1
|
Weng B, Li Y, Feng W, Yao P, Wang Y, Wang Q, Wang X, Li Y, Li L, Wang Q. Azithromycin inhibits the intracellular persistence of Acinetobacter baumannii by inducing host cell autophagy in human bronchial epithelial cells. Microb Pathog 2025; 198:107152. [PMID: 39586339 DOI: 10.1016/j.micpath.2024.107152] [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: 08/28/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
The invasion of host cells by bacteria, leading to intracellular infections, is a major cause of infection recurrence. Drug-resistant Acinetobacter baumannii (A. baumannii) is one of the most challenging public health issues worldwide, with very limited clinical treatment options available. A. baumannii has been found to be able to invade host cells and proliferate within them in recent studies. In addition to the direct antimicrobial effect of antibiotics, the activation of host autophagic flux also plays an important role in eliminating intracellular pathogens. Herein, this study aimes to evaluate the clearance effect of antibiotics on intracellular A. baumannii both in vivo and in vitro, and explore the relationship between this effect and autophagy. The results showed that intracellular pathogens resulted in a significant increase in the minimum bactericidal concentration, while azithromycin can significantly eliminate intracellular A. baumannii in vitro and in vivo. Notably, 60 μg/mL azithromycin demonstrated intracellular clearance against multidrug-resistant A. baumannii and markedly induced autophagosomes in BEAS-2B cells with a mild stimulation of autophagosomes degradation. These findings indicated that azithromycin can significantly clear intracellular A. baumannii and its ability to clear intracellular A. baumannii may be related to the stimulation of autophagosome formation and the induction of host autophagy, which has important implications for the clinical treatment of A. baumannii infections, especially when intracellular infections are present.
Collapse
Affiliation(s)
- Bangbi Weng
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yuliang Li
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Wei Feng
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Pu Yao
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yu Wang
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qianmei Wang
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaowen Wang
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yang Li
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Li Li
- Department of Pain Medicine, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qian Wang
- Department of Pharmacy, Southwest Hospital of Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
2
|
Liu L, Huang Y, Wang Y, Jiang Y, Liu K, Pei Z, Li Z, Zhu Y, Liu D, Li X. Molecular Epidemiology and Genetic Characterization of Carbapenem-Resistant Acinetobacter baumannii Isolates from the ICU of a Tertiary Hospital in East China. Infect Drug Resist 2024; 17:5925-5945. [PMID: 39759767 PMCID: PMC11699857 DOI: 10.2147/idr.s491858] [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: 09/25/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
Purpose To evaluate the clinical characteristics, antimicrobial resistance (AMR) phenotypes and genotypes, and homology features of carbapenem-resistant Acinetobacter baumannii (CRAB) in intensive care unit (ICU) and to provide basis for effectively prevention, control and treatment of nosocomial infections caused by CRAB. Methods A total of 39 CRAB strains isolated from hospitalized patients in the ICU and neurosurgical ICU (NICU) between 2020 and 2023 were subjected to antimicrobial susceptibility testing and whole-genome sequencing (WGS). Virulence factor genes (VFGs), antimicrobial resistance genes (ARGs), multilocus sequencing typing (MLST), complete genome multilocus sequencing typing (cgMLST), average nucleotide identity (ANI), and single nucleotide polymorphism (SNP) analyses were performed using WGS. Results All CRAB strains were 100% resistant to ciprofloxacin, ceftazidime, piperacillin/tazobactam, and ticarcillin/clavulanic acid. A total of 48 antimicrobial resistance genes (ARGs) were found in the 39 CRAB strains, including blaOXA-66, blaOXA-23, blaADC-30, blaADC-73, gyrA, ant(3″)-IIa, aph(3″)-Ib, aph(6)-Id, tetB, tetR, sul1, sul2, LpxC and LpxA which confered resistance to carbapenems, cephalosporins, fluoroquinolones, aminoglycosides, tetracycline and sulfonamides. There were 128 VFGs, including genes encoding the AdeFGH efflux pump, lipopolysaccharide (LpsBLC), outer membrane protein A (OmpA), penicillin-binding protein (PbpG), biofilm-associated proteins (bap, pgaBCD, CsuABCDE), type VI secretion system protein (Tss), quorum sensing protein (AbaI/AbaR). Six clonal lineages were identified by Oxford MLST method, whereas one sequence type (ST2) was identified using the Pasteur MLST method. ANI analysis, heat map of SNP analysis, and phylogenetic tree based on core SNP revealed six clusters, and the strain classification results were consistent with these different methods. Ten clonal lineages were identified by cgMLST. Conclusion The CRAB strains were ST2 clones accompanied by severe resistance to commonly used antibiotics and abundant ARGs and VFGs in genotype. Strict measures should be implemented to prevent and control transmissions and infections. CgMLST and SNPs analyses showed excellent discriminatory power in homology analysis.
Collapse
Affiliation(s)
- Lili Liu
- Department of Nosocomial Infection, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Yuan Huang
- Department of Science and Education, Anqing Municipal Hospital, Anqing City, Anhui Province, People’s Republic of China
| | - Yaping Wang
- Department of Clinical Laboratory, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Yunlan Jiang
- Department of Nosocomial Infection, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Kang Liu
- Department of Clinical Laboratory, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Zhongxia Pei
- Department of Nosocomial Infection, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Zhiping Li
- Department of Nosocomial Infection, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Yuqiong Zhu
- Department of Nosocomial Infection, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Dan Liu
- Department of Nosocomial Infection, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| | - Xiaoyue Li
- Subdean Office, Anqing First People’s Hospital of Anhui Medical University, Anqing City, Anhui Province, People’s Republic of China
| |
Collapse
|
3
|
Huang Q, Zhu L, Huang F, Zhao Y, Wang H, Luan S, Xiao C. Novel quinazolin-6-yl Isoindolinone: Altering polysaccharide chemstructure for antibacterial efficacy against Staphylococcus aureus. Int J Biol Macromol 2024; 280:135650. [PMID: 39278453 DOI: 10.1016/j.ijbiomac.2024.135650] [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: 08/02/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
The ongoing development of novel strategies to combat Staphylococcus aureus and eliminate its biofilm formation has gained significant attention for human health. Antibiotic-resistant S. aureus necessitates the development of novel antibacterial agents with new mechanism of action. This study introduced a promising recently synthesized quinazolin-6-yl isoindolinone (IQE-X1), which exhibited potent antibacterial and antibiofilm efficacy with average median inhibitory concentration (IC50) of 3.37 μg mL-1 and minimal inhibitory concentration (MIC) of 12.5 μg mL-1, coupled with its ability to reduce cell surface hydrophobicity. IQE-X1 dose-dependently decreased extracellular polysaccharides (EPS) and its component monosaccharides, including rhamnose, arabinose, glucosamine, galactose, glucose, xylose, mannose, and ribose, accompanied by an increase in capsular polysaccharides (CP) and its individual monosaccharides, especially glucosamine. IQE-X1 demonstrated specificity in modulating the structural profiles of EPS and CP by altering the compositional ratios of their component monosaccharides. The potential mechanism of polysaccharide modulation was preliminarily elucidated through the response of β-N-acetylaminoglucosidase to IQE-X1 and their direct binding interaction. These findings provide new insights into the potential manipulation of the chemstructure of these biologically important macromolecules, EPS and CP, and highlight the antibacterial potential of IQE-X1 as a polysaccharide modulator for the development of more effective polysaccharide-targeted strategies against S. aureus.
Collapse
Affiliation(s)
- Qingchun Huang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China.
| | - Lisong Zhu
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Fengcheng Huang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Yanjun Zhao
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Hongye Wang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Shaorong Luan
- School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Ciying Xiao
- School of Biotechnology, East China University of Science and Technology, Shanghai 200237, PR China
| |
Collapse
|
4
|
Saoudi B, Bariz K, Saci S, Belounis Y, Ait Issad H, Abbaci M, Mustapha MA, Nabti EH, Alenazy R, Alhussaini MS, Alyahya AAI, Alqasmi M, Alhumaidi MS, Almufarriji FM, Houali K. Enhancing Antibiotic Efficacy and Combating Biofilm Formation: Evaluating the Synergistic Potential of Origanum vulgare Essential Oil against Multidrug-Resistant Gram-Negative Bacteria. Microorganisms 2024; 12:1651. [PMID: 39203493 PMCID: PMC11356740 DOI: 10.3390/microorganisms12081651] [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: 07/30/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Multidrug-resistant (MDR) Gram-negative bacteria remain a global public health issue due to the barrier imposed by their outer membrane and their propensity to form biofilms. It is becoming imperative to develop new antibacterial strategies. In this context, this study aims to evaluate the antibacterial efficacy of Origanum vulgare essential oil (OEO), alone and in combination with antibiotics, as well as its antibiofilm action against multidrug-resistant Gram-negative strains. OEO components were identified by gas chromatography-mass spectrometry (GC-MS), and antibacterial activity was assessed using the agar diffusion test and the microdilution method. Interactions between OEO and antibiotics were examined using the checkerboard method, while antibiofilm activity was analyzed using the crystal violet assay. Chemical analysis revealed that carvacrol was the major compound in OEO (61.51%). This essential oil demonstrated activity against all the tested strains, with inhibition zone diameters (IZDs) reaching 32.3 ± 1.5 mm. The combination of OEO with different antibiotics produced synergistic and additive effects, leading to a reduction of up to 98.44% in minimum inhibitory concentrations (MICs). In addition, this essential oil demonstrated an ability to inhibit and even eradicate biofilm formation. These results suggest that OEO could be exploited in the development of new molecules, combining its metabolites with antibiotics.
Collapse
Affiliation(s)
- Bilal Saoudi
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Karim Bariz
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Sarah Saci
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Yousra Belounis
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| | - Hakima Ait Issad
- Laboratoire Ressources Naturelles, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria;
| | - Mohamed Abbaci
- Centre de Recherche Scientifique et Technique en Analyses Physico—Chimiques CRAPC, Bou Ismail 42004, Algeria; (M.A.); (M.A.M.)
| | - Mohamed Abou Mustapha
- Centre de Recherche Scientifique et Technique en Analyses Physico—Chimiques CRAPC, Bou Ismail 42004, Algeria; (M.A.); (M.A.M.)
| | - El-Hafid Nabti
- Laboratoire de Maitrise des Energies Renouvelables, Faculté des Sciences de la Nature et de le Vie, Université de Bejaïa, Bejaïa 06000, Algeria;
| | - Rawaf Alenazy
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Mohammed Sanad Alhussaini
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Abdulrahman A. I. Alyahya
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Mohammed Alqasmi
- Department of Medical Laboratory, College of Applied Medical Sciences-Shaqra, Shaqra University, Shaqra 11961, Saudi Arabia; (M.S.A.); (A.A.I.A.); (M.A.)
| | - Maryam S. Alhumaidi
- Department of Biology, College of Science, University of Hafr Al Batin, P.O. Box 1803, Hafr Al Batin 31991, Saudi Arabia;
| | - Fawaz M. Almufarriji
- Medical Laboratories Department, College of Applied Medical Sciences in Al-Quwayiyah, Shaqra University, Shaqra 11961, Saudi Arabia;
| | - Karim Houali
- Laboratory of Analytical Biochemistry and Biotechnology, Faculty of Biological and Agronomic Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria; (B.S.); (K.B.); (S.S.); (Y.B.)
| |
Collapse
|
5
|
Rajangam SL, Narasimhan MK. Current treatment strategies for targeting virulence factors and biofilm formation in Acinetobacter baumannii. Future Microbiol 2024; 19:941-961. [PMID: 38683166 PMCID: PMC11290764 DOI: 10.2217/fmb-2023-0263] [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: 11/24/2023] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
A higher prevalence of Acinetobacter baumannii infections and mortality rate has been reported recently in hospital-acquired infections (HAI). The biofilm-forming capability of A. baumannii makes it an extremely dangerous pathogen, especially in device-associated hospital-acquired infections (DA-HAI), thereby it resists the penetration of antibiotics. Further, the transmission of the SARS-CoV-2 virus was exacerbated in DA-HAI during the epidemic. This review specifically examines the complex interconnections between several components and genes that play a role in the biofilm formation and the development of infections. The current review provides insights into innovative treatments and therapeutic approaches to combat A. baumannii biofilm-related infections, thereby ultimately improving patient outcomes and reducing the burden of HAI.
Collapse
Affiliation(s)
- Seetha Lakshmi Rajangam
- Department of Genetic Engineering, School of Bioengineering, College of Engineering & Technology, SRM Institute of Science & Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Manoj Kumar Narasimhan
- Department of Genetic Engineering, School of Bioengineering, College of Engineering & Technology, SRM Institute of Science & Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| |
Collapse
|
6
|
Mushtaq F, Nadeem A, Yabrag A, Bala A, Karah N, Zlatkov N, Nyunt Wai S, Uhlin BE, Ahmad I. Colony phase variation switch modulates antimicrobial tolerance and biofilm formation in Acinetobacter baumannii. Microbiol Spectr 2024; 12:e0295623. [PMID: 38205963 PMCID: PMC10845969 DOI: 10.1128/spectrum.02956-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/09/2023] [Indexed: 01/12/2024] Open
Abstract
Carbapenem-resistant Acinetobacter baumannii causes one of the most difficult-to-treat nosocomial infections. Polycationic drugs like polymyxin B or colistin and tetracycline drugs such as doxycycline or minocycline are commonly used to treat infections caused by carbapenem-resistant A. baumannii. Here, we show that a subpopulation of cells associated with the opaque/translucent colony phase variation by A. baumannii AB5075 displays differential tolerance to subinhibitory concentrations of colistin and tetracycline. Using a variety of microscopic techniques, we demonstrate that extracellular polysaccharide moieties mediate colistin tolerance to opaque A. baumannii at single-cell level and that mushroom-shaped biofilm structures protect opaque bacteria at the community level. The colony switch phenotype is found to alter several traits of A. baumannii, including long-term survival under desiccation, tolerance to ethanol, competition with Escherichia coli, and intracellular survival in the environmental model host Acanthamoeba castellanii. Additionally, our findings suggest that extracellular DNA associated with membrane vesicles can promote colony switching in a DNA recombinase-dependent manner.IMPORTANCEAs a WHO top-priority drug-resistant microbe, Acinetobacter baumannii significantly contributes to hospital-associated infections worldwide. One particularly intriguing aspect is its ability to reversibly switch its colony morphotype on agar plates, which has been remarkably underexplored. In this study, we employed various microscopic techniques and phenotypic assays to investigate the colony phase variation switch under different clinically and environmentally relevant conditions. Our findings reveal that the presence of a poly N-acetylglucosamine-positive extracellular matrix layer contributes to the protection of bacteria from the bactericidal effects of colistin. Furthermore, we provide intriguing insights into the multicellular lifestyle of A. baumannii, specifically in the context of colony switch variation within its predatory host, Acanthamoeba castellanii.
Collapse
Affiliation(s)
- Fizza Mushtaq
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
| | - Aftab Nadeem
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Abdelbasset Yabrag
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Anju Bala
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Nabil Karah
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Nikola Zlatkov
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Sun Nyunt Wai
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Bernt Eric Uhlin
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Irfan Ahmad
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
7
|
Ahmad I, Nadeem A, Mushtaq F, Zlatkov N, Shahzad M, Zavialov AV, Wai SN, Uhlin BE. Csu pili dependent biofilm formation and virulence of Acinetobacter baumannii. NPJ Biofilms Microbiomes 2023; 9:101. [PMID: 38097635 PMCID: PMC10721868 DOI: 10.1038/s41522-023-00465-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Acinetobacter baumannii has emerged as one of the most common extensive drug-resistant nosocomial bacterial pathogens. Not only can the bacteria survive in hospital settings for long periods, but they are also able to resist adverse conditions. However, underlying regulatory mechanisms that allow A. baumannii to cope with these conditions and mediate its virulence are poorly understood. Here, we show that bi-stable expression of the Csu pili, along with the production of poly-N-acetyl glucosamine, regulates the formation of Mountain-like biofilm-patches on glass surfaces to protect bacteria from the bactericidal effect of colistin. Csu pilus assembly is found to be an essential component of mature biofilms formed on glass surfaces and of pellicles. By using several microscopic techniques, we show that clinical isolates of A. baumannii carrying abundant Csu pili mediate adherence to epithelial cells. In addition, Csu pili suppressed surface-associated motility but enhanced colonization of bacteria into the lungs, spleen, and liver in a mouse model of systemic infection. The screening of c-di-GMP metabolizing protein mutants of A. baumannii 17978 for the capability to adhere to epithelial cells led us to identify GGDEF/EAL protein AIS_2337, here denoted PdeB, as a major regulator of Csu pili-mediated virulence and biofilm formation. Moreover, PdeB was found to be involved in the type IV pili-regulated robustness of surface-associated motility. Our findings suggest that the Csu pilus is not only a functional component of mature A. baumannii biofilms but also a major virulence factor promoting the initiation of disease progression by mediating bacterial adherence to epithelial cells.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden.
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan.
| | - Aftab Nadeem
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
| | - Fizza Mushtaq
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
| | - Nikola Zlatkov
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
| | - Muhammad Shahzad
- Department of Pharmacology, University of Health Sciences, Lahore, Pakistan
| | - Anton V Zavialov
- Department of Biochemistry, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Sun Nyunt Wai
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden
| | - Bernt Eric Uhlin
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
| |
Collapse
|
8
|
Chen X, Xue J, Dong X, Lu P. Uncovering virulence factors in Cronobacter sakazakii: insights from genetic screening and proteomic profiling. Appl Environ Microbiol 2023; 89:e0102823. [PMID: 37750707 PMCID: PMC10617496 DOI: 10.1128/aem.01028-23] [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: 06/20/2023] [Accepted: 07/15/2023] [Indexed: 09/27/2023] Open
Abstract
The increasing problem of antibiotic resistance has driven the search for virulence factors in pathogenic bacteria, which can serve as targets for the development of new antibiotics. Although whole-genome Tn5 transposon mutagenesis combined with phenotypic assays has been a widely used approach, its efficiency remains low due to labor-intensive processes. In this study, we aimed to identify specific genes and proteins associated with the virulence of Cronobacter sakazakii, a pathogenic bacterium known for causing severe infections, particularly in infants and immunocompromised individuals. By employing a combination of genetic screening, comparative proteomics, and in vivo validation using zebrafish and rat models, we rapidly screened highly virulent strains and identified two genes, rcsA and treR, as potential regulators of C. sakazakii toxicity toward zebrafish and rats. Proteomic profiling revealed upregulated proteins upon knockout of rcsA and treR, including FabH, GshA, GppA, GcvH, IhfB, RfaC, MsyB, and three unknown proteins. Knockout of their genes significantly weakened bacterial virulence, confirming their role as potential virulence factors. Our findings contribute to understanding the pathogenicity of C. sakazakii and provide insights into the development of targeted interventions and therapies against this bacterium.IMPORTANCEThe emergence of antibiotic resistance in pathogenic bacteria has become a critical global health concern, necessitating the identification of virulence factors as potential targets for the development of new antibiotics. This study addresses the limitations of conventional approaches by employing a combination of genetic screening, comparative proteomics, and in vivo validation to rapidly identify specific genes and proteins associated with the virulence of Cronobacter sakazakii, a highly pathogenic bacterium responsible for severe infections in vulnerable populations. The identification of two genes, rcsA and treR, as potential regulators of C. sakazakii toxicity toward zebrafish and rats and the proteomic profiling upon knockout of rcsA and treR provides novel insights into the mechanisms underlying bacterial virulence. The findings contribute to our understanding of C. sakazakii's pathogenicity, shed light on the regulatory pathways involved in bacterial virulence, and offer potential targets for the development of novel interventions against this highly virulent bacterium.
Collapse
Affiliation(s)
- Xi Chen
- Tianjin Eye Hospital, Tianjin Eye Institute,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, China
| | - Juan Xue
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaoli Dong
- Tianjin Eye Hospital, Tianjin Eye Institute,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, China
| | - Ping Lu
- Tianjin Eye Hospital, Tianjin Eye Institute,Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, China
| |
Collapse
|
9
|
Ji F, Tian G, Shang D, Jiang F. Antimicrobial peptide 2K4L disrupts the membrane of multidrug-resistant Acinetobacter baumannii and protects mice against sepsis. Front Microbiol 2023; 14:1258469. [PMID: 37942076 PMCID: PMC10628664 DOI: 10.3389/fmicb.2023.1258469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Antimicrobial peptides represent a promising therapeutic alternative for the treatment of antibiotic-resistant bacterial infections. 2K4L is a rationally-designed analog of a short peptide temporin-1CEc, a natural peptide isolated and purified from the skin secretions of the Chinese brown frog Rana chensinensis by substituting amino acid residues. 2K4L adopt an α-helical confirm in a membrane-mimetic environment and displayed an improved and broad-spectrum antibacterial activity against sensitive and multidrug-resistant Gram-negative and Gram-positive bacterial strains. Here, the action mechanism of 2K4L on multidrug resistant Acinetobacter baumannii (MRAB) and protection on MRAB-infected mice was investigated. The results demonstrated high bactericidal activity of 2K4L against both a multidrug resistant A. baumannii 0227 strain (MRAB 0227) and a sensitive A. baumannii strain (AB 22934), indicating a potential therapeutic advantage of this peptide. Strong positively-charged residues significantly promoted the electrostatic interaction on 2K4L with lipopolysaccharides (LPS) of the bacterial outer membrane. High hydrophobicity and an α-helical confirm endowed 2K4L remarkably increase the permeability of A. baumannii cytoplasmic membrane by depolarization of membrane potential and disruption of membrane integration, as well as leakage of fluorescein from the liposomes. Additionally, 2K4L at low concentrations inhibited biofilm formation and degraded mature 1-day-old MRAB 0227 biofilms by reducing the expression of biofilm-related genes. In an invasive A. baumannii infection model, 2K4L enhanced the survival of sepsis mice and decreased the production of the proinflammatory cytokines downregulating the phosphorylation level of signaling protein in MAPK and NF-κB signaling pathways, indicating that 2K4L represents a novel therapeutic antibiotic candidate against invasive multidrug-resistant bacterial strain infections.
Collapse
Affiliation(s)
- Fangyu Ji
- School of Life Science, Liaoning Normal University, Dalian, China
| | - Guoxu Tian
- School of Life Science, Liaoning Normal University, Dalian, China
| | - Dejing Shang
- School of Life Science, Liaoning Normal University, Dalian, China
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Fengquan Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Cavallo I, Oliva A, Pages R, Sivori F, Truglio M, Fabrizio G, Pasqua M, Pimpinelli F, Di Domenico EG. Acinetobacter baumannii in the critically ill: complex infections get complicated. Front Microbiol 2023; 14:1196774. [PMID: 37425994 PMCID: PMC10325864 DOI: 10.3389/fmicb.2023.1196774] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Acinetobacter baumannii is increasingly associated with various epidemics, representing a serious concern due to the broad level of antimicrobial resistance and clinical manifestations. During the last decades, A. baumannii has emerged as a major pathogen in vulnerable and critically ill patients. Bacteremia, pneumonia, urinary tract, and skin and soft tissue infections are the most common presentations of A. baumannii, with attributable mortality rates approaching 35%. Carbapenems have been considered the first choice to treat A. baumannii infections. However, due to the widespread prevalence of carbapenem-resistant A. baumannii (CRAB), colistin represents the main therapeutic option, while the role of the new siderophore cephalosporin cefiderocol still needs to be ascertained. Furthermore, high clinical failure rates have been reported for colistin monotherapy when used to treat CRAB infections. Thus, the most effective antibiotic combination remains disputed. In addition to its ability to develop antibiotic resistance, A. baumannii is also known to form biofilm on medical devices, including central venous catheters or endotracheal tubes. Thus, the worrisome spread of biofilm-producing strains in multidrug-resistant populations of A. baumannii poses a significant treatment challenge. This review provides an updated account of antimicrobial resistance patterns and biofilm-mediated tolerance in A. baumannii infections with a special focus on fragile and critically ill patients.
Collapse
Affiliation(s)
- Ilaria Cavallo
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Rebecca Pages
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Francesca Sivori
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Mauro Truglio
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Giorgia Fabrizio
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Martina Pasqua
- Department of Biology and Biotechnology "C. Darwin" Sapienza University of Rome, Rome, Italy
| | - Fulvia Pimpinelli
- Microbiology and Virology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Enea Gino Di Domenico
- Department of Biology and Biotechnology "C. Darwin" Sapienza University of Rome, Rome, Italy
| |
Collapse
|
11
|
Berre ML, Paulovčáková T, Verissimo CDM, Doyle S, Dalton JP, Masterson C, Martínez ER, Walsh L, Gormley C, Laffey JG, McNicholas B, Simpkin AJ, Kilcoyne M. A new multiplex SARS-CoV-2 antigen microarray showed correlation of IgG, IgA, and IgM antibodies from patients with COVID-19 disease severity and maintenance of relative IgA and IgM antigen binding over time. PLoS One 2023; 18:e0283537. [PMID: 36996259 PMCID: PMC10062637 DOI: 10.1371/journal.pone.0283537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 03/12/2023] [Indexed: 04/01/2023] Open
Abstract
Zoonotic spillover of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to humans in December 2019 caused the coronavirus disease 2019 (COVID-19) pandemic. Serological monitoring is critical for detailed understanding of individual immune responses to infection and protection to guide clinical therapeutic and vaccine strategies. We developed a high throughput multiplexed SARS-CoV-2 antigen microarray incorporating spike (S) and nucleocapsid protein (NP) and fragments expressed in various hosts which allowed simultaneous assessment of serum IgG, IgA, and IgM responses. Antigen glycosylation influenced antibody binding, with S glycosylation generally increasing and NP glycosylation decreasing binding. Purified antibody isotypes demonstrated a binding pattern and intensity different from the same isotype in whole serum, probably due to competition from the other isotypes present. Using purified antibody isotypes from naïve Irish COVID-19 patients, we correlated antibody isotype binding to different panels of antigens with disease severity, with binding to the S region S1 expressed in insect cells (S1 Sf21) significant for IgG, IgA, and IgM. Assessing longitudinal response for constant concentrations of purified antibody isotypes for a patient subset demonstrated that the relative proportion of antigen-specific IgGs decreased over time for severe disease, but the relative proportion of antigen-specific IgA binding remained at the same magnitude at 5 and 9 months post-first symptom onset. Further, the relative proportion of IgM binding decreased for S antigens but remained the same for NP antigens. This may support antigen-specific serum IgA and IgM playing a role in maintaining longer-term protection, important for developing and assessing vaccine strategies. Overall, these data demonstrate the multiplexed platform as a sensitive and useful platform for expanded humoral immunity studies, allowing detailed elucidation of antibody isotypes response against multiple antigens. This approach will be useful for monoclonal antibody therapeutic studies and screening of donor polyclonal antibodies for patient infusions.
Collapse
Affiliation(s)
- Marie Le Berre
- Carbohydrate Signalling Group, Infectious Disease Section, School of Chemical and Biological Sciences, University of Galway, Galway, Ireland
| | - Terézia Paulovčáková
- Carbohydrate Signalling Group, Infectious Disease Section, School of Chemical and Biological Sciences, University of Galway, Galway, Ireland
| | - Carolina De Marco Verissimo
- Molecular Parasitology Lab, Centre for One Health and Ryan Institute, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Seán Doyle
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - John P. Dalton
- Molecular Parasitology Lab, Centre for One Health and Ryan Institute, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Claire Masterson
- School of Medicine, and Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - Eduardo Ribes Martínez
- Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Laura Walsh
- University College Dublin, Belfield, Dublin, Ireland
| | - Conor Gormley
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - John G. Laffey
- School of Medicine, and Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Saolta University Hospital Group, Galway, Ireland
| | - Bairbre McNicholas
- School of Medicine, and Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
- Department of Anaesthesia and Intensive Care Medicine, University Hospital Galway, Saolta University Hospital Group, Galway, Ireland
| | - Andrew J. Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
| | - Michelle Kilcoyne
- Carbohydrate Signalling Group, Infectious Disease Section, School of Chemical and Biological Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
12
|
Remodeling of the Enterococcal Cell Envelope during Surface Penetration Promotes Intrinsic Resistance to Stress. mBio 2022; 13:e0229422. [PMID: 36354750 PMCID: PMC9765498 DOI: 10.1128/mbio.02294-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Enterococcus faecalis is a normal commensal of the human gastrointestinal tract (GIT). However, upon disruption of gut homeostasis, this nonmotile bacterium can egress from its natural niche and spread to distal organs. While this translocation process can lead to life-threatening systemic infections, the underlying mechanisms remain largely unexplored. Our prior work showed that E. faecalis migration across diverse surfaces requires the formation of matrix-covered multicellular aggregates and the synthesis of exopolysaccharides, but how enterococcal cells are reprogrammed during this process is unknown. Whether surface penetration endows E. faecalis with adaptive advantages is also uncertain. Here, we report that surface penetration promotes the generation of a metabolically and phenotypically distinct E. faecalis population with an enhanced capacity to endure various forms of extracellular stress. Surface-invading enterococci demonstrated major ultrastructural alterations in their cell envelope characterized by increased membrane glycolipid content. These changes were accompanied by marked induction of specific transcriptional programs enhancing cell envelope biogenesis and glycolipid metabolism. Notably, the surface-invading population demonstrated superior tolerance to membrane-damaging antimicrobials, including daptomycin and β-defensins produced by epithelial cells. Genetic mutations impairing glycolipid biosynthesis sensitized E. faecalis to envelope stressors and reduced the ability of this bacterium to penetrate semisolid surfaces and translocate through human intestinal epithelial cell monolayers. Our study reveals that surface penetration induces distinct transcriptional, metabolic, and ultrastructural changes that equip E. faecalis with enhanced capacity to resist external stressors and thrive in its surrounding environment. IMPORTANCE Enterococcus faecalis inhabits the GIT of multiple organisms, where its establishment could be mediated by the formation of biofilm-like aggregates. In susceptible individuals, this bacterium can overgrow and breach intestinal barriers, a process that may lead to lethal systemic infections. While the formation of multicellular aggregates promotes E. faecalis migration across surfaces, little is known about the metabolic and physiological states of the enterococci encased in these surface-penetrating structures. The present study reveals that E. faecalis cells capable of migrating through semisolid surfaces genetically reprogram their metabolism toward increased cell envelope and glycolipid biogenesis, which confers superior tolerance to membrane-damaging agents. E. faecalis's success as a pathobiont depends on its antimicrobial resistance, as well as on its rapid adaptability to overcome multiple environmental challenges. Thus, targeting adaptive genetic and/or metabolic pathways induced during E. faecalis surface penetration may be useful to better confront infections by this bacterium in the clinic.
Collapse
|
13
|
Kerantzas CA, Merwede J, Snyder EL, Hendrickson JE, Tormey CA, Kazmierczak BI, Peaper DR. Assessment of polymicrobial interactions in bacterial isolates from transfused platelet units associated with sepsis. Transfusion 2022; 62:2458-2463. [PMID: 36178430 PMCID: PMC11472026 DOI: 10.1111/trf.17136] [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: 12/31/2021] [Revised: 07/31/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND In 2019 the Centers for Disease Control and Prevention (CDC) reported a series of 4 transfusion reactions that resulted from contamination of apheresis platelet products. Products involved in all 4 cases were contaminated with Acinetobacter calcoaceticus-baumannii complex (ACBC) and in 3 products Staphylococcus saprophyticus was found as well. CDC investigation found that bacterial isolates from the cases were genetically related and suggested a common source of contamination. The contamination of blood products with ACBC is rare and polymicrobial contamination of blood products even less common. ACBC and S. saprophyticus have been observed to adhere to one another and sediment out of suspension in vitro, a process referred to as coaggregation, and we hypothesized that there was an interaction between the strains from these cases that contributed to their co-contamination of blood products. STUDY DESIGN AND METHODS To test the hypothesis of bacterial interaction, we performed coaggregation experiments and observed the growth characteristics of ACBC and S. saprophyticus strains recovered from contaminated blood products involved in a subset of the CDC cases. RESULTS An increase in S. saprophyticus CFU concentration was observed after several days of co-culture with ACBC in LB and plasma; however, no other findings suggested coaggregation or augmentative growth interaction between the bacterial strains. CONCLUSION Ultimately, an interaction between ACBC and S. saprophyticus that could help explain their co-occurrence and growth in contaminated platelet units was not found; however future studies of potential interactions may be warranted.
Collapse
Affiliation(s)
| | - Jacob Merwede
- Department of Laboratory Medicine, Yale-New Haven Hospital, New Haven, Connecticut, USA
| | - Edward L. Snyder
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, USA
| | | | | | - Barbara I. Kazmierczak
- Department of Internal Medicine, Section of Infectious Disease, Yale University, New Haven, Connecticut, USA
- Department of Microbial Pathogenesis, Yale University, New Haven, Connecticut, USA
| | - David R. Peaper
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Ramezanalizadeh F, Rasooli I, Owlia P, Darvish Alipour Astaneh S, Abdolhamidi R. Vaccination with a combination of planktonic and biofilm virulence factors confers protection against carbapenem-resistant Acinetobacter baumannii strains. Sci Rep 2022; 12:19909. [PMID: 36402866 PMCID: PMC9675771 DOI: 10.1038/s41598-022-24163-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022] Open
Abstract
Acinetobacter baumannii is a multi-drug resistant pathogen with the ability to switch between planktonic and biofilm phenotypes. Although there is no vaccine against A. baumannii infections, many attempts have been made to develop vaccines using planktonic or biofilm antigens. To cover the different phenotypes of A. baumannii during growth and attachment, we combined planktonic upregulated antigens of iron receptors with biofilm upregulated antigens of pilus rods and evaluated immune responses and protective efficacies of the combined vaccine using lethal and sub-lethal murine sepsis models. The results showed that the combined vaccine elicited high IgG antibody titers and conferred protection against lethal doses of two Carbapenem-resistant high adherent A. baumannii strains. Complete bacterial clearance from all the affected tissues of the mice challenged with A. baumannii was an excellent achievement with our quadrivalent immunogen. These results demonstrate both planktonic and biofilm antigens are important during antigen selection for vaccine design.
Collapse
Affiliation(s)
| | - Iraj Rasooli
- Department of Biology, Shahed University, Tehran, Iran.
- Department of Biology, Molecular Microbiology Research Center, Shahed University, Tehran-Qom Express Way, Tehran, 3319118651, Iran.
| | - Parviz Owlia
- Department of Biology, Molecular Microbiology Research Center, Shahed University, Tehran-Qom Express Way, Tehran, 3319118651, Iran
| | - Shakiba Darvish Alipour Astaneh
- Department of Biotechnology, Semnan University, Central Administration of Semnan University, Campus 1, P.O. Box 35131-19111, Semnan, Islamic Republic of Iran
| | | |
Collapse
|
15
|
Kasthuri T, Swetha TK, Bhaskar JP, Pandian SK. Rapid-killing efficacy substantiates the antiseptic property of the synergistic combination of carvacrol and nerol against nosocomial pathogens. Arch Microbiol 2022; 204:590. [PMID: 36053368 PMCID: PMC9438373 DOI: 10.1007/s00203-022-03197-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/08/2022] [Accepted: 08/18/2022] [Indexed: 12/05/2022]
Abstract
Globally, new classes of synthetic and natural antibiotics and antivirulents have continuously been validated for their potential broad-spectrum antagonistic activity with the aim of identifying an effective active molecule to prevent the spread of infectious agents in both food industry and medical field. In view of this, present study is aimed at evaluating the rapid killing efficacy of bioactive molecules Carvacrol (C) and Nerol (N) through British Standard European Norm 1276: phase2/step1 (EN1276) protocol. Active molecules C and N showed broad-spectrum antimicrobial activity against the test strains Staphylococcus aureus, Pseudomonas aeruginosa, Escherichia coli and Enterococcus hirae at concentration range of 78.125, 625, 156.25 and 312.5 μg/mL, respectively, for C, and 625 μg/mL for N. Whereas, combinatorial approach showed efficient activity with four times reduced concentration of C and N at 78.125 and 156.25 µg/mL, respectively, against test strains. Further, EN1276 results proved the rapid killing efficacy of test strains in 1 min of contact time with significant (> 5 log) growth reduction at 100X concentration of actives. SEM analysis and reduced concentration of protease, lipids and carbohydrate contents of treated group biofilm components ascertained preformed biofilm disruption potential of C + N on polystyrene and nail surfaces. C + N at synergistic concentration exhibited no adverse effect on HaCaT cells at 78.125 µg/mL (C) + 156.25 µg/mL (N). Taken together, based on the observed experimental results, present study evidence the antiseptic/disinfectant ability of C + N and suggest that the combination can preferentially be used in foam-based hand wash formulations.
Collapse
Affiliation(s)
- Thirupathi Kasthuri
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India
| | | | | | - Shunmugiah Karutha Pandian
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India.
| |
Collapse
|
16
|
Law SKK, Tan HS. The Role of Quorum Sensing, Biofilm Formation, and Iron Acquisition as Key Virulence Mechanisms in Acinetobacter baumannii and the Corresponding Anti-virulence Strategies. Microbiol Res 2022; 260:127032. [DOI: 10.1016/j.micres.2022.127032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022]
|
17
|
Upmanyu K, Haq QMR, Singh R. Factors mediating Acinetobacter baumannii biofilm formation: Opportunities for developing therapeutics. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100131. [PMID: 35909621 PMCID: PMC9325880 DOI: 10.1016/j.crmicr.2022.100131] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Acinetobacter baumannii has notably become a superbug due to its mounting risk of infection and escalating rates of antimicrobial resistance, including colistin, the last-resort antibiotic. Its propensity to form biofilm on biotic and abiotic surfaces has contributed to the majority of nosocomial infections. Bacterial cells in biofilms are resistant to antibiotics and host immune response, and pose challenges in treatment. Therefore current scenario urgently requires the development of novel therapeutic strategies for successful treatment outcomes. This article provides a holistic understanding of sequential events and regulatory mechanisms directing A. baumannii biofilm formation. Understanding the key factors functioning and regulating the biofilm machinery of A. baumannii will provide us insight to develop novel approaches to combat A. baumannii infections. Further, the review article deliberates promising strategies for the prevention of biofilm formation on medically relevant substances and potential therapeutic strategies for the eradication of preformed biofilms which can help tackle biofilm-associated A. baumannii infections. Advances in emerging therapeutic opportunities such as phage therapy, nanoparticle therapy and photodynamic therapy are also discussed to comprehend the current scenario and future outlook for the development of successful treatment against biofilm-associated A. baumannii infections.
Collapse
Affiliation(s)
- Kirti Upmanyu
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | | | - Ruchi Singh
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| |
Collapse
|
18
|
Roy S, Chowdhury G, Mukhopadhyay AK, Dutta S, Basu S. Convergence of Biofilm Formation and Antibiotic Resistance in Acinetobacter baumannii Infection. Front Med (Lausanne) 2022; 9:793615. [PMID: 35402433 PMCID: PMC8987773 DOI: 10.3389/fmed.2022.793615] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/31/2022] [Indexed: 07/30/2023] Open
Abstract
Acinetobacter baumannii (A. baumannii) is a leading cause of nosocomial infections as this pathogen has certain attributes that facilitate the subversion of natural defenses of the human body. A. baumannii acquires antibiotic resistance determinants easily and can thrive on both biotic and abiotic surfaces. Different resistance mechanisms or determinants, both transmissible and non-transmissible, have aided in this victory over antibiotics. In addition, the propensity to form biofilms (communities of organism attached to a surface) allows the organism to persist in hospitals on various medical surfaces (cardiac valves, artificial joints, catheters, endotracheal tubes, and ventilators) and also evade antibiotics simply by shielding the bacteria and increasing its ability to acquire foreign genetic material through lateral gene transfer. The biofilm formation rate in A. baumannii is higher than in other species. Recent research has shown how A. baumannii biofilm-forming capacity exerts its effect on resistance phenotypes, development of resistome, and dissemination of resistance genes within biofilms by conjugation or transformation, thereby making biofilm a hotspot for genetic exchange. Various genes control the formation of A. baumannii biofilms and a beneficial relationship between biofilm formation and "antimicrobial resistance" (AMR) exists in the organism. This review discusses these various attributes of the organism that act independently or synergistically to cause hospital infections. Evolution of AMR in A. baumannii, resistance mechanisms including both transmissible (hydrolyzing enzymes) and non-transmissible (efflux pumps and chromosomal mutations) are presented. Intrinsic factors [biofilm-associated protein, outer membrane protein A, chaperon-usher pilus, iron uptake mechanism, poly-β-(1, 6)-N-acetyl glucosamine, BfmS/BfmR two-component system, PER-1, quorum sensing] involved in biofilm production, extrinsic factors (surface property, growth temperature, growth medium) associated with the process, the impact of biofilms on high antimicrobial tolerance and regulation of the process, gene transfer within the biofilm, are elaborated. The infections associated with colonization of A. baumannii on medical devices are discussed. Each important device-related infection is dealt with and both adult and pediatric studies are separately mentioned. Furthermore, the strategies of preventing A. baumannii biofilms with antibiotic combinations, quorum sensing quenchers, natural products, efflux pump inhibitors, antimicrobial peptides, nanoparticles, and phage therapy are enumerated.
Collapse
Affiliation(s)
- Subhasree Roy
- Division of Bacteriology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Goutam Chowdhury
- Division of Molecular Microbiology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Asish K. Mukhopadhyay
- Division of Molecular Microbiology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shanta Dutta
- Division of Bacteriology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sulagna Basu
- Division of Bacteriology, Indian Council of Medical Research (ICMR)-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
19
|
Calculating Half Maximal Inhibitory Concentration (IC 50) Values from Glycomics Microarray Data Using GraphPad Prism. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2460:89-111. [PMID: 34972932 DOI: 10.1007/978-1-0716-2148-6_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Half maximal inhibitory concentration (IC50) is a measurement often used to compare the efficiency of various carbohydrates and their derivatives for inhibition of lectin binding to particular ligands. IC50 values can be calculated using experimental data from various platforms including enzyme-linked immunosorbent assay- (ELISA-)type microtiter plate assays, isothermal titration calorimetry (ITC), or glycan microarrays. In this chapter, we describe methods to fluorescently label a lectin, to carry out a lectin binding inhibition experiment on glycan microarrays, and to calculate the IC50 value of a binding inhibitory molecule using GraphPad Prism software. In the example used to illustrate the method in this chapter, IC50 calculation is demonstrated for inhibition of Maackia amurensis agglutinin (MAA) binding to 3'sialyl-N-acetyllactosamine (3SLN) using free lactose.
Collapse
|
20
|
Cunningham S, Gerlach JQ, Flannery A, Kilcoyne M. Bacterial Staining and Profiling for Glycan Interactions on Glycan Microarrays for t-Test Calculation. Methods Mol Biol 2022; 2460:223-237. [PMID: 34972940 DOI: 10.1007/978-1-0716-2148-6_14] [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] [Indexed: 06/14/2023]
Abstract
The use of glycan microarrays to study carbohydrate interactions of bacterial cells is of great interest owing to the key roles these interactions play in bacterial colonization and infection of a host. In this chapter, the methods to fluorescently stain Gram-positive or Gram-negative bacteria and profiling them for glycan interactions using glycan microarrays are described in detail. The application of the Student's t-test to glycan microarray data using an example data set comparing glycan microarray binding of an Acinetobacter baumannii wild type and mutant strain is also described in step-by-step detail.
Collapse
Affiliation(s)
- Stephen Cunningham
- Glycoscience Group, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
- Advanced Glycoscience Research Cluster, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Jared Q Gerlach
- Advanced Glycoscience Research Cluster, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Andrea Flannery
- Advanced Glycoscience Research Cluster, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
- Carbohydrate Signalling Group, Discipline of Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Michelle Kilcoyne
- Advanced Glycoscience Research Cluster, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
- Carbohydrate Signalling Group, Discipline of Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
21
|
Kumar S, Anwer R, Azzi A. Virulence Potential and Treatment Options of Multidrug-Resistant (MDR) Acinetobacter baumannii. Microorganisms 2021; 9:microorganisms9102104. [PMID: 34683425 PMCID: PMC8541637 DOI: 10.3390/microorganisms9102104] [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: 08/26/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Acinetobacter baumannii is an opportunistic pathogen which is undoubtedly known for a high rate of morbidity and mortality in hospital-acquired infections. A. baumannii causes life-threatening infections, including; ventilator-associated pneumonia (VAP), meningitis, bacteremia, and wound and urinary tract infections (UTI). In 2017, the World Health Organization listed A. baumannii as a priority-1 pathogen. The prevalence of A. baumannii infections and outbreaks emphasizes the direct need for the use of effective therapeutic agents for treating such infections. Available antimicrobials, such as; carbapenems, tigecycline, and colistins have insufficient effectiveness due to the appearance of multidrug-resistant strains, accentuating the need for alternative and novel therapeutic remedies. To understand and overcome this menace, the knowledge of recent discoveries on the virulence factors of A. baumannii is needed. Herein, we summarized the role of various virulence factors, including; outer membrane proteins, efflux pumps, biofilm, penicillin-binding proteins, and siderophores/iron acquisition systems. We reviewed the recent scientific literature on different A. baumannii virulence factors and the effective antimicrobial agents for the treatment and management of bacterial infections.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India;
| | - Razique Anwer
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317-4233, Saudi Arabia;
| | - Arezki Azzi
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317-4233, Saudi Arabia
- Correspondence:
| |
Collapse
|
22
|
Tram G, Poole J, Adams FG, Jennings MP, Eijkelkamp BA, Atack JM. The Acinetobacter baumannii Autotransporter Adhesin Ata Recognizes Host Glycans as High-Affinity Receptors. ACS Infect Dis 2021; 7:2352-2361. [PMID: 34339169 DOI: 10.1021/acsinfecdis.1c00021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acinetobacter baumannii is a significant opportunistic pathogen responsible for infections of the lung, blood, skin, urinary tract, and soft tissues, with some strains exhibiting almost complete resistance to commonly used antibiotics. This multidrug resistance, together with a dearth of new antibiotic development, mean novel methods of treatment and prevention are urgently needed. Although many A. baumannii factors required to colonize the host have been identified, little is known about the specific host molecules recognized by these factors. A. baumannii produces a trimeric autotransporter adhesin known as Ata that has been previously demonstrated to bind components of the host cell's extracellular matrix, which are often heavily glycosylated. We hypothesized that Ata would exhibit lectin activity which would play a role in adherence to the host cell surface. Our biophysical analysis using glycan arrays and surface plasmon resonance demonstrated that Ata binds galactose, N-acetylglucosamine, and galactose (β1-3/4) N-acetylglucosamine with high-affinity. These structures are present on many of the proteins which were previously reported to be bound by Ata. We also demonstrated that the recognition of human plasma fibronectin by Ata requires this ability to bind glycans, as the interaction between Ata and fibronectin does not occur when fibronectin is deglycosylated. This strongly suggests a key role for Ata lectin activity during host adherence. This information will assist in directing the development of new and effective treatments to block host interactions using glycans and/or novel compounds in multidrug resistant A. baumannii infections.
Collapse
Affiliation(s)
- Greg Tram
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4215, Australia
| | - Jessica Poole
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4215, Australia
| | - Felise G. Adams
- Molecular Sciences & Technology, College of Science and Engineering, Flinders University, Sturt Road, Bedford Park, South Australia 5042, Australia
| | - Michael P. Jennings
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4215, Australia
| | - Bart A. Eijkelkamp
- Molecular Sciences & Technology, College of Science and Engineering, Flinders University, Sturt Road, Bedford Park, South Australia 5042, Australia
| | - John M. Atack
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4215, Australia
| |
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: 6.8] [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
|
Pimenta AI, Kilcoyne M, Bernardes N, Mil-Homens D, Joshi L, Fialho AM. Burkholderia cenocepacia BCAM2418-induced antibody inhibits bacterial adhesion, confers protection to infection and enables identification of host glycans as adhesin targets. Cell Microbiol 2021; 23:e13340. [PMID: 33822465 DOI: 10.1111/cmi.13340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 12/18/2022]
Abstract
Trimeric Autotransporter Adhesins (TAA) found in Gram-negative bacteria play a key role in virulence. This is the case of Burkholderia cepacia complex (Bcc), a group of related bacteria able to cause infections in patients with cystic fibrosis. These bacteria use TAAs, among other virulence factors, to bind to host protein receptors and their carbohydrate ligands. Blocking such contacts is an attractive approach to inhibit Bcc infections. In this study, using an antibody produced against the TAA BCAM2418 from the epidemic strain Burkholderia cenocepacia K56-2, we were able to uncover its roles as an adhesin and the type of host glycan structures that serve as recognition targets. The neutralisation of BCAM2418 was found to cause a reduction in the adhesion of the bacteria to bronchial cells and mucins. Moreover, in vivo studies have shown that the anti-BCAM2418 antibody exerted an inhibitory effect during infection in Galleria mellonella. Finally, inferred by glycan arrays, we were able to predict for the first time, host glycan epitopes for a TAA. We show that BCAM2418 favoured binding to 3'sialyl-3-fucosyllactose, histo-blood group A, α-(1,2)-linked Fuc-containing structures, Lewis structures and GM1 gangliosides. In addition, the glycan microarrays demonstrated similar specificities of Burkholderia species for their most intensely binding carbohydrates.
Collapse
Affiliation(s)
- Andreia I Pimenta
- iBB-Institute for Bioengineering and Biosciences, and Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Michelle Kilcoyne
- Carbohydrate Signalling Group, Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Nuno Bernardes
- iBB-Institute for Bioengineering and Biosciences, and Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Dalila Mil-Homens
- iBB-Institute for Bioengineering and Biosciences, and Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Lokesh Joshi
- Glycoscience Group, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Arsenio M Fialho
- iBB-Institute for Bioengineering and Biosciences, and Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| |
Collapse
|