
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Teppei Sawada
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tomoko Okada
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yuki Kaya
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Norihiko Takemoto
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kinnosuke Yahiro
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
15
|
Choudhary M, Shrivastava R, Vashistt J. Acinetobacter baumannii Biofilm Formation: Association with Antimicrobial Resistance and Prolonged Survival under Desiccation. Curr Microbiol 2022; 79:361. [PMID: 36253556 DOI: 10.1007/s00284-022-03071-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/29/2022] [Indexed: 11/30/2022]
Abstract
Biofilm-forming multidrug-resistant Acinetobacter baumannii has emerged as a global pathogen. This study investigated the impact of biofilm formation by A. baumannii on antimicrobial resistance and prolonged survival under desiccation, which is essential for effective infection control of A. baumannii in hospital settings. Seventy-eight clinical isolates of A. baumannii were identified, and antibiotic susceptibility profiles were assessed. All the isolates were investigated for their biofilm-forming abilities at 24 and 48 h. The biofilm inhibitory concentrations of antibiotics were evaluated for selected biofilm-forming isolates to determine the influence of biofilm on antibiotic tolerance. The impact of biofilm formation on desiccation tolerance was also evaluated for up to 48 days. The results revealed that out of 78 A. baumannii clinical isolates, 83% were MDR and 17% non-MDR. Overall, 79% of isolates formed high biofilm after 24 h. The extent of biofilm formation gets significantly increased after 48 h, and 87% of isolates formed high biofilm. It was observed that eradicating mature biofilm requires up to a thousandfold higher concentration of antibiotics than MICs, and biofilm-forming isolates can survive for a prolonged period under desiccation. In conclusion, our findings revealed that both MDR and non-MDR isolates of A. baumannii could form biofilms on abiotic surfaces. A. baumannii biofilms contribute to endurance in the presence of antimicrobials and desiccation conditions, which are significant trouble for hospital patient care management. The present findings may offer insights for developing preventive measures to tackle biofilm-associated A. baumannii infection.
Collapse
Affiliation(s)
- Monika Choudhary
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Rahul Shrivastava
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Jitendraa Vashistt
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India.
| |
Collapse
|
16
|
Phenotypic, genomic, and transcriptomic changes in an Acinetobacter baumannii strain after spaceflight in China's Tiangong-2 space laboratory. Braz J Microbiol 2022; 53:1447-1464. [PMID: 35763257 DOI: 10.1007/s42770-022-00772-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 05/11/2022] [Indexed: 11/02/2022] Open
Abstract
Acinetobacter baumannii is an opportunistic pathogen often found in patients with low immunity. It causes nosocomial infections, which are difficult to treat. This bacterium can rapidly mutate, developing resistance to antimicrobials and adapting to environmental stress, thereby increasing its survival. Understanding such adaptive mechanisms will be beneficial for controlling the spread of A. baumannii. Astrobiology studies have demonstrated that microbiomes from astronauts and manned spaceflight environments show resistance to stress and antibiotics. Astronauts also encounter low immunity during spaceflight missions. The extreme conditions of spaceflight provide a unique research platform for studying how opportunistic pathogens such as A. baumannii adapt to conditions such as microgravity and mutate during spaceflight. In this study, we compared phenotypic variations and analyzed genomic and transcriptomic variations in A. baumannii strains exposed to three different conditions: ST1 (64 days on Tiangong-2 space laboratory), GT1 (ground control), and Aba (original strain). Biofilm formation ability of the ST1 strain increased after 64 days of spaceflight. In addition, high-throughput sequencing revealed that some differentially expressed genes were upregulated in the ST1 strain compared to the GT1 strain. These results provide insights into the environmental adaptation of this widespread pathogen.
Collapse
|
17
|
Hashemizadeh Z, Hatam G, Fathi J, Aminazadeh F, Hosseini-Nave H, Hadadi M, Shakib NH, Kholdi S, Bazargani A. The Spread of Insertion Sequences Element and Transposons in Carbapenem Resistant Acinetobacter baumannii in a Hospital Setting in Southwestern Iran. Infect Chemother 2022; 54:275-286. [PMID: 35706082 PMCID: PMC9259918 DOI: 10.3947/ic.2022.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/08/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Acinetobacter baumannii is one of the most important hospital pathogenic bacteria that cause infectious diseases. The present study aimed to determine the frequency of carbapenem resistance genes in association with transposable elements and molecular typing of carbapenem-resistant A. baumannii bacteria collected from patients in Shiraz, Iran. MATERIALS AND METHODS A total of 170 carbapenem-resistant A. baumannii isolates were obtained from different clinical specimens in two hospitals. The minimum inhibitory concentrations (MIC) of imipenem were determined and the prevalence of OXA Carbapenemases, Metallo-β-lactamases genes, insertion sequences (IS) elements, and transposons were evaluated by the polymerase chain reaction (PCR) method. Finally, molecular typing of the isolates was performed by the Enterobacterial Repetitive Intergenic Consensus-PCR method. RESULTS The MICs ranged from 16 to 1,024 µg/mL for imipenem-resistant A. baumannii isolates. Out of the 170 carbapenem resistant A. baumannii isolates, blaOXA-24-like (94, 55.3%) followed by blaOXA-23-like (71, 41.7%) were predominant. In addition, A. baumannii isolates carried blaVIM (71, 41.7%), blaGES (32, 18.8%), blaSPM (4, 2.3%), and blaKPC (1, 0.6%). Moreover, ISAba1 (94.2%) and Tn2009 (39.2%) were the most frequent transposable elements. Furthermore, (71, 44.0%) and (161, 94.7%) of the ISAba1 of the isolates were associated with blaOXA-23 and blaOXA-51 genes, respectively. Besides (3, 1.7%), (1, 0.6%) and (5, 2.9%) of blaOXA-23 were associated with IS18, ISAba4, and ISAba2, respectively. Considering an 80.0% cut off, clusters and four singletons were detected. CONCLUSION According to the results, transposable elements played an important role in the development of resistance genes and resistance to carbapenems. The results also indicated carbapenem-resistant A. baumannii bacteria as a public health concern.
Collapse
Affiliation(s)
- Zahra Hashemizadeh
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Hatam
- Basic Sciences in Infectious Diseases Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Fathi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Aminazadeh
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Hosseini-Nave
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahtab Hadadi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nafiseh Hosseinzadeh Shakib
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sodeh Kholdi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdollah Bazargani
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
18
|
Evaluating the Effectiveness of Hospital Antiseptics on Multidrug-Resistant Acinetobacter baumannii: Understanding the Relationship between Microbicide and Antibiotic Resistance. Antibiotics (Basel) 2022; 11:antibiotics11050614. [PMID: 35625258 PMCID: PMC9137960 DOI: 10.3390/antibiotics11050614] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 12/22/2022] Open
Abstract
Acinetobacter baumannii hospital infections are difficult to treat due to the rapid emergence of multidrug-resistant (MDR) strains. In addition, A. baumannii can survive in numerous adverse environments, including in the presence of common hospital antiseptics. We hypothesized that in addition to accumulating drug resistance determinants, MDR A. baumannii strains also accumulate mutations that allow for greater microbicide tolerance when compared to pan-susceptible (PS) strains. To test this hypothesis, we compared the survival of five MDR and five PS patient isolates when exposed to bleach, ethanol, quaternary ammonium compounds, chlorhexidine gluconate, and povidone. We evaluated bacteria in a free-living planktonic state and under biofilm conditions. Each disinfectant eliminated 99.9% of planktonic bacteria, but this was not the case for bacterial biofilms. Next, we characterized strains for the presence of the known microbicide-resistance genes cepA, qacEΔ1, qacE, and qacA. MDR strains did not survive more than PS strains in the presence of microbicides, but microbicide-resistant strains had higher survival rates under some conditions. Interestingly, the PS strains were more likely to possess microbicide-resistance genes. Microbicide resistance remains an important topic in healthcare and may be independent of antimicrobial resistance. Hospitals should consider stricter isolation precautions that take pan-susceptible strains into account.
Collapse
|
19
|
Pourhajibagher M, Bahador A. Physico-mechanical properties, antimicrobial activities, and anti-biofilm potencies of orthodontic adhesive containing cerium oxide nanoparticles against Streptococcus mutans. Folia Med (Plovdiv) 2022; 64:252-259. [DOI: 10.3897/folmed.64.e60418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/08/2021] [Indexed: 11/12/2022] Open
Abstract
Introduction: White spot lesions around orthodontic brackets may lead to the formation of dental caries during and following fixed orthodontic treatment.
Aim: This study aimed to evaluate the physico-mechanical properties and antimicrobial potencies of orthodontic adhesive doped with cerium oxide nanoparticles (CeO2-NPs) against Streptococcus mutans.
Materials and methods: After synthesis and conformation of CeO2-NPs by transmission electron microscope (TEM), shear bond strength (SBS) and adhesive remnant index (ARI) of modified orthodontic adhesive containing different concentrations of CeO2-NPs (0, 1, 2, 5, and 10 wt%) were measured. The antimicrobial effects of modified orthodontic adhesive were evaluated by disk agar diffusion method and biofilm formation inhibition assay.
Results: The pseudo-spherical shapes of CeO2-NPs were observed in TEM micrographs. The physico-mechanical finding showed that 5 wt% CeO2-NPs showed the highest concentration of CeO2-NPs and SBS value (18.21±9.06 MPa, p<0.05) simultaneously with no significant differences in ARI compared with the control group (p>0.05). There was a significant reduction in cell viability of S. mutans with increasing CeO2-NPs concentration. The 3.1 Log10 and 4.6 Log10 reductions were observed in the count of treated S. mutans with 5 and 10 wt% CeO2-NPs, respectively (p<0.05).
Conclusions: Overall, an orthodontic adhesive containing 5 wt% CeO2-NPs had antimicrobial properties against S. mutans without adverse effects on SBS and ARI.
Collapse
|
20
|
Liu X, Wu Y, Zhu Y, Jia P, Li X, Jia X, Yu W, Cui Y, Yang R, Xia W, Xu Y, Yang Q. Emergence of colistin-resistant hypervirulent Klebsiella pneumoniae (CoR-HvKp) in China. Emerg Microbes Infect 2022; 11:648-661. [PMID: 35086435 PMCID: PMC8896207 DOI: 10.1080/22221751.2022.2036078] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Colistin is regarded as a last-resort agent to combat infections caused by multidrug-resistant (MDR) Gram-negative bacteria, especially carbapenem-resistant isolates. In recent years, reports of colistin-resistant Klebsiella pneumoniae (CoRKp) are increasing. However, the molecular mechanism and relevance of colistin resistance and virulence remain unclear. Fourteen CoRKp strains were retrospectively screened from 1884 clinical K. pneumoniae isolates during 2017–2018 in China. Six CoRKp strains belonging to ST11 were MDR strains. Plasmid-mediated mobile colistin-resistance genes had a low prevalence in CoRKp. Our results revealed that up-regulated expression of two-component systems, especially phoPQ, contributed more to colistin resistance. mgrB mutation was the most common molecular mechanism of colistin resistance, caused by either nonsense mutations or insertion sequences, which drove the overexpression of phoPQ system. This study also identified three novel point mutations in pmrAB system, in which D313N mutation in pmrB was proved to increase the MIC to colistin by 16-fold. In addition, 6 out of 14 CoRKP strains independently carried hypervirulence genes. All six strains showed medium-to-high virulence phenotype compared with NTUH-K2044 strain in mice intraperitoneal challenge models. We found that 4 strains were biofilm strong producers and transcriptome analysis revealed that three of them significantly up-regulated expression of type III fimbrial shaft gene mrkA. In conclusion, our result revealed the emergence of colistin-resistant and hypervirulent MDR K. pneumoniae, which is a noticeable superbug and could cause a severe challenge to public health.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Medical Technology Academy, Beihua University, Jilin, Jilin Province, China; Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China; Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yarong Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Ying Zhu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Peiyao Jia
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xue Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xinmiao Jia
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Central Research Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Wei Yu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Wei Xia
- Medical Technology Academy, Beihua University, Jilin, Jilin Province, China; Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China; Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingchun Xu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiwen Yang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Farshadzadeh Z, Pourhajibagher M, Taheri B, Ekrami A, Modarressi MH, Azimzadeh M, Bahador A. Antimicrobial and anti-biofilm potencies of dermcidin-derived peptide DCD-1L against Acinetobacter baumannii: an in vivo wound healing model. BMC Microbiol 2022; 22:25. [PMID: 35026999 PMCID: PMC8756727 DOI: 10.1186/s12866-022-02439-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The global emergence of Acinetobacter baumannii resistance to most conventional antibiotics presents a major therapeutic challenge and necessitates the discovery of new antibacterial agents. The purpose of this study was to investigate in vitro and in vivo anti-biofilm potency of dermcidin-1L (DCD-1L) against extensively drug-resistant (XDR)-, pandrug-resistant (PDR)-, and ATCC19606-A. baumannii. METHODS After determination of minimum inhibitory concentration (MIC) of DCD-1L, in vitro anti-adhesive and anti-biofilm activities of DCD-1L were evaluated. Cytotoxicity, hemolytic activity, and the effect of DCD-1L treatment on the expression of various biofilm-associated genes were determined. The inhibitory effect of DCD-1L on biofilm formation in the model of catheter-associated infection, as well as, histopathological examination of the burn wound sites of mice treated with DCD-1L were assessed. RESULTS The bacterial adhesion and biofilm formation in all A. baumannii isolates were inhibited at 2 × , 4 × , and 8 × MIC of DCD-1L, while only 8 × MIC of DCD-1L was able to destroy the pre-formed biofilm in vitro. Also, reduce the expression of genes involved in biofilm formation was observed following DCD-1L treatment. DCD-1L without cytotoxic and hemolytic activities significantly reduced the biofilm formation in the model of catheter-associated infection. In vivo results showed that the count of A. baumannii in infected wounds was significantly decreased and the promotion in wound healing by the acceleration of skin re-epithelialization in mice was observed following treatment with 8 × MIC of DCD-1L. CONCLUSIONS Results of this study demonstrated that DCD-1L can inhibit bacterial attachment and biofilm formation and prevent the onset of infection. Taking these properties together, DCD-1L appears as a promising candidate for antimicrobial and anti-biofilm drug development.
Collapse
Affiliation(s)
- Zahra Farshadzadeh
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Behrouz Taheri
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Ekrami
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Masoud Azimzadeh
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Bahador
- Fellowship in Clinical Laboratory Sciences, BioHealth Lab, Tehran, Iran.
| |
Collapse
|
22
|
AL-Dulaimi M, Algburi A, Abdelhameed A, Mazanko MS, Rudoy DV, Ermakov AM, Chikindas ML. Antimicrobial and Anti-Biofilm Activity of Polymyxin E Alone and in Combination with Probiotic Strains of Bacillus subtilis KATMIRA1933 and Bacillus amyloliquefaciens B-1895 against Clinical Isolates of Selected Acinetobacter spp.: A Preliminary Study. Pathogens 2021; 10:1574. [PMID: 34959528 PMCID: PMC8707300 DOI: 10.3390/pathogens10121574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 01/23/2023] Open
Abstract
Acinetobacter spp., the nosocomial pathogen, forms strong biofilms and is resistant to numerous antibiotics, causing persistent infections. This study investigates the antibacterial and anti-biofilm activity of polymyxin E alone and in combination with the cell-free supernatants (CFS) of the tested probiotic bacilli, Bacillus subtilis KATMIRA1933 and Bacillus amyloliquefaciens B-1895 against the selected Acinetobacter spp. starins. Three isolates of Acinetobacter spp., designated as Acinetobacter spp. isolate 1; Acinetobacter spp. isolate 2, and Acinetobacter spp. isolate 3, were collected from patients with burns, wounds, and blood infections, respectively. Bacterial identification and antibiotic susceptibility testing were conducted using the VITEK2 system. Auto-aggregation and coaggregation of the tested bacilli strains with the selected Acinetobacter spp. isolates were evaluated. A disk diffusion assay was used to identify the microorganism's susceptibility to the selected antibiotics, alone and in combination with the CFS of the bacilli. The MIC and MBIC (minimum inhibitory and minimum biofilm inhibitory concentrations) of polymyxin E combined with bacilli CFS were determined. Acinetobacter spp. isolates were (i) sensitive to polymyxin E, (ii) able to form a strong biofilm, and (iii) resistant to the tested antibiotics and the CFS of tested bacilli. Significant inhibition of biofilm formation was noticed when CFS of the tested bacilli were combined with polymyxin E. The bacilli CFS showed synergy with polymyxin E against planktonic cells and biofilms of the isolated pathogens.
Collapse
Affiliation(s)
- Munaf AL-Dulaimi
- Educational Laboratories, Baqubah General Hospital, Baqubah 32001, Iraq;
| | - Ammar Algburi
- Scholarship and Cultural Relations Department, University of Diyala, Baqubah 32001, Iraq;
- Biotechnology Department, College of Science, University of Diyala, Baqubah 32001, Iraq
| | - Alyaa Abdelhameed
- Scholarship and Cultural Relations Department, University of Diyala, Baqubah 32001, Iraq;
| | - Maria S. Mazanko
- Center for Agrobiotechnology, Don State Technical University, 344002 Rostov-on-Don, Russia; (M.S.M.); (D.V.R.); (A.M.E.); (M.L.C.)
| | - Dmitry V. Rudoy
- Center for Agrobiotechnology, Don State Technical University, 344002 Rostov-on-Don, Russia; (M.S.M.); (D.V.R.); (A.M.E.); (M.L.C.)
| | - Alexey M. Ermakov
- Center for Agrobiotechnology, Don State Technical University, 344002 Rostov-on-Don, Russia; (M.S.M.); (D.V.R.); (A.M.E.); (M.L.C.)
| | - Michael L. Chikindas
- Center for Agrobiotechnology, Don State Technical University, 344002 Rostov-on-Don, Russia; (M.S.M.); (D.V.R.); (A.M.E.); (M.L.C.)
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ 08904, USA
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| |
Collapse
|
23
|
Pompilio A, Scribano D, Sarshar M, Di Bonaventura G, Palamara AT, Ambrosi C. Gram-Negative Bacteria Holding Together in a Biofilm: The Acinetobacter baumannii Way. Microorganisms 2021; 9:1353. [PMID: 34206680 PMCID: PMC8304980 DOI: 10.3390/microorganisms9071353] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022] Open
Abstract
Bacterial biofilms are a serious public-health problem worldwide. In recent years, the rates of antibiotic-resistant Gram-negative bacteria associated with biofilm-forming activity have increased worrisomely, particularly among healthcare-associated pathogens. Acinetobacter baumannii is a critically opportunistic pathogen, due to the high rates of antibiotic resistant strains causing healthcare-acquired infections (HAIs). The clinical isolates of A. baumannii can form biofilms on both biotic and abiotic surfaces; hospital settings and medical devices are the ideal environments for A. baumannii biofilms, thereby representing the main source of patient infections. However, the paucity of therapeutic options poses major concerns for human health infections caused by A. baumannii strains. The increasing number of multidrug-resistant A. baumannii biofilm-forming isolates in association with the limited number of biofilm-eradicating treatments intensify the need for effective antibiofilm approaches. This review discusses the mechanisms used by this opportunistic pathogen to form biofilms, describes their clinical impact, and summarizes the current and emerging treatment options available, both to prevent their formation and to disrupt preformed A. baumannii biofilms.
Collapse
Affiliation(s)
- Arianna Pompilio
- Center for Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, Service of Clinical Microbiology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.P.); (G.D.B.)
| | - Daniela Scribano
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy;
- Dani Di Giò Foundation-Onlus, 00193 Rome, Italy
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Giovanni Di Bonaventura
- Center for Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, Service of Clinical Microbiology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.P.); (G.D.B.)
| | - Anna Teresa Palamara
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy;
- Laboratory Affiliated to Institute Pasteur Italia-Cenci Bolognetti Foundation, Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Cecilia Ambrosi
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, IRCCS, 00166 Rome, Italy
| |
Collapse
|
24
|
Ilsan NA, Lee YJ, Kuo SC, Lee IH, Huang TW. Antimicrobial Resistance Mechanisms and Virulence of Colistin- and Carbapenem-Resistant Acinetobacter baumannii Isolated from a Teaching Hospital in Taiwan. Microorganisms 2021; 9:microorganisms9061295. [PMID: 34198665 PMCID: PMC8232278 DOI: 10.3390/microorganisms9061295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 01/01/2023] Open
Abstract
Acinetobacter baumannii, a Gram-negative bacterium, is an important nosocomial pathogen. Colistin-resistant A. baumannii is becoming a new concern, since colistin is one of the last-line antibiotics for infections by carbapenem-resistant A. baumannii. From 452 carbapenem-resistant isolates collected in a teaching hospital in Taipei, Taiwan, we identified seven that were resistant to colistin. Carbapenem resistance in these isolates is attributed to the presence of carbapenemase gene blaOXA-23 in their genomes. Colistin resistance is presumably conferred by mutations in the sensor kinase domain of PmrB found in these isolates, which are known to result in modification of colistin target lipid A via the PmrB-PmrA-PmrC signal transduction pathway. Overexpression of pmrC, eptA, and naxD was observed in all seven isolates. Colistin resistance mediated by pmrB mutations has never been reported in Taiwan. One of the seven isolates contained three mutations in lpxD and exhibited an altered lipopolysaccharide profile, which may contribute to its colistin resistance. No significant difference in growth rates was observed between the isolates and the reference strain, suggesting no fitness cost of colistin resistance. Biofilm formation abilities of the isolates were lower than that of the reference. Interestingly, one of the isolates was heteroresistant to colistin. Four of the isolates were significantly more virulent to wax moth larvae than the reference.
Collapse
Affiliation(s)
- Noor Andryan Ilsan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yuarn-Jang Lee
- Department of Internal Medicine, Division of Infectious Diseases, Taipei Medical University Hospital, Taipei 11031, Taiwan;
- Department of Internal Medicine, Division of Infectious Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan;
| | - I-Hui Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Tzu-Wen Huang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: ; Tel./Fax: +886-2-2736-1661 (ext. 3925); (ext. 3921)
| |
Collapse
|
25
|
Dickstein Y, Lellouche J, Ben Dalak Amar M, Schwartz D, Nutman A, Daitch V, Yahav D, Leibovici L, Skiada A, Antoniadou A, Daikos GL, Andini R, Zampino R, Durante-Mangoni E, Mouton JW, Friberg LE, Dishon Benattar Y, Bitterman R, Neuberger A, Carmeli Y, Paul M. Treatment Outcomes of Colistin- and Carbapenem-resistant Acinetobacter baumannii Infections: An Exploratory Subgroup Analysis of a Randomized Clinical Trial. Clin Infect Dis 2020; 69:769-776. [PMID: 30462182 DOI: 10.1093/cid/ciy988] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/15/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND We evaluated the association between mortality and colistin resistance in Acinetobacter baumannii infections and the interaction with antibiotic therapy. METHODS This is a secondary analysis of a randomized controlled trial of patients with carbapenem-resistant gram-negative bacterial infections treated with colistin or colistin-meropenem combination. We evaluated patients with infection caused by carbapenem-resistant A. baumannii (CRAB) identified as colistin susceptible (CoS) at the time of treatment and compared patients in which the isolate was confirmed as CoS with those whose isolates were retrospectively identified as colistin resistant (CoR) when tested by broth microdilution (BMD). The primary outcome was 28-day mortality. RESULTS Data were available for 266 patients (214 CoS and 52 CoR isolates). Patients with CoR isolates had higher baseline functional capacity and lower rates of mechanical ventilation than patients with CoS isolates. All-cause 28-day mortality was 42.3% (22/52) among patients with CoR strains and 52.8% (113/214) among patients with CoS isolates (P = .174). After adjusting for variables associated with mortality, the mortality rate was lower among patients with CoR isolates (odds ratio [OR], 0.285 [95% confidence interval {CI}, .118-.686]). This difference was associated with treatment arm: Mortality rates among patients with CoR isolates were higher in those randomized to colistin-meropenem combination therapy compared to colistin monotherapy (OR, 3.065 [95% CI, 1.021-9.202]). CONCLUSIONS Colistin resistance determined by BMD was associated with lower mortality among patients with severe CRAB infections. Among patients with CoR isolates, colistin monotherapy was associated with a better outcome compared to colistin-meropenem combination therapy. CLINICAL TRIALS REGISTRATION NCT01732250.
Collapse
Affiliation(s)
- Yaakov Dickstein
- National Institute for Antibiotic Resistance and Infection Control, Ministry of Health, Tel-Aviv
| | - Jonathan Lellouche
- National Institute for Antibiotic Resistance and Infection Control, Ministry of Health, Tel-Aviv.,Molecular Epidemiology Laboratory, Tel-Aviv Sourasky Medical Center
| | - Maayan Ben Dalak Amar
- National Institute for Antibiotic Resistance and Infection Control, Ministry of Health, Tel-Aviv.,Molecular Epidemiology Laboratory, Tel-Aviv Sourasky Medical Center
| | - David Schwartz
- National Institute for Antibiotic Resistance and Infection Control, Ministry of Health, Tel-Aviv.,Molecular Epidemiology Laboratory, Tel-Aviv Sourasky Medical Center
| | - Amir Nutman
- National Institute for Antibiotic Resistance and Infection Control, Ministry of Health, Tel-Aviv.,Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv
| | - Vered Daitch
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv.,Department of Medicine E, Beilinson Hospital, Petah Tikva, Israel
| | - Dafna Yahav
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv.,Unit of Infectious Diseases, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Leonard Leibovici
- Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv.,Department of Medicine E, Beilinson Hospital, Petah Tikva, Israel
| | - Anna Skiada
- First Department of Medicine, Laikon General Hospital, Athens, Greece.,National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Antoniadou
- National and Kapodistrian University of Athens, Athens, Greece.,Fourth Department of Medicine, Attikon University General Hospital, Athens, Greece
| | - George L Daikos
- First Department of Medicine, Laikon General Hospital, Athens, Greece.,National and Kapodistrian University of Athens, Athens, Greece
| | - Roberto Andini
- Department of Precision Medicine, University of Campania 'L Vanvitelli' and Azienda Ospedaliera di Rilievo Nazionale dei Colli-Monaldi Hospital, Napoli, Italy
| | - Rosa Zampino
- Department of Precision Medicine, University of Campania 'L Vanvitelli' and Azienda Ospedaliera di Rilievo Nazionale dei Colli-Monaldi Hospital, Napoli, Italy
| | - Emanuele Durante-Mangoni
- Department of Precision Medicine, University of Campania 'L Vanvitelli' and Azienda Ospedaliera di Rilievo Nazionale dei Colli-Monaldi Hospital, Napoli, Italy
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lena E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Sweden
| | - Yael Dishon Benattar
- Institute of Infectious Diseases, Rambam Health Care Campus.,Cheryl Spencer Department of Nursing, University of Haifa
| | - Roni Bitterman
- Institute of Infectious Diseases, Rambam Health Care Campus
| | - Ami Neuberger
- Institute of Infectious Diseases, Rambam Health Care Campus
| | - Yehuda Carmeli
- National Institute for Antibiotic Resistance and Infection Control, Ministry of Health, Tel-Aviv.,Molecular Epidemiology Laboratory, Tel-Aviv Sourasky Medical Center.,Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv
| | - Mical Paul
- Institute of Infectious Diseases, Rambam Health Care Campus.,Faculty of Medicine, Technion, Israel Institute of Technology, Haifa
| | | |
Collapse
|
26
|
Karakonstantis S. A systematic review of implications, mechanisms, and stability of in vivo emergent resistance to colistin and tigecycline in Acinetobacter baumannii. J Chemother 2020; 33:1-11. [PMID: 32677578 DOI: 10.1080/1120009x.2020.1794393] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The potential of A. baumannii for acquired resistance to last resort antibiotics (colistin and tigecycline) during treatment has important clinical implications, especially when dealing with patients failing to improve despite treatment with an active antimicrobial. However, the relevant literature remains scattered. Therefore, a systematic search was conducted in PubMed and Scopus. Several studies reported emergence of resistance to colistin or tigecycline during treatment, in most cases (86%) resulting in persistent or recurrent infections, especially in cases of emergent resistance without fitness cost. Lipopolysaccharide modification in the case of colistin and overexpression of efflux pumps in the case of tigecycline were the main mechanisms of resistance. Emergent colistin resistance is often associated with fitness cost which may result in re-emergence of the fitter and more virulent colistin susceptible strain after cessation of antibiotic pressure. Prospective studies are needed to determine the frequency of emergent resistance during treatment and its impact on patient outcomes.
Collapse
Affiliation(s)
- Stamatis Karakonstantis
- Internal Medicine Department, General Hospital of Heraklion Venizeleio, Heraklion, Crete, Greece.,School of medicine, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
27
|
Karakonstantis S, Saridakis I. Colistin heteroresistance in Acinetobacter spp.: systematic review and meta-analysis of the prevalence and discussion of the mechanisms and potential therapeutic implications. Int J Antimicrob Agents 2020; 56:106065. [PMID: 32599229 DOI: 10.1016/j.ijantimicag.2020.106065] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/20/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND Colistin is one of the few remaining options for carbapenem-resistant Acinetobacter baumannii (A. baumannii); however, emergence of resistance from heteroresistant populations is possible. This review aimed to systematically search and consolidate the literature on the prevalence, mechanisms and therapeutic implications of colistin heteroresistance in Acinetobacter spp. METHODS A systematic search was conducted in PubMed and Scopus. The pooled prevalence of colistin heteroresistance was calculated using meta-analysis of proportions with the Freeman-Tukey transformation and the random-effects (DerSimonian and Laird) method. RESULTS Based on 15 studies the prevalence of colistin heteroresistance was 33% (95% CI 16-53%) but considerable heterogeneity was observed (I2 = 96%, P < 0.001). Prior exposure to colistin was associated with a higher proportion of resistant subpopulations. Colistin heteroresistance may result from chromosomal mutations in resistant subpopulations (predominantly in PmrAB and lpx genes) resulting in lipopolysaccharide modification or loss, or overexpression of efflux pumps. No dosage scheme of colistin monotherapy can prevent the emergence of resistant subpopulations in vitro, but few studies have reported in vivo emergence of resistance from heteroresistant A. baumannii during treatment, and studies examining the correlation between heteroresistance and clinical/microbiological outcomes are lacking. Several colistin-based combinations have been shown in vitro to prevent the emergence of the resistant subpopulations but none have been translated so far into clinical benefit. Reasons for this discrepancy are discussed. CONCLUSIONS Colistin heteroresistance was common but highly variable between studies. The impact of colistin heteroresistance (frequency of emergent resistance during treatment and correlation with treatment outcomes) requires further study.
Collapse
Affiliation(s)
- Stamatis Karakonstantis
- School of Medicine, University of Crete, Heraklion, Crete, Greece; Internal Medicine Department, General Hospital of Heraklion Venizeleio, Heraklion, Crete, Greece.
| | - Ioannis Saridakis
- Internal Medicine Department, General Hospital of Heraklion Venizeleio, Heraklion, Crete, Greece
| |
Collapse
|
28
|
Kamoshida G, Akaji T, Takemoto N, Suzuki Y, Sato Y, Kai D, Hibino T, Yamaguchi D, Kikuchi-Ueda T, Nishida S, Unno Y, Tansho-Nagakawa S, Ubagai T, Miyoshi-Akiyama T, Oda M, Ono Y. Lipopolysaccharide-Deficient Acinetobacter baumannii Due to Colistin Resistance Is Killed by Neutrophil-Produced Lysozyme. Front Microbiol 2020; 11:573. [PMID: 32373082 PMCID: PMC7183746 DOI: 10.3389/fmicb.2020.00573] [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] [Received: 10/07/2019] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
Acinetobacter baumannii causes nosocomial infections due to its multidrug resistance and high environmental adaptability. Colistin is a polypeptide antibacterial agent that targets lipopolysaccharide (LPS) and is currently used to control serious multidrug-resistant Gram-negative bacterial infections, including those caused by A. baumannii. However, A. baumannii may acquire colistin resistance by losing their LPS. In mouse models, LPS-deficient A. baumannii have attenuated virulence. Nevertheless, the mechanism through which the pathogen is cleared by host immune cells is unknown. Here, we established colistin-resistant A. baumannii strains and analyzed possible mechanisms through which they are cleared by neutrophils. Colistin-resistant, LPS-deficient strains harbor mutations or insertion sequence (IS) in lpx genes, and introduction of intact lpx genes restored LPS deficiency. Analysis of interactions between these strains and neutrophils revealed that compared with wild type, LPS-deficient A. baumannii only weakly stimulated neutrophils, with consequent reduced levels of reactive oxygen species (ROS) and inflammatory cytokine production. Nonetheless, neutrophils preferentially killed LPS-deficient A. baumannii compared to wild-type strains. Moreover, LPS-deficient A. baumannii strains presented with increased sensitivities to antibacterial lysozyme and lactoferrin. We revealed that neutrophil-secreted lysozyme was the antimicrobial factor during clearance of LPS-deficient A. baumannii strains. These findings may inform the development of targeted therapeutics aimed to treat multidrug-resistant infections in immunocompromised patients who are unable to mount an appropriate cell-mediated immune response.
Collapse
Affiliation(s)
- Go Kamoshida
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan.,Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Takuya Akaji
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Norihiko Takemoto
- Pathogenic Microbe Laboratory, Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Yoshinori Sato
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Daichi Kai
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Taishi Hibino
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Daiki Yamaguchi
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Takane Kikuchi-Ueda
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Satoshi Nishida
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Yuka Unno
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Shigeru Tansho-Nagakawa
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Tsuneyuki Ubagai
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masataka Oda
- Department of Microbiology and Infection Control Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yasuo Ono
- Department of Microbiology and Immunology, School of Medicine, Teikyo University, Tokyo, Japan
| |
Collapse
|
29
|
Skariyachan S, Muddebihalkar AG, Badrinath V, Umashankar B, Eram D, Uttarkar A, Niranjan V. Natural epiestriol-16 act as potential lead molecule against prospective molecular targets of multidrug resistant Acinetobacter baumannii-Insight from in silico modelling and in vitro investigations. INFECTION GENETICS AND EVOLUTION 2020; 82:104314. [PMID: 32268193 DOI: 10.1016/j.meegid.2020.104314] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 03/05/2020] [Accepted: 04/01/2020] [Indexed: 01/09/2023]
Abstract
The current study aimed to identify putative drug targets of multidrug resistant Acinetobacter baumannii (MDRAb) and study the therapeutic potential of natural epiestriol-16 by computer aided virtual screening and in vitro studies. The clinical isolates (n = 5) showed extreme dug resistance to carbapenems and colistins (p ≤ .05). Computational screening suggested that out of 236 natural molecules selected, 06 leads were qualified for drug likeliness, pharmacokinetic features and one potential molecule namely natural epiestriol-16 (16b-Hydroxy-17a-estradiol) exhibited significant binding potential towards four prioritised drug targets in comparison with the binding of faropenem to their usual target. Natural epiestriol demonstrated profound binding to the outer membrane protein (Omp38), protein RecA (RecA), orotate phosphoribosyltransferase (PyrE) and orotidine 5'-phosphate decarboxylase (PyrF) with binding energy of -6.0, -7.3, -7.3 and -8.0 kcal/mol respectively. MD simulations suggested that 16-epiestriol-receptor complexes demonstrated stability throughout the simulation. The growth curve and time kill assays revealed that MDRAb showed resistance to faropenem and polymyxin-B and the pure epiestriol-16 showed significant inhibitory properties at a concentration of 200 μg/mL (p ≤ .5). Thus, natural epiestriol-16 can be used as potential inhibitor against the prioritised targets of MDRAb and this study provide insight for drug development against carbapenem and colistin resistant A. baumannii.
Collapse
Affiliation(s)
- Sinosh Skariyachan
- Department of Microbiology, St. Pius X College Rajapuram, Kasaragod, Kerala, India; Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, India.
| | - Aditi G Muddebihalkar
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, India
| | - Vaishnavi Badrinath
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, India
| | - Bindu Umashankar
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, India
| | - Daniya Eram
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, India
| | - Akshay Uttarkar
- Department of Biotechnology, RV College of Engineering, Bangalore, Karnataka, India
| | - Vidya Niranjan
- Department of Biotechnology, RV College of Engineering, Bangalore, Karnataka, India
| |
Collapse
|
30
|
Karthika S, Midhun SJ, Jisha MS. A potential antifungal and growth-promoting bacterium Bacillus sp. KTMA4 from tomato rhizosphere. Microb Pathog 2020; 142:104049. [PMID: 32045643 DOI: 10.1016/j.micpath.2020.104049] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 10/25/2022]
Abstract
Plant growth-promoting rhizobacteria are indigenous beneficial bacteria that will enhance plant growth as well as suppress phytopathogens. In the present study, the isolate KTMA4 showed the highest inhibition against major phytopathogens of tomato; Fusarium oxysporum (66%) and Alternaria solani (54%) after seven days of incubation. Analysis of the 16S rRNA gene sequence revealed that the isolate KTMA4 is Bacillus cereus (MG547975). The isolate produced in vitro plants growth-promoting factors such as Indole-3-acetic acid, ammonia, catalase, siderophore and 1-aminocyclopropane-1-carboxylate deaminase and it has nitrogen fixation ability. The bacterial strain has also produced lytic enzymes such as amylase, cellulase, xylanase, lipase, and protease. Moreover, the bacterium Bacillus cereus KTMA4 effectively produced biofilm, biosurfactants and salt-tolerant (5% NaCl). The bacterium exhibited intrinsic antibiotic resistance. The in vivo studies using tomato plants grown from seeds treated with the bacterial strain KTMA4 demonstrated an enhancement in seed germination percentage (86.66 ± 2.88) and vigour index (637.5 ± 21.65) over the uninoculated control (germination percentage- 28.33 ± 2.88 and vigour index- 42.5 ± 4.33). 60 days of greenhouse study revealed that the bacterial isolate enhanced the plant growth significantly (P ≤ 0.05) compared to the uninoculated control and the treated plants. Therefore the study suggests that the newly isolated rhizosphere bacterial strain can be used as a potential biocontrol agent against a multitude of fungal pathogens as well as a biofertilizer inoculant for tomato cultivation.
Collapse
Affiliation(s)
- S Karthika
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India
| | | | - M S Jisha
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India.
| |
Collapse
|
31
|
Pourhajibagher M, Partoazar A, Alaeddini M, Etemad-Moghadam S, Bahador A. Photodisinfection effects of silver sulfadiazine nanoliposomes doped-curcumin on Acinetobacter baumannii: a mouse model. Nanomedicine (Lond) 2020; 15:437-452. [DOI: 10.2217/nnm-2019-0315] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Aim: To evaluate the antimicrobial effects of photoexcited silver sulfadiazine nanoliposomes (AgSD-NLs) doped by curcumin (AgSD-NLs@Cur) on Acinetobacter baumannii. Materials & methods: Following characterization, the cytotoxic and hemolytic activities of AgSD-NLs@Cur were evaluated. The antimicrobial activities of AgSD-NLs@Cur-mediated antimicrobial photodynamic therapy (aPDT) were determined. Histopathological examination of the burn wound sites of infected mice treated with photoexcited AgSD-NLs@Cur was assessed. Results: No significant cytotoxic and hemolytic activities were observed. There was a decrease in the Acinetobacter baumannii count in planktonic and biofilm forms and the gene expression level using AgSD-NLs@Cur-aPDT (p < 0.05). Histopathological analysis indicated the epidermis developed markedly and the bacterial load decreased significantly after aPDT. Conclusion: Photoexcited AgSD-NLs@Cur has an antimicrobial potential against A. baumannii.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Partoazar
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Alaeddini
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahroo Etemad-Moghadam
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Bahador
- Oral Microbiology Laboratory, Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Nikfarjam S, Tohidkia MR, Mehdipour T, Soleimani R, Rahimi AAR, Nouri M. ---Successful Application of Whole Cell Panning for Isolation of Phage Antibody Fragments Specific to Differentiated Gastric Cancer Cells. Adv Pharm Bull 2019; 9:624-631. [PMID: 31857967 PMCID: PMC6912189 DOI: 10.15171/apb.2019.072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 11/24/2022] Open
Abstract
Purpose: Generation of antibodies which potentially discriminate between malignant and healthy cells is an important prerequisite for early diagnosis and treatment of gastric cancer (GC). Comparative analysis of cell surface protein landscape will provide an experimental basis for biomarker discovery, which is essential for targeted molecular therapies. This study aimed to isolate phage-displayed antibody fragments recognizing cell surface proteins, which were differently expressed between two closely related GC cell lines, namely AGS and MKN-45. Methods: We selected and screened a semisynthetic phage-scFv library on AGS, MKN-45, and NIH-3T3 cell lines by utilizing a tailored selection scheme that was designed to isolate phagescFvs that not only recognize the differentiated AGS cells but also distinguish them from NIH3T3 fibroblasts and the poorly differentiated MKN-45 cells. Results: After four rounds of subtractive whole cell panning, 14 unique clones were identified by ELISA screening and nucleotide sequencing. For further characterization, we focused on four phage-scFvs with strong signals in screening, and their specificity was confirmed by cell-based ELISA. Furthermore, the selected phage-scFvs were able to specifically stain AGS cells with 38.74% (H1), 11.04% (D11), 76.93% (G11), and 69.03% (D1) in flow cytometry analysis which supported the ability of these phage scFvs in distinguishing AGS from MKN-45 and NIH-3T3 cells. Conclusion: Combined with other proteomic techniques, these phage-scFvs can be applied to membrane proteome analysis and, subsequently, identification of novel tumor-related antigens mediating proliferation and differentiation of cells. Furthermore, such antibody fragments can be exploited for diagnostic purposes as well as targeted drug delivery of GC.
Collapse
Affiliation(s)
- Sepideh Nikfarjam
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tayebeh Mehdipour
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Soleimani
- Department of Molecular Biology, Research and Diagnostic Laboratory of Dook, Sari, Iran
| | - Ali Akbar Rahim Rahimi
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Taheri B, Mohammadi M, Momenzadeh N, Farshadzadeh Z, Roozbehani M, Dehghani P, Hajian S, Darvishi S, Shamseddin J. Substitution of lysine for isoleucine at the center of the nonpolar face of the antimicrobial peptide, piscidin-1, leads to an increase in the rapidity of bactericidal activity and a reduction in toxicity. Infect Drug Resist 2019; 12:1629-1647. [PMID: 31354312 PMCID: PMC6585414 DOI: 10.2147/idr.s195872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 03/25/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose: Piscidin-1 is an effective antimicrobial peptide (AMP) against a variety of microbes. However, its toxicity has been reported as a limitation for its potential therapeutic applications. The toxicity of piscidin-1 may be related to the long nonpolar face of this AMP. Here, we investigated different piscidin-1 analogs to reach a peptide with the reduced toxicity. Material and methods: In vitro and in vivo antibacterial activity and toxicity of piscidin-1 analogs generated by replacement of isoleucine at the border (I9) or the center (I16) of the nonpolar face of piscidin-1 by alanine or lysine were investigated. Results: The results indicated that among all peptides, piscidin-1 with the highest HPLC retention time (RT) and I16K-piscidin-1 with the lowest RT had the highest and lowest cytotoxicity, respectively. Although I16K-piscidin-1 possessed the same MIC value as the parent peptide (piscidin-1) and other analogs, I16K-piscidin-1 exhibited a higher rapidity of bactericidal action at 5×MIC. The β-galactosidase leakage and propidium iodide staining assays indicated a higher pore-forming capacity of I16K-piscidin-1 relative to the parent peptide (piscidin-1). Taken together, RT is suggested to have a direct association with the toxicity and an inverse association with the rapidity of bactericidal action and pore-forming capacity. After infection of mice with clinical colistin-resistant Acinetobacter baumannii or clinical methicillin-resistant Staphylococcus aureus strains, treatment with I16K-piscidin-1, but not piscidin-1 and other analogs, resulted in a significantly stronger bactericidal potency. Furthermore, I16K-piscidin-1 exhibited the lowest in vivo toxicity. Conclusion: Overall, in vitro and in vivo comparison of piscidin-1 and its analogs together documented that replacement of isoleucine at the center of the nonpolar face of piscidin-1(I16) by lysine leads to not only a decrease in toxicity potential but also an increase in bactericidal potential.
Collapse
Affiliation(s)
- Behrouz Taheri
- Department of Medical Laboratory Sciences, School of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohsen Mohammadi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Niloofar Momenzadeh
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Zahra Farshadzadeh
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mona Roozbehani
- Department of Medical Parasitology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parva Dehghani
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Sobhan Hajian
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Sadegh Darvishi
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Jebreil Shamseddin
- Infection and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
34
|
Nadhe SB, Singh R, Wadhwani SA, Chopade BA. Acinetobacter sp. mediated synthesis of AgNPs, its optimization, characterization and synergistic antifungal activity against C. albicans. J Appl Microbiol 2019; 127:445-458. [PMID: 31074075 DOI: 10.1111/jam.14305] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 04/25/2019] [Accepted: 05/06/2019] [Indexed: 12/21/2022]
Abstract
AIMS To synthesize silver nanoparticles (AgNPs) with cell free extract of Acinetobacter sp. and evaluate antifungal activity against planktonic and biofilm of Candida. Also, to study mechanism of antifungal action of AgNPs. METHODS AND RESULT Acinetobacter spp were screened for synthesis of AgNPs. Physio-chemical parameters were optimized to obtained monodispersed nanoparticles. Optimized nanoparticles were characterized using spectroscopic, microscopic and diffraction techniques. Antifungal and biofilm disruption activity of AgNPs (10 ± 5 nm) were investigated against C. albicans. Mechanism of antifungal activity of nanosilver was deduced by growth curve, reactive oxygen species generation, thiol interaction and microscopic analysis. Acinetobacter sp. GWRFH 45 gave maximum synthesis of AgNPs. At optimized condition monodispersed, spherical nanoparticles were obtained which were crystalline with negative surface charge. AgNPs exhibited antifungal activity against planktonic cells and biofilm of Candida. AgNPs showed synergistic effect with amphotericin B as well as fluconazole against biofilm disruption. AgNPs were found to affect growth of Candida, generate reactive oxygen species and disrupt cellular morphology. CONCLUSIONS Cell free extract of A. calcoaceticus GWRFH 45 has ability to synthesize AgNPs. AgNPs alone and in combination with drugs have potential to inhibit C. albicans. SIGNIFICANCE AND IMPACT OF THE STUDY This is the first report of bacteriogenic AgNPs used in combination with antifungal drugs against Candida.
Collapse
Affiliation(s)
- S B Nadhe
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - R Singh
- Department of Biotechnology, SIES College of Arts Science and Commerce, Mumbai, Maharashtra, India
| | - S A Wadhwani
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - B A Chopade
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, India.,Dr. Babasaheb Ambedkar, Marathwada University, Aurangabad, Maharashtra, India
| |
Collapse
|
35
|
Ušjak D, Ivković B, Božić DD, Bošković L, Milenković M. Antimicrobial activity of novel chalcones and modulation of virulence factors in hospital strains of Acinetobacter baumannii and Pseudomonas aeruginosa. Microb Pathog 2019; 131:186-196. [PMID: 30980878 DOI: 10.1016/j.micpath.2019.04.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 01/01/2023]
Abstract
Acinetobacter baumannii and Pseudomonas aeruginosa are frequent multiresistant nosocomial pathogens that cause wound and pulmonary infections in hospitalized patients. As being increasingly resistant to most clinically available antibiotics, there is a constant need for exploration of new substances that could kill them or inhibit their growth, or alternatively inhibit some of their essential virulence factors. Chalcones are chemical compounds with well-documented antimicrobial potential. The aim of this study was to examine effectiveness of four newly-synthesized chalcones against the multiresistant clinical strains of A. baumannii and P. aeruginosa. Antibacterial activity of chalcones was investigated with broth-microdilution test and time-dependent killing assay. Synergistic effects of tested compounds with antibiotics (meropenem, amikacin and ciprofloxacin) were determined by checkerboard assay. The effects of chalcones on expression of virulence factors in P. aeruginosa (pyocyanin production, swimming and swarming motility) and A. baumannii (twitching and surface-associated motility), along with their biofilm production, were also examined. The obtained results indicate substantial antimicrobial activity of the tested chalcones (MICs = 100-175 μg/mL) and several synergistic interactions with antibiotics, as well as notable reduction in expression of all investigated virulence factors. These promising results may constitute a good basis for further research.
Collapse
Affiliation(s)
- Dušan Ušjak
- Department of Microbiology and Immunology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Branka Ivković
- Department of Pharmaceutical Chemistry, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Dragana D Božić
- Department of Microbiology and Immunology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Lidija Bošković
- Department of Laboratory Diagnostics, Clinical Hospital Centre "Dr. Dragiša Mišović", Heroja Milana Tepića 1, 11000, Belgrade, Serbia
| | - Marina Milenković
- Department of Microbiology and Immunology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia.
| |
Collapse
|
36
|
Dose Optimization of Colistin Combinations against Carbapenem-Resistant Acinetobacter baumannii from Patients with Hospital-Acquired Pneumonia in China by Using an In Vitro Pharmacokinetic/Pharmacodynamic Model. Antimicrob Agents Chemother 2019; 63:AAC.01989-18. [PMID: 30745385 DOI: 10.1128/aac.01989-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/19/2019] [Indexed: 12/13/2022] Open
Abstract
Colistin-based combination therapy has become an important strategy to combat the carbapenem-resistant Acinetobacter baumannii (CRAB). However, the optimal dosage regimen selection for the combination with maximum efficacy is challenging. Checkerboard assay was employed to evaluate the synergy of colistin in combination with meropenem, rifampin, fosfomycin, and minocycline against nine carbapenem-resistant A. baumannii isolates (MIC of meropenem [MICMEM], ≥32 mg/liter) isolated from Chinese hospital-acquired pneumonia (HAP) patients. A static time-kill assay, in vitro dynamic pharmacokinetic/pharmacodynamic (PK/PD) model, and semimechanistic PK/PD modeling were conducted to predict and validate the synergistic effect of the most efficacious combination. Both checkerboard and static time-kill assays demonstrated the superior synergistic effect of the colistin-meropenem combination against all CRAB isolates. In the in vitro PK/PD model, the dosage regimen of 2 g meropenem daily via 3-h infusion combined with steady-state 1 mg/liter colistin effectively suppressed the bacterial growth at 24 h with a 2-log10 decrease, compared with the initial inocula against two CRAB isolates. The semimechanistic PK/PD model predicted that more than 2 mg/liter colistin combined with meropenem (2 g, 3-h infusion) was required to achieve the killing below the limit of detection (<LOD; i.e., 1 log10CFU/ml) at 24 h with an MICMEM of ≥32 mg/liter. Colistin combined with meropenem exerted synergistic killing against CRAB even with an MICMEM of ≥32 mg/liter and MIC of colistin (MICCST) of ≤1 mg/liter. However, it is predicted that a higher concentration of colistin combined with meropenem was crucial to kill bacteria to <LOD. Our study provides important PK/PD information for optimization of the colistin and meropenem combination against CRAB.
Collapse
|
37
|
Li FJ, Starrs L, Burgio G. Tug of war between Acinetobacter baumannii and host immune responses. Pathog Dis 2019; 76:5290314. [PMID: 30657912 DOI: 10.1093/femspd/ftz004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/16/2019] [Indexed: 02/06/2023] Open
Abstract
Acinetobacter baumannii is an emerging nosocomial, opportunistic pathogen with growing clinical significance. Acinetobacter baumannii has an exceptional ability to rapidly develop drug resistance and to adhere to abiotic surfaces, including medical equipment, significantly promoting bacterial spread and also limiting our ability to control A. baumannii infections. Consequently, A. baumannii is frequently responsible for ventilator-associated pneumonia in clinical settings. In order to develop an effective treatment strategy, understanding host-pathogen interactions during A. baumannii infection is crucial. Various A. baumannii virulence factors have been identified as targets of host innate pattern-recognition receptors, which leads to activation of downstream inflammasomes to develop inflammatory responses, and the recruitment of innate immune effectors against A. baumannii infection. To counteract host immune attack, A. baumannii regulates its expression of different virulence factors. This review summarizes the significance of mechanisms of host-bacteria interaction, as well as different bacteria and host defense mechanisms during A. baumannii infection.
Collapse
Affiliation(s)
- Fei-Ju Li
- Department of Immunology and infectious Diseases, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Acton, ACT 2601, Australia
| | - Lora Starrs
- Department of Immunology and infectious Diseases, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Acton, ACT 2601, Australia
| | - Gaetan Burgio
- Department of Immunology and infectious Diseases, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Acton, ACT 2601, Australia
| |
Collapse
|
38
|
Zhao X, Yu Y, Zhang X, Huang B, Bai P, Xu C, Li D, Zhang B, Liu C. Decreased biofilm formation ability of Acinetobacter baumannii after spaceflight on China's Shenzhou 11 spacecraft. Microbiologyopen 2018; 8:e00763. [PMID: 30379419 PMCID: PMC6562233 DOI: 10.1002/mbo3.763] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/30/2022] Open
Abstract
China has prepared for construction of a space station by the early 2020s. The mission will require astronauts to stay on the space station for at least 180 days. Microbes isolated from the International Space Station (ISS) have shown profound resistance to clinical antibiotics and environmental stresses. Previous studies have demonstrated that the space environment could affect microbial survival, growth, virulence, biofilms, metabolism, as well as their antibiotic‐resistant phenotypes. Furthermore, several studies have reported that astronauts experience a decline in their immunity during long‐duration spaceflights. Monitoring microbiomes in the ISS or the spacecraft will be beneficial for the prevention of infection among the astronauts during spaceflight. The development of a manned space program worldwide not only provides an opportunity to investigate the impact of this extreme environment on opportunistic pathogenic microbes, but also offers a unique platform to detect mutations in pathogenic bacteria. Various microorganisms have been carried on a spacecraft for academic purposes. Acinetobacter baumannii is a common multidrug‐resistant bacterium often prevalent in hospitals. Variations in the ability to cope with environmental hazards increase the chances of microbial survival. Our study aimed to compare phenotypic variations and analyze genomic and transcriptomic variations in A. baumannii among three different groups: SS1 (33 days on the Shenzhou 11 spacecraft), GS1 (ground control), and Aba (reference strain). Consequently, the biofilm formation ability of the SS1 strain decreased after 33 days of spaceflight. Furthermore, high‐throughput sequencing revealed that some differentially expressed genes were downregulated in the SS1 strain compared with those in the GS1 strain. In conclusion, this present study provides insights into the environmental adaptation of A. baumannii and might be useful for understanding changes in the opportunistic pathogenic microbes on our spacecraft and on China's future ISS.
Collapse
Affiliation(s)
- Xian Zhao
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Yi Yu
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Xuelin Zhang
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Bing Huang
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Po Bai
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Chou Xu
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Diangeng Li
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Bin Zhang
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| | - Changting Liu
- Nanlou Respiratory Diseases Department, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
39
|
The Relationship Between Antibiotic Resistance Phenotypes and Biofilm Formation Capacity in Clinical Isolates of Acinetobacter baumannii. Jundishapur J Microbiol 2018. [DOI: 10.5812/jjm.74315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|