1
|
Hosseini SF, Jalali Nadoushan M, Fekrirad Z, Rasooli I. Omp34-Mediated Acinetobacter baumannii Invasion of Human Cervical Carcinoma Epithelial, HeLa Cells, and the Influence of Anti-Omp34 Antibodies. Anal Cell Pathol (Amst) 2025; 2025:1931119. [PMID: 40256153 PMCID: PMC12006715 DOI: 10.1155/ancp/1931119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/13/2025] [Indexed: 04/22/2025] Open
Abstract
Acinetobacter baumannii is known for its ability to invade and persist within eukaryotic cells, impacting infection outcomes and disease progression. This study investigates the role of Omp34, a key outer membrane protein (Omp), in A. baumannii interaction with epithelial cells and the protective effects of anti-Omp34 antibodies (Abs). Omp34 is a key regulator of A. baumannii epithelial cell invasion, influencing bacterial adherence, internalization, and intracellular proliferation. The presence of anti-Omp34 Abs mitigates A. baumannii-induced cellular damage and enhances bacterial clearance. The process involved the expression and purification of Omp34, which in turn induced Abs in BALB/c mice against Omp34. The acute toxicity of Omp34 was studied through a histological analysis conducted on six distinct organs in mice. HeLa cells were infected by A. baumannii ATCC 19606 and a clinical strain. Various aspects of A. baumannii behavior with HeLa cells, including HeLa cell viability, adherence, serum resistance, cell internalization, and intracellular proliferation with and without anti-Omp34 sera. Cytoskeleton inhibitors were used to study the potential roles played in the process of A. baumannii invasion by microfilaments and microtubules. Omp34 effectively triggered Ab production in mice without resulting in any toxicity. The assay for serum resistance revealed potent bactericidal and antibiofilm effects on both A. baumannii strains. Bacterial internalization was constrained when actin polymerization was inhibited. Examination under the microscope revealed instances of adherence, alterations in the cell membrane, apoptosis, vacuolization, and cell damage. HeLa cells exposed to anti-Omp34 serum showed decreased cell damage. The results provide substantial evidence of the adherence capacity of A. baumannii to proliferate in the epithelial cells. In conclusion, Omp34 plays a substantial role in regulating interactions between epithelial cells and A. baumannii, the multifaceted nature of which intricately modifies the trajectory of infection within host cells by A. baumannii.
Collapse
Affiliation(s)
| | - Mohammadreza Jalali Nadoushan
- Department of Pathology, School of Medicine, Shahed University, Tehran, Iran
- Molecular Microbiology Research Center and Department of Biology, Shahed University, Tehran, Iran
| | | | - Iraj Rasooli
- Department of Biology, Shahed University, Tehran, Iran
- Molecular Microbiology Research Center and Department of Biology, Shahed University, Tehran, Iran
| |
Collapse
|
2
|
Wei SY, Li YL, Wang L, Chu ZY, Qin YC, Zeng H. α-Pinene: Inhibitor of Acinetobacter baumannii biofilms and potential therapeutic agent for pneumonia. Int Immunopharmacol 2025; 151:114287. [PMID: 40015205 DOI: 10.1016/j.intimp.2025.114287] [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: 12/10/2024] [Revised: 02/08/2025] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Abstract
Acinetobacter baumannii is a Gram-negative bacterium whose biofilm formation and mechanisms contribute to its persistent infectivity and drug resistance in clinical settings. Inhibition or disruption of biofilms might hold the key to resolving the issue of drug resistance in A. baumannii. α-Pinene, a bicyclic terpene olefin derived from the essential oils of plants, exhibits multiple biological activities, including antimicrobial, antioxidant, and anti-inflammatory effects. In this investigation, we discovered that α-Pinene had powerful antimicrobial activity against A. baumannii 390015, and its minimum inhibitory concentration was 0.625 μL/mL. In vitro experiments demonstrated that α-Pinene exerted an inhibitory effect on biofilm formation and impacted the production of extracellular polymers and the twitching motility of A. baumannii. Moreover, qRT-PCR experiments in combination with proteomic validation revealed that bmfR, csuAB, ompA, and bap were down-regulated in A. baumannii after the action of α-Pinene. In vivo experiments indicated that α-Pinene decreased the expression of inflammatory factors, including interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α) in tissues. Additionally, the expression levels of JNK, P38, and ERK in the downstream pathways of TRAF6 were evaluated, and it was found that α-Pinene decreased the expression levels of JNK, P38, and ERK. Notably, the expression levels of these markers increased as the concentration of α-Pinene decreased. These findings suggest that α-Pinene can inhibit biofilm formation in A. baumannii and mitigate inflammation, highlighting its therapeutic potential for A. baumannii infections.
Collapse
Affiliation(s)
- Shu-Yun Wei
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise 533000, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Clinical Prevention and Control Technology and Leading Drug for Microorganisms with Drug Resistance in Border Ethnic Areasn, Baise 533000, China; Key Laboratory of the Prevention and Treatment of Drug-Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Education Department of Guangxi Zhuang Autonomous Region, Baise 533000, China
| | - Yu-Long Li
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise 533000, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Clinical Prevention and Control Technology and Leading Drug for Microorganisms with Drug Resistance in Border Ethnic Areasn, Baise 533000, China; Key Laboratory of the Prevention and Treatment of Drug-Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Education Department of Guangxi Zhuang Autonomous Region, Baise 533000, China
| | - Lin Wang
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise 533000, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Clinical Prevention and Control Technology and Leading Drug for Microorganisms with Drug Resistance in Border Ethnic Areasn, Baise 533000, China; Key Laboratory of the Prevention and Treatment of Drug-Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Education Department of Guangxi Zhuang Autonomous Region, Baise 533000, China
| | - Zi-Yong Chu
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise 533000, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Clinical Prevention and Control Technology and Leading Drug for Microorganisms with Drug Resistance in Border Ethnic Areasn, Baise 533000, China; Key Laboratory of the Prevention and Treatment of Drug-Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Education Department of Guangxi Zhuang Autonomous Region, Baise 533000, China; College of Life Science and Technology, Xinjiang University, Urumqi 830046, Xinjiang, China
| | - Yan-Chun Qin
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise 533000, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Clinical Prevention and Control Technology and Leading Drug for Microorganisms with Drug Resistance in Border Ethnic Areasn, Baise 533000, China; Key Laboratory of the Prevention and Treatment of Drug-Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Education Department of Guangxi Zhuang Autonomous Region, Baise 533000, China.
| | - Hong Zeng
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Guangxi Technology Innovation Cooperation Base of Prevention and Control Pathogenic Microbes with Drug Resistance, Youjiang Medical University for Nationalities, Baise 533000, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Clinical Prevention and Control Technology and Leading Drug for Microorganisms with Drug Resistance in Border Ethnic Areasn, Baise 533000, China; Key Laboratory of the Prevention and Treatment of Drug-Resistant Microbial Infecting, Youjiang Medical University for Nationalities, Education Department of Guangxi Zhuang Autonomous Region, Baise 533000, China.
| |
Collapse
|
3
|
Zhang R, Li D, Fang H, Xie Q, Tang H, Chen L. Iron-dependent mechanisms in Acinetobacter baumannii: pathogenicity and resistance. JAC Antimicrob Resist 2025; 7:dlaf039. [PMID: 40110557 PMCID: PMC11920509 DOI: 10.1093/jacamr/dlaf039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
Acinetobacter baumannii is a Gram-negative opportunistic pathogen that poses a significant challenge in healthcare settings, particularly in ICUs, due to its MDR and high mortality rates, especially among critically ill coronavirus disease 2019 patients. Iron is crucial for the survival, growth and pathogenicity of A. baumannii, and the bacterium has developed multiple iron acquisition systems, including siderophore production, haem uptake and TonB-dependent transport mechanisms, to adapt to the iron-limited environment within the host. Although specific studies on A. baumannii are limited, mechanisms from other bacterial species suggest that similar iron acquisition strategies may play a key role in its virulence. Therapeutic approaches targeting these iron-dependent systems, such as the siderophore-conjugated cephalosporin cefiderocol, have shown potential in overcoming MDR A. baumannii infections. Additionally, strategies such as synthetic siderophores, TonB receptor inhibitors and iron chelators are under investigation to enhance treatment outcomes. Future research should prioritize validating these mechanisms in A. baumannii, advancing clinical trials for these therapies and exploring combination treatments to mitigate resistance and improve clinical outcomes in severely affected patients.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Pulmonary and Critical Care Medicine, Southwest Medical University, Luzhou, China
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Li
- Department of Pulmonary and Critical Care Medicine, Southwest Medical University, Luzhou, China
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Fang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Xie
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Huan Tang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Chen
- Department of Pulmonary and Critical Care Medicine, Southwest Medical University, Luzhou, China
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Lucidi M, Visaggio D, Migliaccio A, Capecchi G, Visca P, Imperi F, Zarrilli R. Pathogenicity and virulence of Acinetobacter baumannii: Factors contributing to the fitness in healthcare settings and the infected host. Virulence 2024; 15:2289769. [PMID: 38054753 PMCID: PMC10732645 DOI: 10.1080/21505594.2023.2289769] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023] Open
Abstract
Acinetobacter baumannii is a common cause of healthcare-associated infections and hospital outbreaks, particularly in intensive care units. Much of the success of A. baumannii relies on its genomic plasticity, which allows rapid adaptation to adversity and stress. The capacity to acquire novel antibiotic resistance determinants and the tolerance to stresses encountered in the hospital environment promote A. baumannii spread among patients and long-term contamination of the healthcare setting. This review explores virulence factors and physiological traits contributing to A. baumannii infection and adaptation to the hospital environment. Several cell-associated and secreted virulence factors involved in A. baumannii biofilm formation, cell adhesion, invasion, and persistence in the host, as well as resistance to xeric stress imposed by the healthcare settings, are illustrated to give reasons for the success of A. baumannii as a hospital pathogen.
Collapse
Affiliation(s)
- Massimiliano Lucidi
- Department of Science, Roma Tre University, Rome, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | - Daniela Visaggio
- Department of Science, Roma Tre University, Rome, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- Santa Lucia Foundation IRCCS, Rome, Italy
| | | | | | - Paolo Visca
- Department of Science, Roma Tre University, Rome, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- Santa Lucia Foundation IRCCS, Rome, Italy
| | - Francesco Imperi
- Department of Science, Roma Tre University, Rome, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- Santa Lucia Foundation IRCCS, Rome, Italy
| | - Raffaele Zarrilli
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
McGillin M, Tokman JI, Hsu E, Alcaine SD. Assessment of resistance to colicinogenic synthetic phage antimicrobial system. Microbiol Spectr 2024; 12:e0079324. [PMID: 39405458 PMCID: PMC11537092 DOI: 10.1128/spectrum.00793-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/05/2024] [Indexed: 11/07/2024] Open
Abstract
This work presents a multi-hurdle approach that addresses antimicrobial resistance by minimizing the selective pressure of antimicrobials using a novel colicinogenic-phage system. We have created two synthetic T7 phages (T7-E1 and T7-M) by inserting the gene of colicin E1 (Cea) or colicin M (Cma) into the genome of the T7 phage, thereby adding an additional colicin-based hurdle to the T7 lytic cycle. The colicin-phages' efficacy in suppressing the outgrowth of a T7-resistant sub-population within a mixed culture of Escherichia coli was demonstrated using a challenge matrix design under planktonic and structured conditions. When T7-resistant cells were present at 1% of the total planktonic population, T7-E1 delayed the outgrowth. At 0.1% resistance, T7-M delayed resistant outgrowth, whereas T7-E1 suppressed the resistant sub-population. When T7-E1 and T7-M were combined into a triple-hurdle treatment, the T7-E1/T7-M cocktail completely suppressed a mixed planktonic population of 50% resistance cell concentrations. In structured environments, the colicin-phage treatments formed clear and confluent plaque-like zones of clearing in the mixed populations of 50% resistant cells with a lawn density of 1 × 106 CFU/mL. Reducing the lawn density to 1 × 105 CFU/mL diminished the multi-hurdle treatments' effectiveness, as demonstrated by localized zones of clearing within turbid bacterial lawns, highlighting the relationship between bacterial lawn density and phage effectiveness in structured environments. Fluctuation assays revealed persistence as the predominant mechanism for overcoming the treatments by T7-sensitive E. coli. Results indicate that T7-M treatment significantly reduces persister formation compared to WT-T7, while T7-E1 unexpectedly increases persister formation significantly. This suggests a complex relationship between antimicrobial stress and persister formation. IMPORTANCE Antimicrobial resistance (AMR) poses a significant challenge in treating bacterial infections. To address this, we present a multi-hurdle approach that combines the power of different antimicrobials to target resistance. We have weaponized the natural predator of Escherichia coli, the T7-phage, by engineering it to produce toxins called colicins, resulting in a colicin-phage antimicrobial. This multi-hurdled approach aims to decrease resistance risk because survival requires different tactics to overcome the phage and colicin activity, thus adding a hurdle in a bacterium's pathway to resistance. In cases of pre-existing resistance, the colicin effectively controlled the sub-population resistant to the phage. When investigating the emergence of resistance, we discovered that antimicrobial persistence was the predominant survival strategy. These findings reveal an essential slice of the AMR pie by emphasizing bacterial survival tactics that are not based on resistance genes. By expanding our AMR lens to include persistence, we can more effectively address treatment failure.
Collapse
Affiliation(s)
- Meghan McGillin
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Jeffrey I. Tokman
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Ella Hsu
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Samuel D. Alcaine
- Department of Food Science, Cornell University, Ithaca, New York, USA
| |
Collapse
|
6
|
Pumirat P, Santajit S, Tunyong W, Kong-Ngoen T, Tandhavanant S, Lohitthai S, Rungruengkitkun A, Chantratita N, Ampawong S, Reamtong O, Indrawattana N. Impact of AbaI mutation on virulence, biofilm development, and antibiotic susceptibility in Acinetobacter baumannii. Sci Rep 2024; 14:21521. [PMID: 39277662 PMCID: PMC11401864 DOI: 10.1038/s41598-024-72740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024] Open
Abstract
The quorum sensing (QS) system mediated by the abaI gene in Acinetobacter baumannii is crucial for various physiological and pathogenic processes. In this study, we constructed a stable markerless abaI knockout mutant (ΔabaI) strain using a pEXKm5-based allele replacement method to investigate the impact of abaI on A. baumannii. Proteomic analysis revealed significant alterations in protein expression between the wild type (WT) and ΔabaI mutant strains, particularly in proteins associated with membrane structure, antibiotic resistance, and virulence. Notably, the downregulation of key outer membrane proteins such as SurA, OmpA, OmpW, and BamA suggests potential vulnerabilities in outer membrane integrity, which correlate with structural abnormalities in the ΔabaI mutant strain, including irregular cell shapes and compromised membrane integrity, observed by scanning and transmission electron microscopy. Furthermore, diminished expression of regulatory proteins such as OmpR and GacA-GacS highlights the broader regulatory networks affected by abaI deletion. Functional assays revealed impaired biofilm formation and surface-associated motility in the mutant strain, indicative of altered colonization capabilities. Interestingly, the mutant showed a complex antibiotic susceptibility profile. While it demonstrated increased susceptibility to membrane-targeting antibiotics, its response to beta-lactams was more nuanced. Despite increased expression of metallo-beta-lactamase (MBL) superfamily proteins and DcaP-like protein, the mutant unexpectedly showed lower MICs for carbapenems (imipenem and meropenem) compared to the wild-type strain. This suggests that abaI deletion affects antibiotic susceptibility through multiple, potentially competing mechanisms. Further investigation is needed to fully elucidate the interplay between quorum sensing, antibiotic resistance genes, and overall antibiotic susceptibility in A. baumannii. Our findings underscore the multifaceted role of the abaI gene in modulating various cellular processes and highlight its significance in A. baumannii physiology, pathogenesis, and antibiotic resistance. Targeting the abaI QS system may offer novel therapeutic strategies for this clinically significant pathogen.
Collapse
Affiliation(s)
- Pornpan Pumirat
- Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Sirijan Santajit
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Witawat Tunyong
- Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Thida Kong-Ngoen
- Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Sarunporn Tandhavanant
- Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
- Department of Bacteriology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Sanisa Lohitthai
- Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | | | - Narisara Chantratita
- Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Tropical Molecular Biology and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nitaya Indrawattana
- Biomedical Research Incubator Unit, Department of Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Department of Research, Siriraj Center of Research Excellence in Allergy and Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
7
|
Pereira IL, Cardoso TL, Wozeak DR, Caballero PS, Buchhorn de Freitas S, Pinto Seixas Neto AC, da Silva Pinto L, Hartwig DD. Antibodies anti-rFilF protein has anti-biofilm activity against carbapenem-resistant Acinetobacter baumannii. Microbes Infect 2024; 26:105347. [PMID: 38679227 DOI: 10.1016/j.micinf.2024.105347] [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: 09/26/2023] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
Acinetobacter baumannii is an opportunistic bacterium that causes infection in several sites. Carbapenem-resistant A. baumannii strains (CRAb) lead the World Health Organization's list of 12 pathogens considered a priority for developing new antimicrobials. The pathogenicity of A. baumannii is related to the different virulence factors employed in the colonization of biotic and abiotic surfaces, biofilm formation and multidrug resistance. We analyze the outer membrane protein FilF from A. baumannii in silico and produce it in recombinant form (rFilF). rFilF protein was successfully expressed in Escherichia coli BL21 Star in an insoluble form. Immunization with rFilF induced significant anti-rFilF IgG antibody production in mice, detected by indirect enzyme-linked immunosorbent assay, since the first evaluation until 49th. On the last experimentation day, the predominant immunoglobulin found was IgG1 followed by IgG2a, IgG2b, IgM, IgG3, and IgA. We observe that interleukins 4 and 10 show significant production after the 28th day of experimentation in mice immunized with rFilF. Anti-rFilF pAbs were able to inhibit biofilm formation in nine CRAb strains evaluated, and in the standard strain ATCC® 19606. These results demonstrate the anti-biofilm activity of anti-rFilF antibodies, promising in the development of a non-antibiotic approach based on the control of CRAb strains.
Collapse
Affiliation(s)
- Isabel Ladeira Pereira
- Biotechnology Unit, Center of Technological Development, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil
| | - Thayná Laner Cardoso
- Biotechnology Unit, Center of Technological Development, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil
| | - Daniela Rodriguero Wozeak
- Biotechnology Unit, Center of Technological Development, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil
| | - Pamela Scaraffuni Caballero
- Biotechnology Unit, Center of Technological Development, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil
| | - Stella Buchhorn de Freitas
- Biotechnology Unit, Center of Technological Development, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil
| | - Amilton Clair Pinto Seixas Neto
- Department of Microbiology and Parasitology, Biology Institute, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil
| | - Luciano da Silva Pinto
- Biotechnology Unit, Center of Technological Development, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil
| | - Daiane Drawanz Hartwig
- Biotechnology Unit, Center of Technological Development, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil; Department of Microbiology and Parasitology, Biology Institute, Federal University of Pelotas, CEP 96010-900, Pelotas, RS, Brazil.
| |
Collapse
|
8
|
Singh S, Singh S, Trivedi M, Dwivedi M. An insight into MDR Acinetobacter baumannii infection and its pathogenesis: Potential therapeutic targets and challenges. Microb Pathog 2024; 192:106674. [PMID: 38714263 DOI: 10.1016/j.micpath.2024.106674] [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: 10/19/2023] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024]
Abstract
Acinetobacter baumannii is observed as a common species of Gram-negative bacteria that exist in soil and water. Despite being accepted as a typical component of human skin flora, it has become an important opportunistic pathogen, especially in healthcare settings. The pathogenicity of A. baumannii is attributed to its virulence factors, which include adhesins, pili, lipopolysaccharides, outer membrane proteins, iron uptake systems, autotransporter, secretion systems, phospholipases etc. These elements provide the bacterium the ability to cling to and penetrate host cells, get past the host immune system, and destroy tissue. Its infection is a major contributor to human pathophysiological conditions including pneumonia, bloodstream infections, urinary tract infections, and surgical site infections. It is challenging to treat infections brought on by this pathogen since this bacterium has evolved to withstand numerous drugs and further emergence of drug-resistant A. baumannii results in higher rates of morbidity and mortality. The long-term survival of this bacterium on surfaces of medical supplies and hospital furniture facilitates its frequent spread in humans from one habitat to another. There is a need for urgent investigations to find effective drug targets for A. baumannii as well as designing novel drugs to reduce the survival and spread of infection. In the current review, we represent the specific features, pathogenesis, and molecular intricacies of crucial drug targets of A. baumannii. This would also assist in proposing strategies and alternative therapies for the prevention and treatment of A. baumannii infections and their spread.
Collapse
Affiliation(s)
- Sukriti Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India
| | - Sushmita Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India
| | - Mala Trivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India; Research Cell, Amity University Uttar Pradesh, Lucknow, 226028, India.
| |
Collapse
|
9
|
Brychcy M, Nguyen B, Tierney GA, Casula P, Kokodynski A, Godoy VG. The metabolite vanillic acid regulates Acinetobacter baumannii surface attachment. Mol Microbiol 2024; 121:833-849. [PMID: 38308563 DOI: 10.1111/mmi.15234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/05/2024]
Abstract
The nosocomial bacterium Acinetobacter baumannii is protected from antibiotic treatment by acquiring antibiotic resistances and by forming biofilms. Cell attachment, one of the first steps in biofilm formation, is normally induced by environmental metabolites. We hypothesized that vanillic acid (VA), the oxidized form of vanillin and a widely available metabolite, may play a role in A. baumannii cell attachment. We first discovered that A. baumannii actively breaks down VA through the evolutionarily conserved vanABKP genes. These genes are under the control of the repressor VanR, which we show binds directly to VanR binding sites within the vanABKP genes bidirectional promoter. VA in turn counteracts VanR inhibition. We identified a VanR binding site and searched for it throughout the genome, especially in pili encoding promoter genes. We found a VanR binding site in the pilus encoding csu operon promoter and showed that VanR binds specifically to it. As expected, a strain lacking VanR overproduces Csu pili and makes robust biofilms. Our study uncovers the role that VA plays in facilitating the attachment of A. baumannii cells to surfaces, a crucial step in biofilm formation. These findings provide valuable insights into a previously obscure catabolic pathway with significant clinical implications.
Collapse
Affiliation(s)
- Merlin Brychcy
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Brian Nguyen
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | | | - Pranav Casula
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Alexis Kokodynski
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Veronica G Godoy
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
10
|
de Oliveira PAA, Baboghlian J, Ramos COA, Mançano ASF, Porcari ADM, Girardello R, Ferraz LFC. Selection and validation of reference genes suitable for gene expression analysis by Reverse Transcription Quantitative real-time PCR in Acinetobacter baumannii. Sci Rep 2024; 14:3830. [PMID: 38360762 PMCID: PMC10869792 DOI: 10.1038/s41598-024-51499-5] [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: 07/18/2023] [Accepted: 01/05/2024] [Indexed: 02/17/2024] Open
Abstract
Acinetobacter baumannii is a Gram-negative bacterium considered an emerging multi-drug-resistant pathogen. Furthermore, this bacterium can survive in extreme environmental conditions, which makes it a frequent cause of nosocomial infection outbreaks. Gene expression analyses by Reverse Transcription Quantitative real-time PCR (RT-qPCR) depend on a reference gene, also called an endogenous gene, which is used to normalize the generated data and thus ensure an accurate analysis with minimal errors. Currently, gene expression analyses in A. baumannii are compromised, as there are no reports in the literature describing the identification of validated reference genes for use in RT-qPCR analyses. For this reason, we selected twelve candidate reference genes of A. baumannii and assessed their expression profile under different experimental and culture conditions. The expression stability of the candidate genes was evaluated by using statistical algorithms such as BestKeeper, geNorm, NormFinder, Delta CT, and RefFinder, in order to identify the most suitable candidate reference genes for RT-qPCR analyses. The statistical analyses indicated rpoB, rpoD, and fabD genes as the most adequate to ensure accurate normalization of RT-qPCR data in A. baumannii. The accuracy of the proposed reference genes was validated by using them to normalize the expression of the ompA gene, encoding the outer membrane protein A, in A. baumannii sensible and resistant to the antibiotic polymyxin. The present work provides suitable reference genes for precise RT-qPCR data normalization on future gene expression studies with A. baumannii.
Collapse
Affiliation(s)
| | - Juliana Baboghlian
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, SP, CEP 12916-900, Brazil
| | | | | | - Andréia de Melo Porcari
- Laboratório Multidisciplinar de Pesquisa, Universidade São Francisco, Bragança Paulista, SP, CEP 12916-900, Brazil
| | - Raquel Girardello
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, SP, CEP 12916-900, Brazil
| | - Lúcio Fábio Caldas Ferraz
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, SP, CEP 12916-900, Brazil.
| |
Collapse
|
11
|
Ching C, Brychcy M, Nguyen B, Muller P, Pearson AR, Downs M, Regan S, Isley B, Fowle W, Chai Y, Godoy VG. RecA levels modulate biofilm development in Acinetobacter baumannii. Mol Microbiol 2024; 121:196-212. [PMID: 37918886 DOI: 10.1111/mmi.15188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Infections caused by Acinetobacter baumannii, a Gram-negative opportunistic pathogen, are difficult to eradicate due to the bacterium's propensity to quickly gain antibiotic resistances and form biofilms, a protective bacterial multicellular community. The A. baumannii DNA damage response (DDR) mediates the antibiotic resistance acquisition and regulates RecA in an atypical fashion; both RecALow and RecAHigh cell types are formed in response to DNA damage. The findings of this study demonstrate that the levels of RecA can influence formation and dispersal of biofilms. RecA loss results in surface attachment and prominent biofilms, while elevated RecA leads to diminished attachment and dispersal. These findings suggest that the challenge to treat A. baumannii infections may be explained by the induction of the DDR, common during infection, as well as the delicate balance between maintaining biofilms in low RecA cells and promoting mutagenesis and dispersal in high RecA cells. This study underscores the importance of understanding the fundamental biology of bacteria to develop more effective treatments for infections.
Collapse
Affiliation(s)
- Carly Ching
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Merlin Brychcy
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Brian Nguyen
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Paul Muller
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | | | - Margaret Downs
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Samuel Regan
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Breanna Isley
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - William Fowle
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Yunrong Chai
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Veronica G Godoy
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Suresh G, Srivastava S. A concise review on genes involved in biofilm-related disease and differential gene expression in medical-related biofilms. MICROBIAL BIOFILMS 2024:215-235. [DOI: 10.1016/b978-0-443-19252-4.00012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Kasperski T, Romaniszyn D, Jachowicz-Matczak E, Pomorska-Wesołowska M, Wójkowska-Mach J, Chmielarczyk A. Extensive Drug Resistance of Strong Biofilm-Producing Acinetobacter baumannii Strains Isolated from Infections and Colonization Hospitalized Patients in Southern Poland. Pathogens 2023; 12:975. [PMID: 37623935 PMCID: PMC10459043 DOI: 10.3390/pathogens12080975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Acinetobacter baumannii (AB) is a bacterium that causes infections, particularly in immunocompromised patients. Treatment is challenging due to biofilm formation by AB strains, which hinders antibiotic effectiveness and promotes drug resistance. The aim of our study was to analyze the biofilm-producing capacity of AB isolates from various forms of infections in relation to biofilm-related genes and their drug resistance. We tested one hundred isolates for biofilm formation using the crystal violet microplate method. Drug resistance analyses were performed based on EUCAST and CLSI guidelines, and biofilm genes were detected using PCR. All tested strains were found to form biofilms, with 50% being ICU strains and 72% classified as strong biofilm producers. Among these, 87% were extensively drug-resistant (XDR) and 2% were extra-extensively drug-resistant (E-XDR). The most common gene set was bap, bfmS, csuE, and ompA, found in 57% of all isolates. Our research shows that, regardless of the form of infection, biofilm-forming strains can be expected among AB isolates. The emergence of E-XDR and XDR strains among non-ICU infections highlights the necessity for the rational use of antibiotics to stop or limit the further acquisition of drug resistance by A. baumannii.
Collapse
Affiliation(s)
- Tomasz Kasperski
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 31-008 Krakow, Poland
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland
| | - Dorota Romaniszyn
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland
| | - Estera Jachowicz-Matczak
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland
| | - Monika Pomorska-Wesołowska
- Department of Microbiology, Analytical and Microbiological Laboratory of Ruda Slaska, KORLAB NZOZ, 41-703 Ruda Slaska, Poland
| | - Jadwiga Wójkowska-Mach
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland
| | - Agnieszka Chmielarczyk
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland
| |
Collapse
|
14
|
Hajiagha MN, Kafil HS. Efflux pumps and microbial biofilm formation. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023:105459. [PMID: 37271271 DOI: 10.1016/j.meegid.2023.105459] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/06/2023]
Abstract
Biofilm-related infections are resistant forms of pathogens that are regarded as a medical problem, particularly due to the spread of multiple drug resistance. One of the factors associated with biofilm drug resistance is the presence of various types of efflux pumps in bacteria. Efflux pumps also play a role in biofilm formation by influencing Physical-chemical interactions, mobility, gene regulation, quorum sensing (QS), extracellular polymeric substances (EPS), and toxic compound extrusion. According to the findings of studies based on efflux pump expression analysis, their role in the anatomical position within the biofilm will differ depending on the biofilm formation stage, encoding gene expression level, the type and concentration of substrate. In some cases, the function of the efflux pumps can overlap with each other, so it seems necessary to accurate identify the efflux pumps of biofilm-forming bacteria along with their function in this process. Such studies will help to choose treatment strategy, at least in combination with antibiotics. Furthermore, if the goal of treatment is an efflux pump manipulation, we should not limit it to inhibition.
Collapse
Affiliation(s)
- Mahdyeh Neghabi Hajiagha
- Department of Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Pourhajibagher M, Hosseini N, Bahador A. Antimicrobial activity of D-amino acid in combination with photo-sonoactivated hypericin nanoparticles against Acinetobacter baumannii. BMC Microbiol 2023; 23:23. [PMID: 36658487 PMCID: PMC9850556 DOI: 10.1186/s12866-023-02758-4] [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: 09/21/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The emergence of multidrug-resistant Acinetobacter baumannii strains is increasing worldwide. To overcome these life-threatening infections, the development of new treatment approaches is critical. For this purpose, this study was conducted to determine the antimicrobial photo-sonodynamic therapy (aPSDT) using hypericin nanoparticles (HypNP) in combination with D-Tryptophan (D-Trp) against A. baumannii. MATERIALS AND METHODS HypNP was synthesized and characterized, followed by the determination of the fractional inhibitory concentration (FIC) index of HypNP and D-Trp by checkerboard assay. Next, the antimicrobial and anti-biofilm potential of HypNP@D-Trp-mediated aPSDT against A. baumannii was evaluated. Finally, the anti-virulence activity of aPSDT using HypNP@D-Trp was accessed following the characterization of HypNP@D-Trp interaction with AbaI using in silico virtual screening and molecular docking. RESULTS A synergistic activity in the combination of HypNP and D-Trp against A. baumannii was observed with a FIC index value of 0.5. There was a 5.10 log10 CFU/mL reduction in the cell viability of A. baumannii when the bacterial cells were treated with 1/2 × MIC of HypNP@D-Trp and subsequently exposed to ultrasound waves and blue light (P < 0.05). Moreover, a significant biofilm degradation effect on biofilm-associated cells of A. baumannii was observed after treatment with aPSDT using 2 × MIC of HypNP@D-Trp in comparison with the control groups (P < 0.05). According to the molecular docking analysis of the protein-ligand complex, Hyp with a high affinity for AbaI showed a binding affinity of - 9.41 kcal/mol. Also, the expression level of abaI gene was significantly downregulated by 10.32-fold in A. baumannii treated with aPSDT as comprised with the control group (P < 0.05). CONCLUSIONS It can be concluded that HypNP@D-Trp-mediated aPSDT can be considered a promising strategy to overcome the infections caused by A. baumannii by reducing the growth of bacterial biofilm and decreasing the expression of abaI as a gene involved in A. baumannii biofilm formation.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- grid.411705.60000 0001 0166 0922Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nava Hosseini
- grid.23856.3a0000 0004 1936 8390Institut de Biologie Intégrative et des Systèmes (IBIS), Pavillon Charles-Eugène-Marchand, Université Laval, Quebec City, QC G1V 0A6 Canada ,grid.23856.3a0000 0004 1936 8390Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Quebec City, QC G1V 0A6 Canada ,grid.421142.00000 0000 8521 1798Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5 Canada
| | - Abbas Bahador
- Fellowship in Clinical Laboratory Sciences, BioHealth Lab, Tehran, Iran ,grid.411705.60000 0001 0166 0922Department of Microbiology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Sharma S, Kaushik V, Kulshrestha M, Tiwari V. Different Efflux Pump Systems in Acinetobacter baumannii and Their Role in Multidrug Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023. [DOI: 10.1007/5584_2023_771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
17
|
Patel KD, Ahmed SF, MacDonald MR, Gulick AM. Structural Studies of Modular Nonribosomal Peptide Synthetases. Methods Mol Biol 2023; 2670:17-46. [PMID: 37184698 DOI: 10.1007/978-1-0716-3214-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The non-ribosomal peptide synthetases (NRPSs) are a family of modular enzymes involved in the production of peptide natural products. Not restricted by the constraints of ribosomal peptide and protein production, the NRPSs are able to incorporate unusual amino acids and other suitable building blocks into the final product. The NRPSs operate with an assembly line strategy in which peptide intermediates are covalently tethered to a peptidyl carrier protein and transported to different catalytic domains for the multiple steps in the biosynthesis. Often the carrier and catalytic domains are joined into a single large multidomain protein. This chapter serves to introduce the NRPS enzymes, using the nocardicin NRPS system as an example that highlights many common features to NRPS biochemistry. We then describe recent advances in the structural biology of NRPSs focusing on large multidomain structures that have been determined.
Collapse
Affiliation(s)
- Ketan D Patel
- Department of Structural Biology, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Syed Fardin Ahmed
- Department of Structural Biology, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Monica R MacDonald
- Department of Structural Biology, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Andrew M Gulick
- Department of Structural Biology, University at Buffalo, SUNY, Buffalo, NY, USA.
- Department of Structural Biology, Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY, USA.
| |
Collapse
|
18
|
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
|
19
|
Tomlinson BR, Denham GA, Torres NJ, Brzozowski RS, Allen JL, Jackson JK, Eswara PJ, Shaw LN. Assessing the Role of Cold-Shock Protein C: a Novel Regulator of Acinetobacter baumannii Biofilm Formation and Virulence. Infect Immun 2022; 90:e0037622. [PMID: 36121221 PMCID: PMC9584223 DOI: 10.1128/iai.00376-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022] Open
Abstract
Acinetobacter baumannii is a formidable opportunistic pathogen that is notoriously difficult to eradicate from hospital settings. This resilience is often attributed to a proclivity for biofilm formation, which facilitates a higher tolerance toward external stress, desiccation, and antimicrobials. Despite this, little is known regarding the mechanisms orchestrating A. baumannii biofilm formation. Here, we performed RNA sequencing (RNA-seq) on biofilm and planktonic populations for the multidrug-resistant isolate AB5075 and identified 438 genes with altered expression. To assess the potential role of genes upregulated within biofilms, we tested the biofilm-forming capacity of their respective mutants from an A. baumannii transposon library. In so doing, we uncovered 24 genes whose disruption led to reduced biofilm formation. One such element, cold shock protein C (cspC), had a highly mucoid colony phenotype, enhanced tolerance to polysaccharide degradation, altered antibiotic tolerance, and diminished adherence to abiotic surfaces. RNA-seq of the cspC mutant revealed 201 genes with altered expression, including the downregulation of pili and fimbria genes and the upregulation of multidrug efflux pumps. Using transcriptional arrest assays, it appears that CspC mediates its effects, at least in part, through RNA chaperone activity, influencing the half-life of several important transcripts. Finally, we show that CspC is required for survival during challenge by the human immune system and is key for A. baumannii dissemination and/or colonization during systemic infection. Collectively, our work identifies a cadre of new biofilm-associated genes within A. baumannii and provides unique insight into the global regulatory network of this emerging human pathogen.
Collapse
Affiliation(s)
- Brooke R. Tomlinson
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Grant A. Denham
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Nathanial J. Torres
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Robert S. Brzozowski
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Jessie L. Allen
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Jessica K. Jackson
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Prahathees J. Eswara
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Lindsey N. Shaw
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
20
|
The biofilm life cycle: expanding the conceptual model of biofilm formation. Nat Rev Microbiol 2022; 20:608-620. [PMID: 35922483 PMCID: PMC9841534 DOI: 10.1038/s41579-022-00767-0] [Citation(s) in RCA: 523] [Impact Index Per Article: 174.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 01/18/2023]
Abstract
Bacterial biofilms are often defined as communities of surface-attached bacteria and are typically depicted with a classic mushroom-shaped structure characteristic of Pseudomonas aeruginosa. However, it has become evident that this is not how all biofilms develop, especially in vivo, in clinical and industrial settings, and in the environment, where biofilms often are observed as non-surface-attached aggregates. In this Review, we describe the origin of the current five-step biofilm development model and why it fails to capture many aspects of bacterial biofilm physiology. We aim to present a simplistic developmental model for biofilm formation that is flexible enough to include all the diverse scenarios and microenvironments where biofilms are formed. With this new expanded, inclusive model, we hereby introduce a common platform for developing an understanding of biofilms and anti-biofilm strategies that can be tailored to the microenvironment under investigation.
Collapse
|
21
|
Dolma KG, Khati R, Paul AK, Rahmatullah M, de Lourdes Pereira M, Wilairatana P, Khandelwal B, Gupta C, Gautam D, Gupta M, Goyal RK, Wiart C, Nissapatorn V. Virulence Characteristics and Emerging Therapies for Biofilm-Forming Acinetobacter baumannii: A Review. BIOLOGY 2022; 11:biology11091343. [PMID: 36138822 PMCID: PMC9495682 DOI: 10.3390/biology11091343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022]
Abstract
Simple Summary Acinetobacter baumannii (A. baumannii) is one of the ESKAPE organisms and has the competency to build biofilms. These biofilms account for the most nosocomial infections all over the world. This review reflects on the various physicochemical and environmental factors such as adhesion, pili expression, growth surfaces, drug-resistant genes, and virulence factors that profoundly affect its resistant forte. Emerging drug-resistant issues and limitations to newer drugs are other factors affecting the hospital environment. Here, we discuss newer and alternative methods that can significantly enhance the susceptibility to Acinetobacter spp. Many new antibiotics are under trials, such as GSK-3342830, The Cefiderocol (S-649266), Fimsbactin, and similar. On the other hand, we can also see the impact of traditional medicine and the secondary metabolites of these natural products’ application in searching for new treatments. The field of nanoparticles has demonstrated effective antimicrobial actions and has exhibited encouraging results in the field of nanomedicine. The use of various phages such as vWUPSU and phage ISTD as an alternative treatment for its specificity and effectiveness is being investigated. Cathelicidins obtained synthetically or from natural sources can effectively produce antimicrobial activity in the micromolar range. Radioimmunotherapy and photodynamic therapy have boundless prospects if explored as a therapeutic antimicrobial strategy. Abstract Acinetobacter species is one of the most prevailing nosocomial pathogens with a potent ability to develop antimicrobial resistance. It commonly causes infections where there is a prolonged utilization of medical devices such as CSF shunts, catheters, endotracheal tubes, and similar. There are several strains of Acinetobacter (A) species (spp), among which the majority are pathogenic to humans, but A. baumannii are entirely resistant to several clinically available antibiotics. The crucial mechanism that renders them a multidrug-resistant strain is their potent ability to synthesize biofilms. Biofilms provide ample opportunity for the microorganisms to withstand the harsh environment and further cause chronic infections. Several studies have enumerated multiple physiological and virulence factors responsible for the production and maintenance of biofilms. To further enhance our understanding of this pathogen, in this review, we discuss its taxonomy, pathogenesis, current treatment options, global resistance rates, mechanisms of its resistance against various groups of antimicrobials, and future therapeutics.
Collapse
Affiliation(s)
- Karma G. Dolma
- Department of Microbiology, Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok 737102, Sikkim, India
| | - Rachana Khati
- Department of Microbiology, Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok 737102, Sikkim, India
| | - Alok K. Paul
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia
| | - Mohammed Rahmatullah
- Department of Biotechnology & Genetic Engineering, University of Development Alternative, Lalmatia, Dhaka 1207, Bangladesh
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Correspondence: (P.W.); (V.N.)
| | - Bidita Khandelwal
- Department of Medicine, Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok 737102, Sikkim, India
| | - Chamma Gupta
- Department of Biotechnology, Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok 737102, Sikkim, India
| | - Deepan Gautam
- Department of Microbiology, Sikkim Manipal Institute of Medical Sciences, Sikkim Manipal University, Gangtok 737102, Sikkim, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Ramesh K. Goyal
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Christophe Wiart
- Institute for Tropical Biology and Conservation, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
- Correspondence: (P.W.); (V.N.)
| |
Collapse
|
22
|
New C-6 functionalized quinoline NorA inhibitors strongly synergize with ciprofloxacin against planktonic and biofilm growing resistant Staphylococcus aureus strains. Eur J Med Chem 2022; 241:114656. [DOI: 10.1016/j.ejmech.2022.114656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022]
|
23
|
Wang Q, Wang P, Liu P, Ou J. Comparative Transcriptome Analysis Reveals Regulatory Factors Involved in Vibrio Parahaemolyticus Biofilm Formation. Front Cell Infect Microbiol 2022; 12:917131. [PMID: 35899046 PMCID: PMC9309355 DOI: 10.3389/fcimb.2022.917131] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Vibrio parahaemolyticus biofilm poses a serious threat to food safety and human health. However, there is limited knowledge of transcriptional regulatory mechanism during the biofilm formation of this organism. Hence, the RNA sequencing technique was employed to compare the differences in transcriptome profiles between planktonic and biofilm state of V. parahaemolyticus ATCC33847 in this study. Collections of mRNA from planktonic and biofilm cells cultured at 25°C for 36 h were sequenced by studying their biological characteristics. The results showed that there were significant differences in the expression levels of 956 genes in biofilms compared with planktonic cells. These differences suggested that two-component regulatory system (TCS) and quorum sensing (QS) regulated V. parahaemolyticus biofilm formation by affecting important factors such as flagellar motility, Extracellular polymeric substance (EPS) secretion, tripartite ATP-independent (TRAP) transport system and ATP binding cassette (ABC) transport system. The present work in transcriptomics serves as a basis for future studies examining the complex network systems that regulate bacterial biofilm formation.
Collapse
Affiliation(s)
- Qiuyu Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Pengfei Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Pingping Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jie Ou
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai, China
- Shanghai Engineering Research Center of Aquatic-Product Processing and Preservation, Shanghai, China
- *Correspondence: Jie Ou,
| |
Collapse
|
24
|
The Phenylacetic Acid Catabolic Pathway Regulates Antibiotic and Oxidative Stress Responses in Acinetobacter. mBio 2022; 13:e0186321. [PMID: 35467424 PMCID: PMC9239106 DOI: 10.1128/mbio.01863-21] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The opportunistic pathogen Acinetobacter baumannii is responsible for a wide range of infections that are becoming increasingly difficult to treat due to extremely high rates of multidrug resistance. Acinetobacter's pathogenic potential is thought to rely on a "persist and resist" strategy that facilitates its remarkable ability to survive under a variety of harsh conditions. The paa operon is involved in the catabolism of phenylacetic acid (PAA), an intermediate in phenylalanine degradation, and is the most differentially regulated pathway under many environmental conditions. We found that, under subinhibitory concentrations of antibiotics, A. baumannii upregulates expression of the paa operon while simultaneously repressing chaperone-usher Csu pilus expression and biofilm formation. These phenotypes are reverted either by exogenous addition of PAA and its nonmetabolizable derivative 4-fluoro-PAA or by a mutation that blocks PAA degradation. Interference with PAA degradation increases susceptibility to antibiotics and hydrogen peroxide treatment. Transcriptomic and proteomic analyses identified a subset of genes and proteins whose expression is affected by addition of PAA or disruption of the paa pathway. Finally, we demonstrated that blocking PAA catabolism results in attenuated virulence in a murine catheter-associated urinary tract infection (CAUTI) model. We conclude that the paa operon is part of a regulatory network that responds to antibiotic and oxidative stress and is important for virulence. PAA has known regulatory functions in plants, and our experiments suggest that PAA is a cross-kingdom signaling molecule. Interference with this pathway may lead, in the future, to novel therapeutic strategies against A. baumannii infections. IMPORTANCE Acinetobacter baumannii causes a wide range of infections that are difficult to treat due to increasing rates of multidrug resistance; however, the mechanisms that this pathogen uses to respond to stress are poorly understood. Here, we describe a new mechanism of stress signaling in Acinetobacter that is mediated by the metabolite phenylacetic acid (PAA). We found that disrupting PAA catabolism interfered with A. baumannii's ability to adapt to stress, leading to decreased antibiotic tolerance and hydrogen peroxide resistance. We propose that investigating this stress response could lead to the development of novel therapeutics. In fact, PAA derivatives constitute a group of FDA-approved nonsteroidal anti-inflammatory drugs that could potentially be repurposed as antivirulence therapies to target multidrug-resistant Acinetobacter infections.
Collapse
|
25
|
Djahanschiri B, Di Venanzio G, Distel JS, Breisch J, Dieckmann MA, Goesmann A, Averhoff B, Göttig S, Wilharm G, Feldman MF, Ebersberger I. Evolutionarily stable gene clusters shed light on the common grounds of pathogenicity in the Acinetobacter calcoaceticus-baumannii complex. PLoS Genet 2022; 18:e1010020. [PMID: 35653398 PMCID: PMC9162365 DOI: 10.1371/journal.pgen.1010020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022] Open
Abstract
Nosocomial pathogens of the Acinetobacter calcoaceticus-baumannii (ACB) complex are a cautionary example for the world-wide spread of multi- and pan-drug resistant bacteria. Aiding the urgent demand for novel therapeutic targets, comparative genomics studies between pathogens and their apathogenic relatives shed light on the genetic basis of human-pathogen interaction. Yet, existing studies are limited in taxonomic scope, sensing of the phylogenetic signal, and resolution by largely analyzing genes independent of their organization in functional gene clusters. Here, we explored more than 3,000 Acinetobacter genomes in a phylogenomic framework integrating orthology-based phylogenetic profiling and microsynteny conservation analyses. We delineate gene clusters in the type strain A. baumannii ATCC 19606 whose evolutionary conservation indicates a functional integration of the subsumed genes. These evolutionarily stable gene clusters (ESGCs) reveal metabolic pathways, transcriptional regulators residing next to their targets but also tie together sub-clusters with distinct functions to form higher-order functional modules. We shortlisted 150 ESGCs that either co-emerged with the pathogenic ACB clade or are preferentially found therein. They provide a high-resolution picture of genetic and functional changes that coincide with the manifestation of the pathogenic phenotype in the ACB clade. Key innovations are the remodeling of the regulatory-effector cascade connecting LuxR/LuxI quorum sensing via an intermediate messenger to biofilm formation, the extension of micronutrient scavenging systems, and the increase of metabolic flexibility by exploiting carbon sources that are provided by the human host. We could show experimentally that only members of the ACB clade use kynurenine as a sole carbon and energy source, a substance produced by humans to fine-tune the antimicrobial innate immune response. In summary, this study provides a rich and unbiased set of novel testable hypotheses on how pathogenic Acinetobacter interact with and ultimately infect their human host. It is a comprehensive resource for future research into novel therapeutic strategies. The spread of multi- and pan-drug resistant bacterial pathogens is a worldwide threat to human health. Understanding the genetics of host colonization and infection can substantially help in devising novel ways of treatment. Acinetobacter baumannii, a nosocomial pathogen ranked top by the World Health Organization in the list of bacteria for which novel therapeutic approaches are needed, is a prime example. Here, we have carved out the genetic make-up that distinguishes A. baumannii and its pathogenic next relatives from other and mostly apathogenic Acinetobacter species. We found a rich spectrum of pathways and regulatory modules that reveal how the pathogens have modified biofilm formation, iron scavenging, and their carbohydrate metabolism to adapt to their human host. Among these, the capability to metabolize kynurenine is particularly intriguing. Humans produce this substance to contain bacterial invaders and to fine-tune the innate immune response. But A. baumannii and closely related pathogens found a way to feed on kynurenine. This suggests that the pathogens might be able to dysregulate the human immune response. In summary, our study substantially deepens the understanding of how a highly critical pathogen interacts with its host, which substantially eases the identification of novel targets for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Bardya Djahanschiri
- Applied Bioinformatics Group, Inst. of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Gisela Di Venanzio
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Jesus S. Distel
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Jennifer Breisch
- Inst. of Molecular Biosciences, Department of Molecular Microbiology and Bioenergetics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Alexander Goesmann
- Bioinformatics and Systems Biology, Justus Liebig University Gießen, Gießen, Germany
| | - Beate Averhoff
- Inst. of Molecular Biosciences, Department of Molecular Microbiology and Bioenergetics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Stephan Göttig
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| | | | - Mario F. Feldman
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Ingo Ebersberger
- Applied Bioinformatics Group, Inst. of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt am Main, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIKF), Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Frankfurt am Main, Germany
- * E-mail:
| |
Collapse
|
26
|
Yang R, Lai B, Liao K, Liu B, Huang L, Li S, Gu J, Lin Z, Chen Y, Wang S, Qiu Y, Deng J, Chen S, Zhuo C, Zhou Y. Overexpression of BIT33_RS14560 Enhances the Biofilm Formation and Virulence of Acinetobacter baumannii. Front Microbiol 2022; 13:867770. [PMID: 35547150 PMCID: PMC9083411 DOI: 10.3389/fmicb.2022.867770] [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: 02/01/2022] [Accepted: 03/22/2022] [Indexed: 11/26/2022] Open
Abstract
Acinetobacter baumannii, a strictly aerobic, non-lactose fermented Gram-negative bacteria, is one of the important pathogens of nosocomial infection. Major facilitator superfamily (MFS) transporter membrane proteins are a class of proteins that widely exists in microbial genomes and have been revealed to be related to biofilm formation in a variety of microorganisms. However, as one of the MFS transporter membrane proteins, little is known about the role of BIT33_RS14560 in A. baumannii. To explore the effects of BIT33_RS14560 on biofilm formation of A. baumannii, the biofilm formation abilities of 62 isolates were firstly investigated and compared with their transcript levels of BIT33_RS14560. Then, this specific gene was over-expressed in a standard A. baumannii strain (ATCC 19606) and two isolates of extensively drug-resistant A. baumannii (XDR-Ab). Bacterial virulence was observed using a Galleria mellonella infection model. High-throughput transcriptome sequencing (RNA seq) was performed on ATCC 19606 over-expressed strain and its corresponding empty plasmid control strain. Spearman’s correlation analysis indicated a significant negative correlation (R = −0.569, p = 0.000) between the △CT levels of BIT33_RS1456 and biofilm grading of A. baumannii isolates. The amount of A. baumannii biofilm was relatively high within 12–48 h. Regardless of standard or clinical strains; the biofilm biomass in the BIT33_RS14560 overexpression group was significantly higher than that in the control group ( p < 0.0001). Kaplan–Meier survival curve analysis showed that the mortality of G. mellonella was significantly higher when infected with the BIT33_RS14560 overexpression strain (χ2 = 8.462, p = 0.004). RNA-Seq showed that the mRNA expression levels of three genes annotated as OprD family outer membrane porin, glycosyltransferase family 39 protein, and glycosyltransferase family 2 protein, which were related to bacterial adhesion, biofilm formation, and virulence, were significantly upregulated when BIT33_RS14560 was over-expressed. Our findings provided new insights in identifying potential drug targets for the inhibition of biofilm formation. We also developed a practical method to construct an over-expressed vector that can stably replicate in XDR-Ab isolates.
Collapse
Affiliation(s)
- Ruifu Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bipeng Lai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kang Liao
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Baomo Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lixia Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaoli Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jincui Gu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziying Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yili Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuaishuai Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanli Qiu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiating Deng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Simin Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chao Zhuo
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanbin Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Stewart PS, Williamson KS, Boegli L, Hamerly T, White B, Scott L, Hu X, Mumey BM, Franklin MJ, Bothner B, Vital-Lopez FG, Wallqvist A, James GA. Search for a Shared Genetic or Biochemical Basis for Biofilm Tolerance to Antibiotics across Bacterial Species. Antimicrob Agents Chemother 2022; 66:e0002122. [PMID: 35266829 PMCID: PMC9017379 DOI: 10.1128/aac.00021-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/29/2022] [Indexed: 12/19/2022] Open
Abstract
Is there a universal genetically programmed defense providing tolerance to antibiotics when bacteria grow as biofilms? A comparison between biofilms of three different bacterial species by transcriptomic and metabolomic approaches uncovered no evidence of one. Single-species biofilms of three bacterial species (Pseudomonas aeruginosa, Staphylococcus aureus, and Acinetobacter baumannii) were grown in vitro for 3 days and then challenged with respective antibiotics (ciprofloxacin, daptomycin, and tigecycline) for an additional 24 h. All three microorganisms displayed reduced susceptibility in biofilms compared to planktonic cultures. Global transcriptomic profiling of gene expression comparing biofilm to planktonic and antibiotic-treated biofilm to untreated biofilm was performed. Extracellular metabolites were measured to characterize the utilization of carbon sources between biofilms, treated biofilms, and planktonic cells. While all three bacteria exhibited a species-specific signature of stationary phase, no conserved gene, gene set, or common functional pathway could be identified that changed consistently across the three microorganisms. Across the three species, glucose consumption was increased in biofilms compared to planktonic cells, and alanine and aspartic acid utilization were decreased in biofilms compared to planktonic cells. The reasons for these changes were not readily apparent in the transcriptomes. No common shift in the utilization pattern of carbon sources was discerned when comparing untreated to antibiotic-exposed biofilms. Overall, our measurements do not support the existence of a common genetic or biochemical basis for biofilm tolerance against antibiotics. Rather, there are likely myriad genes, proteins, and metabolic pathways that influence the physiological state of individual microorganisms in biofilms and contribute to antibiotic tolerance.
Collapse
Affiliation(s)
- Philip S. Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, USA
| | - Kerry S. Williamson
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Laura Boegli
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
| | - Timothy Hamerly
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Ben White
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Liam Scott
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Xiao Hu
- Gianforte School of Computing, Montana State University, Bozeman, Montana, USA
| | - Brendan M. Mumey
- Gianforte School of Computing, Montana State University, Bozeman, Montana, USA
| | - Michael J. Franklin
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Brian Bothner
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Francisco G. Vital-Lopez
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland, USA
| | - Garth A. James
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
28
|
Kruppa O, Czermak P. Screening for Biofilm-Stimulating Factors in the Freshwater Planctomycete Planctopirus limnophila to Improve Sessile Growth in a Chemically Defined Medium. Microorganisms 2022; 10:microorganisms10040801. [PMID: 35456851 PMCID: PMC9028447 DOI: 10.3390/microorganisms10040801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/31/2022] [Accepted: 04/10/2022] [Indexed: 12/26/2022] Open
Abstract
Planctomycetes such as Planctopirus limnophila offer a promising source of bioactive molecules, particularly when they switch from planktonic to sessile growth, but little is known about the corresponding biosynthetic gene clusters and how they are activated. We therefore screened for factors that promote sessile growth and biofilm formation to enable the cultivation of P. limnophila in a fixed-bed reactor. We carried out screening in microtiter plates focusing on biofilm formation and changes in optical density in response to various C:N ratios, metal ions, and oxidative stress. We used MTT assays and crystal violet staining to quantify biofilm formation. Positive factors were then validated in a fixed-bed bioreactor. The initial screen showed that D1ASO medium supplemented with NH4Cl to achieve a C:N ratio of 5.7:1, as well as 50 µM FeSO4 or CuSO4, increased the biofilm formation relative to the control medium. Exposure to H2O2 did not affect cell viability but stimulated biofilm formation. However, the same results were not replicated in the fixed-bed bioreactor, probably reflecting conditions that are unique to this environment such as the controlled pH and more vigorous aeration. Although we were able to cultivate P. limnophila in a fixed-bed bioreactor using a chemically defined medium, the factors that stimulate biofilm formation and inhibit planktonic growth were only identified in microtiter plates and further evaluation is required to establish optimal growth conditions in the bioreactor system.
Collapse
Affiliation(s)
- Oscar Kruppa
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany;
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany;
- Faculty of Biology and Chemistry, Justus-Liebig University of Giessen, 35390 Giessen, Germany
- Correspondence:
| |
Collapse
|
29
|
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]
|
30
|
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: 19] [Impact Index Per Article: 6.3] [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
|
31
|
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: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [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
|
32
|
Decoding Acinetobacter baumannii biofilm dynamics and associated protein markers: proteomic and bioinformatics approach. Arch Microbiol 2022; 204:200. [PMID: 35239017 DOI: 10.1007/s00203-022-02807-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 01/27/2022] [Accepted: 02/15/2022] [Indexed: 12/11/2022]
Abstract
Biofilm formation by Acinetobacter baumannii is one of the major cause of its persistence in hospital environment. Biofilm phenotypes are more resistant to physical as well as chemical stresses than their planktonic counterparts. The present study was carried in quest of biofilm-associated protein markers and their association with various biological pathways of A. baumannii. The study was designed with an aim to highlight the crucial common factor present in the majority of the A. baumannii strains irrespective of its resistance nature. A label-free proteome comparison of biofilm and planktonic phenotypes of A. baumannii was done using QExactive tandem mass spectrometry. Our investigation suggests key elevation of adhesion factors, acetate metabolism, nutrient transporters, and secretion system proteins are required for biofilm formation in A. baumannii. Elevation of biofilm-associated proteins revealed that biofilm is the unique phenotype with the potential to form robust matrix-embedded colonies and defeat stress condition. Further, core protein markers of biofilm phenotypes could be used as targets for new clinical interventions to combat biofilm-associated infections.
Collapse
|
33
|
Kaushik V, Tiwari M, Joshi R, Tiwari V. Therapeutic strategies against potential antibiofilm targets of multidrug-resistant Acinetobacter baumannii. J Cell Physiol 2022; 237:2045-2063. [PMID: 35083758 DOI: 10.1002/jcp.30683] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/30/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
Acinetobacter baumannii is the causative agent of various hospital-acquired infections. Biofilm formation is one of the various antimicrobial resistance (AMR) strategies and is associated with high mortality and morbidity. Hence, it is essential to review the potential antibiofilm targets in A. baumannii and come up with different strategies to combat these potential targets. This review covers different pathways involved in the regulation of biofilm formation in A. baumannii like quorum sensing (QS), cyclic-di-GMP signaling, two-component system (TCS), outer-membrane protein (ompA), and biofilm-associated protein (BAP). A newly discovered mechanism of electrical signaling-mediated biofilm formation and contact-dependent biofilm modulation has also been discussed. As biofilm formation and its maintenance in A. baumannii is facilitated by these potential targets, the detailed study of these targets and pathways can bring light to different therapeutic strategies such as anti-biofilm peptides, natural and synthetic molecule inhibitors, QS molecule degrading enzymes, and other strategies. These strategies may help in suppressing the lethality of biofilm-mediated infections. Targeting essential proteins/targets which are crucial for biofilm formation and regulation may render new therapeutic strategies that can aid in combating biofilm, thus reducing the recalcitrant infections and morbidity associated with the biofilm of A. baumannii.
Collapse
Affiliation(s)
- Vaishali Kaushik
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Richa Joshi
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
34
|
Robin B, Nicol M, Le H, Tahrioui A, Schaumann A, Vuillemenot JB, Vergoz D, Lesouhaitier O, Jouenne T, Hardouin J, Potron A, Perrot V, Dé E. MacAB-TolC Contributes to the Development of Acinetobacter baumannii Biofilm at the Solid–Liquid Interface. Front Microbiol 2022; 12:785161. [PMID: 35095797 PMCID: PMC8792954 DOI: 10.3389/fmicb.2021.785161] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Acinetobacter baumannii has emerged as one of the most problematic bacterial pathogens responsible for hospital-acquired and community infections worldwide. Besides its high capacity to acquire antibiotic resistance mechanisms, it also presents high adhesion abilities on inert and living surfaces leading to biofilm development. This lifestyle confers additional protection against various treatments and allows it to persist for long periods in various hospital niches. Due to their remarkable antimicrobial tolerance, A. baumannii biofilms are difficult to control and ultimately eradicate. Further insights into the mechanism of biofilm development will help to overcome this challenge and to develop novel antibiofilm strategies. To unravel critical determinants of this sessile lifestyle, the proteomic profiles of two A. baumannii strains (ATTC17978 and SDF) grown in planktonic stationary phase or in mature solid–liquid (S-L) biofilm were compared using a semiquantitative proteomic study. Of interest, among the 69 common proteins determinants accumulated in the two strains at the S-L interface, we sorted out the MacAB-TolC system. This tripartite efflux pump played a role in A. baumannii biofilm formation as demonstrated by using ΔmacAB-tolC deletion mutant. Complementary approaches allowed us to get an overview of the impact of macAB-tolC deletion in A. baumannii physiology. Indeed, this efflux pump appeared to be involved in the envelope stress response occurring in mature biofilm. It contributes to maintain wild type (WT) membrane rigidity and provides tolerance to high osmolarity conditions. In addition, this system is probably involved in the maintenance of iron and sulfur homeostasis. MacAB-TolC might help this pathogen face and adapt to deleterious conditions occurring in mature biofilms. Increasing our knowledge of A. baumannii biofilm formation will undoubtedly help us develop new therapeutic strategies to tackle this emerging threat to human health.
Collapse
Affiliation(s)
- Brandon Robin
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Marion Nicol
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Hung Le
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Ali Tahrioui
- Normandie Univ, UNIROUEN, LMSM EA4312, Evreux, France
| | - Annick Schaumann
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | | | - Delphine Vergoz
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | | | - Thierry Jouenne
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | - Julie Hardouin
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | - Anaïs Potron
- UMR 6249 Chrono-Environnement, CNRS-Université de Bourgogne/Franche-Comté, Besançon, France
| | - Valérie Perrot
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- *Correspondence: Valérie Perrot,
| | - Emmanuelle Dé
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- Emmanuelle Dé,
| |
Collapse
|
35
|
Integrating transcriptomics and metabolomics analysis on kojic acid combating Acinetobacter baumannii biofilm and its potential roles. Microbiol Res 2021; 254:126911. [PMID: 34763140 DOI: 10.1016/j.micres.2021.126911] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022]
Abstract
As a major secondary metabolite derived from a dominant marine filamentous fungus A7, kojic acid might confer the strain a competitive advantage in natural colonization. The bioactivities of kojic acid against bacterial growth and biofilm formation were investigated against Acinetobacter baumannii (A. baumannii) ATCC 19606. Then, transcriptomics and metabolomics were integrated to characterize the underlying mechanisms. It turned out that kojic acid exhibited a significantly suppressive impact against biofilm but a weak bacteriostatic activity. Meanwhile, a variety of transcriptional and metabolomic profiles were altered within biofilm formation as a result of kojic acid exposure. The alterations highlighted the mechanisms underlying biofilm formation, comprising of quorum sensing, fimbria assembly, bacterial virulence and metabolic plasticity, which could somewhat be hampered by kojic acid. The present study comprehensively elucidated multifactorial schemes for kojic acid combating biofilm formation of A. baumannii, which might provide mechanistic insights into the development of therapeutic strategies against this notorious pathogen. Meanwhile, our observations might shed new light on the ecological roles of kojic acid, e.g., serving as chemical deterrents for host adaptation to marine niches, which, however, awaits further validation.
Collapse
|
36
|
Massier S, Robin B, Mégroz M, Wright A, Harper M, Hayes B, Cosette P, Broutin I, Boyce JD, Dé E, Hardouin J. Phosphorylation of Extracellular Proteins in Acinetobacter baumannii in Sessile Mode of Growth. Front Microbiol 2021; 12:738780. [PMID: 34659171 PMCID: PMC8517400 DOI: 10.3389/fmicb.2021.738780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/30/2021] [Indexed: 11/21/2022] Open
Abstract
Acinetobacter baumannii is a problematic nosocomial pathogen owing to its increasing resistance to antibiotics and its great ability to survive in the hospital environment, which is linked to its capacity to form biofilms. Structural and functional investigations of post-translational modifications, such as phosphorylations, may lead to identification of candidates for therapeutic targets against this pathogen. Here, we present the first S/T/Y phosphosecretome of two A. baumannii strains, the reference strain ATCC 17978 and the virulent multi-drug resistant strain AB0057, cultured in two modes of growth (planktonic and biofilm) using TiO2 chromatography followed by high resolution mass spectrometry. In ATCC 17978, we detected a total of 137 (97 phosphoproteins) and 52 (33 phosphoproteins) phosphosites in biofilm and planktonic modes of growth, respectively. Similarly, in AB0057, 155 (119 phosphoproteins) and 102 (74 phosphoproteins) phosphosites in biofilm and planktonic modes of growth were identified, respectively. Both strains in the biofilm mode of growth showed a higher number of phosphosites and phosphoproteins compared to planktonic growth. Several phosphorylated sites are localized in key regions of proteins involved in either drug resistance (β-lactamases), adhesion to host tissues (pilins), or protein secretion (Hcp). Site-directed mutagenesis of the Hcp protein, essential for type VI secretion system-mediated interbacterial competition, showed that four of the modified residues are essential for type VI secretion system activity.
Collapse
Affiliation(s)
- Sébastien Massier
- Normandie Univ., UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | - Brandon Robin
- Normandie Univ., UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Marianne Mégroz
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Amy Wright
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Marina Harper
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Brooke Hayes
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Pascal Cosette
- Normandie Univ., UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| | | | - John D. Boyce
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Emmanuelle Dé
- Normandie Univ., UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
| | - Julie Hardouin
- Normandie Univ., UNIROUEN, INSA Rouen, CNRS, Polymers, Biopolymers, Surfaces Laboratory, Rouen, France
- PISSARO Proteomic Facility, IRIB, Mont-Saint-Aignan, France
| |
Collapse
|
37
|
Silva AMCMDA, Costa Júnior SD, Lima JLC, Farias Filho JLBDE, Cavalcanti IMF, Maciel MAV. Investigation of the association of virulence genes and biofilm production with infection and bacterial colonization processes in multidrug-resistant Acinetobacter spp. AN ACAD BRAS CIENC 2021; 93:e20210245. [PMID: 34550209 DOI: 10.1590/0001-3765202120210245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/24/2021] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to evaluate the phenotypic and molecular patterns of biofilm formation in infection and colonization isolates of Acinetobacter spp. from patients who were admitted in a public hospital of Recife-PE-Brazil in 2018-2019. For the biofilm phenotypic analysis, Acinetobacter spp. isolates were evaluated by the crystal violet staining method; the search of virulence genes (bap, ompA, epsA, csuE and bfmS) was performed by PCR; and the ERIC-PCR was performed for molecular typing. Amongst the 38 Acinetobacter spp. isolates, 20 were isolated from infections and 18 from colonization. The resistance profile pointed that 86.85% (33/38) of the isolates were multidrug-resistant, being three infection isolates, and two colonization isolates resistant to polymyxin B. All the isolates were able to produce biofilm and they had at least one of the investigated virulence genes on their molecular profile, but the bap gene was found in 100% of them. No clones were detected by ERIC-PCR. There was no correlation between biofilm formation and the resistance profile of the bacteria, neither to the molecular profile of the virulence genes. Thus, the ability of Acinetobacter spp. to form biofilm is probably related to the high frequency of virulence genes.
Collapse
Affiliation(s)
- Adriana Maria C M DA Silva
- Federal University of Pernambuco, Medical Science Center, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - Sérgio D Costa Júnior
- Federal University of Pernambuco, Medical Science Center, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - Jailton L C Lima
- Federal University of Pernambuco, Medical Science Center, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - José Luciano B DE Farias Filho
- Federal University of Pernambuco, Medical Science Center, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - Isabella M F Cavalcanti
- Federal University of Pernambuco, Laboratory of Immunopathology Keizo Asami (LIKA), Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - Maria Amélia V Maciel
- Federal University of Pernambuco, Medical Science Center, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50670-901 Recife, PE, Brazil
| |
Collapse
|
38
|
Sun X, Xiang J. Mechanism Underlying the Role of LuxR Family Transcriptional Regulator abaR in Biofilm Formation by Acinetobacter baumannii. Curr Microbiol 2021; 78:3936-3944. [PMID: 34522977 PMCID: PMC8439540 DOI: 10.1007/s00284-021-02654-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 09/02/2021] [Indexed: 11/26/2022]
Abstract
Our study attempted to explore the mechanism underlying the role of LuxR family transcriptional regulator abaR in biofilm formation by Acinetobacter baumannii. The abaR gene was knocked out in ATCC 17978 strain using homologous recombination method. The growth curve and biofilm formation were measured in the wild type and abaR gene knockdown strains. Transcriptome sequencing was performed in the wild type and abaR gene knockdown strains following 8 h of culture. The growth curve in the abaR gene knockdown strain was similar to that of the wild-type strain. Biofilm formation significantly declined in the abaR gene knockdown strain at 8 and 48 h after culture. A total of 137 differentially expressed genes (DEGs) were obtained including 20 downregulated DEGs and 117 upregulated DEGs. Genes with differential expression were closely related to viral procapsid maturation (GO:0046797), acetoin catabolism (GO:0045150), carbon metabolism (ko01200), and the glycolysis/gluconeogenesis (ko00010)-related pathways. The results of the eight verified expression DEGs were consistent with the results predicted by bioinformatics. AbaR gene knockdown significantly affected biofilm formation by A. baumannii ATCC 17978 strain. The glycolysis/gluconeogenesis pathways were significantly dysregulated and induced by abaR gene knockdown in A. baumannii.
Collapse
Affiliation(s)
- Xu Sun
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Xiang
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
Khadke SK, Lee JH, Kim YG, Raj V, Lee J. Assessment of Antibiofilm Potencies of Nervonic and Oleic Acid against Acinetobacter baumannii Using In Vitro and Computational Approaches. Biomedicines 2021; 9:biomedicines9091133. [PMID: 34572317 PMCID: PMC8466663 DOI: 10.3390/biomedicines9091133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 08/29/2021] [Indexed: 12/24/2022] Open
Abstract
Acinetobacter baumannii is a nosocomial pathogen, and its biofilms are tolerant to desiccation, nutrient starvation, and antimicrobial treatment on biotic and abiotic surfaces, tissues, and medical devices. Biofilm formation by A. baumannii is triggered by a quorum sensing cascade, and we hypothesized that fatty acids might inhibit its biofilm formation by interfering with quorum sensing. Initially, we investigated the antibiofilm activities of 24 fatty acids against A. baumannii ATCC 17978 and two clinical isolates. Among these fatty acids, two unsaturated fatty acids, nervonic and oleic acid, at 20 μg/mL significantly inhibited A. baumannii biofilm formation without affecting its planktonic cell growth (MICs were >500 μg/mL) and markedly decreased the motility of A. baumannii but had no toxic effect on the nematode Caenorhabditis elegans. Interestingly, molecular dynamic simulations showed that both fatty acids bind to the quorum sensing acyl homoserine lactone synthase (AbaI), and decent conformational stabilities of interactions between the fatty acids and AbaI were exhibited. Our results demonstrate that nervonic and oleic acid inhibit biofilm formation by A. baumannii strains and may be used as lead molecules for the control of persistent A. baumannii infections.
Collapse
Affiliation(s)
| | | | | | | | - Jintae Lee
- Correspondence: ; Tel.: +82-53-810-2533; Fax: +82-53-810-4631
| |
Collapse
|
40
|
De Silva PM, Patidar R, Graham CI, Brassinga AKC, Kumar A. A response regulator protein with antar domain, AvnR, in Acinetobacter baumannii ATCC 17978 impacts its virulence and amino acid metabolism. MICROBIOLOGY-SGM 2021; 166:554-566. [PMID: 32324528 DOI: 10.1099/mic.0.000913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acinetobacter baumannii, a Gram-negative coccobacillus, is notorious for its involvement in opportunistic infections around the world. Its resistance to antibiotics makes treatment of infections challenging. In this study, we describe a novel response regulator protein, AvnR (A1S_2006) that regulates virulence-related traits in A. baumannii ATCC17978. Sequence analysis suggests that AvnR is a CheY-like response regulator and contains the RNA-binding ANTAR (AmiR and NasR transcription anti-termination regulators) domain. We show that AvnR plays a role in regulating biofilm formation (on glass and plastic surfaces), surface motility, adhesion to A549 cells as well as in nitrogen metabolism in A. baumannii. RNA-Seq analysis revealed that avnR deletion results in altered expression of more than 150 genes (116 upregulated and 42 downregulated). RNA-Seq data suggest that altered biofilm formation and surface motility observed in the avnR deletion mutant is likely mediated by previously unknown pathways. Of note, was the altered expression of genes predicted to be involved in amino acid transport and metabolism in avnR deletion mutant. Biolog phenotypic array showed that deletion of avnR hampered A. baumannii ATCC17978's ability to metabolize various nitrogen sources, particularly that of glutamic acid, serine, histidine, aspartic acid, isoleucine and arginine. Taken together our data show that AvnR, the first ANTAR protein described in A. baumannii, affects virulence phenotypes as well as its ability to metabolize nitrogen sources.
Collapse
Affiliation(s)
- P Malaka De Silva
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Rakesh Patidar
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | |
Collapse
|
41
|
Henderson PJF, Maher C, Elbourne LDH, Eijkelkamp BA, Paulsen IT, Hassan KA. Physiological Functions of Bacterial "Multidrug" Efflux Pumps. Chem Rev 2021; 121:5417-5478. [PMID: 33761243 DOI: 10.1021/acs.chemrev.0c01226] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial multidrug efflux pumps have come to prominence in human and veterinary pathogenesis because they help bacteria protect themselves against the antimicrobials used to overcome their infections. However, it is increasingly realized that many, probably most, such pumps have physiological roles that are distinct from protection of bacteria against antimicrobials administered by humans. Here we undertake a broad survey of the proteins involved, allied to detailed examples of their evolution, energetics, structures, chemical recognition, and molecular mechanisms, together with the experimental strategies that enable rapid and economical progress in understanding their true physiological roles. Once these roles are established, the knowledge can be harnessed to design more effective drugs, improve existing microbial production of drugs for clinical practice and of feedstocks for commercial exploitation, and even develop more sustainable biological processes that avoid, for example, utilization of petroleum.
Collapse
Affiliation(s)
- Peter J F Henderson
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Claire Maher
- School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, New South Wales, Australia
| | - Liam D H Elbourne
- Department of Biomolecular Sciences, Macquarie University, Sydney 2109, New South Wales, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney 2019, New South Wales, Australia
| | - Bart A Eijkelkamp
- College of Science and Engineering, Flinders University, Bedford Park 5042, South Australia, Australia
| | - Ian T Paulsen
- Department of Biomolecular Sciences, Macquarie University, Sydney 2109, New South Wales, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney 2019, New South Wales, Australia
| | - Karl A Hassan
- School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, New South Wales, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney 2019, New South Wales, Australia
| |
Collapse
|
42
|
López-Martín M, Dubern JF, Alexander MR, Williams P. AbaM Regulates Quorum Sensing, Biofilm Formation, and Virulence in Acinetobacter baumannii. J Bacteriol 2021; 203:e00635-20. [PMID: 33495249 PMCID: PMC8088503 DOI: 10.1128/jb.00635-20] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/08/2021] [Indexed: 12/31/2022] Open
Abstract
Acinetobacter baumannii possesses a single divergent luxR/luxRI-type quorum-sensing (QS) locus named abaR/abaI This locus also contains a third gene located between abaR and abaI, which we term abaM, that codes for an uncharacterized member of the RsaM protein family known to regulate N-acylhomoserine lactone (AHL)-dependent QS in other beta- and gammaproteobacteria. Here, we show that disruption of abaM via a T26 insertion in A. baumannii strain AB5075 resulted in increased production of N-(3-hydroxydodecanoyl)-l-homoserine lactone and enhanced surface motility and biofilm formation. In contrast to the wild type and the abaI::T26 mutant, the virulence of the abaM::T26 mutant was completely attenuated in a Galleria mellonella infection model. Transcriptomic analysis of the abaM::T26 mutant revealed that AbaM differentially regulates at least 76 genes, including the csu pilus operon and the acinetin 505 lipopeptide biosynthetic operon, that are involved in surface adherence, biofilm formation and virulence. A comparison of the wild type, abaM::T26 and abaI::T26 transcriptomes, indicates that AbaM regulates ∼21% of the QS regulon including the csu operon. Moreover, the QS genes (abaI and abaR) were among the most upregulated in the abaM::T26 mutant. A. baumanniilux-based abaM reporter gene fusions revealed that abaM expression is positively regulated by QS but negatively autoregulated. Overall, the data presented in this work demonstrates that AbaM plays a central role in regulating A. baumannii QS, virulence, surface motility, and biofilm formation.IMPORTANCEAcinetobacter baumannii is a multiantibiotic-resistant pathogen of global health care importance. Understanding Acinetobacter virulence gene regulation could aid the development of novel anti-infective strategies. In A. baumannii, the abaR and abaI genes that code for the receptor and synthase components of an N-acylhomoserine (AHL) lactone-dependent quorum sensing system (QS) are separated by abaM Here, we show that although mutation of abaM increased AHL production, surface motility, and biofilm development, it resulted in the attenuation of virulence. AbaM was found to control both QS-dependent and QS-independent genes. The significance of this work lies in the identification of AbaM, an RsaM ortholog known to control virulence in plant pathogens, as a modulator of virulence in a human pathogen.
Collapse
Affiliation(s)
- Mario López-Martín
- Biodiscovery Institute, National Biofilms Innovation Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jean-Frédéric Dubern
- Biodiscovery Institute, National Biofilms Innovation Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Paul Williams
- Biodiscovery Institute, National Biofilms Innovation Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
43
|
Blaschke U, Skiebe E, Wilharm G. Novel Genes Required for Surface-Associated Motility in Acinetobacter baumannii. Curr Microbiol 2021; 78:1509-1528. [PMID: 33666749 PMCID: PMC7997844 DOI: 10.1007/s00284-021-02407-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/10/2021] [Indexed: 01/28/2023]
Abstract
Acinetobacter baumannii is an opportunistic and increasingly multi-drug resistant human pathogen rated as a critical priority one pathogen for the development of new antibiotics by the WHO in 2017. Despite the lack of flagella, A. baumannii can move along wet surfaces in two different ways: via twitching motility and surface-associated motility. While twitching motility is known to depend on type IV pili, the mechanism of surface-associated motility is poorly understood. In this study, we established a library of 30 A. baumannii ATCC® 17978™ mutants that displayed deficiency in surface-associated motility. By making use of natural competence, we also introduced these mutations into strain 29D2 to differentiate strain-specific versus species-specific effects of mutations. Mutated genes were associated with purine/pyrimidine/folate biosynthesis (e.g. purH, purF, purM, purE), alarmone/stress metabolism (e.g. Ap4A hydrolase), RNA modification/regulation (e.g. methionyl-tRNA synthetase), outer membrane proteins (e.g. ompA), and genes involved in natural competence (comEC). All tested mutants originally identified as motility-deficient in strain ATCC® 17978™ also displayed a motility-deficient phenotype in 29D2. By contrast, further comparative characterization of the mutant sets of both strains regarding pellicle biofilm formation, antibiotic resistance, and virulence in the Galleria mellonella infection model revealed numerous strain-specific mutant phenotypes. Our studies highlight the need for comparative analyses to characterize gene functions in A. baumannii and for further studies on the mechanisms underlying surface-associated motility.
Collapse
Affiliation(s)
- Ulrike Blaschke
- Robert Koch Institute, Project group P2, Burgstr. 37, 38855, Wernigerode, Germany.
| | - Evelyn Skiebe
- Robert Koch Institute, Project group P2, Burgstr. 37, 38855, Wernigerode, Germany
| | - Gottfried Wilharm
- Robert Koch Institute, Project group P2, Burgstr. 37, 38855, Wernigerode, Germany.
| |
Collapse
|
44
|
Mea HJ, Yong PVC, Wong EH. An overview of Acinetobacter baumannii pathogenesis: Motility, adherence and biofilm formation. Microbiol Res 2021; 247:126722. [PMID: 33618061 DOI: 10.1016/j.micres.2021.126722] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
The Gram-negative opportunistic pathogen Acinetobacter baumannii has gain notoriety in recent decades, primarily due to its propensity to cause nosocomial infections in critically ill patients. Its global spread, multi-drug resistance features and plethora of virulence factors make it a serious threat to public health worldwide. Though much effort has been expended in uncovering its successes, it continues to confound researchers due to its highly adaptive nature, mutating to meet the needs of a given environment. Its persistence in the clinical setting allows it to be in close proximity to a potential host, where contact can be made facilitating infection and colonization. In this article, we aim to provide a current overview of the bacterial virulence factors, specifically focusing on factors involved in the initial stages of infection, highlighting the role of adaptation facilitated by two-component systems and biofilm formation. Finally, the study of host-pathogen interactions using available animal models, their suitability, notable findings and some perspectives moving forward are also discussed.
Collapse
Affiliation(s)
- Hing Jian Mea
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Phelim Voon Chen Yong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
45
|
Dollery SJ, Zurawski DV, Gaidamakova EK, Matrosova VY, Tobin JK, Wiggins TJ, Bushnell RV, MacLeod DA, Alamneh YA, Abu-Taleb R, Escatte MG, Meeks HN, Daly MJ, Tobin GJ. Radiation-Inactivated Acinetobacter baumannii Vaccine Candidates. Vaccines (Basel) 2021; 9:vaccines9020096. [PMID: 33514059 PMCID: PMC7912630 DOI: 10.3390/vaccines9020096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/20/2022] Open
Abstract
Acinetobacter baumannii is a bacterial pathogen that is often multidrug-resistant (MDR) and causes a range of life-threatening illnesses, including pneumonia, septicemia, and wound infections. Some antibiotic treatments can reduce mortality if dosed early enough before an infection progresses, but there are few other treatment options when it comes to MDR-infection. Although several prophylactic strategies have been assessed, no vaccine candidates have advanced to clinical trials or have been approved. Herein, we rapidly produced protective whole-cell immunogens from planktonic and biofilm-like cultures of A. baumannii, strain AB5075 grown using a variety of methods. After selecting a panel of five cultures based on distinct protein profiles, replicative activity was extinguished by exposure to 10 kGy gamma radiation in the presence of a Deinococcus antioxidant complex composed of manganous (Mn2+) ions, a decapeptide, and orthophosphate. Mn2+ antioxidants prevent hydroxylation and carbonylation of irradiated proteins, but do not protect nucleic acids, yielding replication-deficient immunogenic A. baumannii vaccine candidates. Mice were immunized and boosted twice with 1.0 × 107 irradiated bacterial cells and then challenged intranasally with AB5075 using two mouse models. Planktonic cultures grown for 16 h in rich media and biofilm cultures grown in static cultures underneath minimal (M9) media stimulated immunity that led to 80–100% protection.
Collapse
Affiliation(s)
- Stephen J. Dollery
- Biological Mimetics, Inc., 124 Byte Drive, Frederick, MD 21702, USA; (J.K.T.); (T.J.W.); (R.V.B.); (D.A.M.); (G.J.T.)
- Correspondence:
| | - Daniel V. Zurawski
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (D.V.Z.); (Y.A.A.); (R.A.-T.); (M.G.E.)
| | - Elena K. Gaidamakova
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (E.K.G.); (V.Y.M.); (M.J.D.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Vera Y. Matrosova
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (E.K.G.); (V.Y.M.); (M.J.D.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - John K. Tobin
- Biological Mimetics, Inc., 124 Byte Drive, Frederick, MD 21702, USA; (J.K.T.); (T.J.W.); (R.V.B.); (D.A.M.); (G.J.T.)
| | - Taralyn J. Wiggins
- Biological Mimetics, Inc., 124 Byte Drive, Frederick, MD 21702, USA; (J.K.T.); (T.J.W.); (R.V.B.); (D.A.M.); (G.J.T.)
| | - Ruth V. Bushnell
- Biological Mimetics, Inc., 124 Byte Drive, Frederick, MD 21702, USA; (J.K.T.); (T.J.W.); (R.V.B.); (D.A.M.); (G.J.T.)
| | - David A. MacLeod
- Biological Mimetics, Inc., 124 Byte Drive, Frederick, MD 21702, USA; (J.K.T.); (T.J.W.); (R.V.B.); (D.A.M.); (G.J.T.)
| | - Yonas A. Alamneh
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (D.V.Z.); (Y.A.A.); (R.A.-T.); (M.G.E.)
| | - Rania Abu-Taleb
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (D.V.Z.); (Y.A.A.); (R.A.-T.); (M.G.E.)
| | - Mariel G. Escatte
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (D.V.Z.); (Y.A.A.); (R.A.-T.); (M.G.E.)
| | | | - Michael J. Daly
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (E.K.G.); (V.Y.M.); (M.J.D.)
| | - Gregory J. Tobin
- Biological Mimetics, Inc., 124 Byte Drive, Frederick, MD 21702, USA; (J.K.T.); (T.J.W.); (R.V.B.); (D.A.M.); (G.J.T.)
| |
Collapse
|
46
|
Marr CM, MacDonald U, Trivedi G, Chakravorty S, Russo TA. An Evaluation of BfmR-Regulated Antimicrobial Resistance in the Extensively Drug Resistant (XDR) Acinetobacter baumannii Strain HUMC1. Front Microbiol 2020; 11:595798. [PMID: 33193275 PMCID: PMC7658413 DOI: 10.3389/fmicb.2020.595798] [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: 08/17/2020] [Accepted: 10/06/2020] [Indexed: 11/13/2022] Open
Abstract
Acinetobacter baumannii is a problematic pathogen due to its common expression of extensive drug resistance (XDR) and ability to survive in the healthcare environment. These characteristics are mediated, in part, by the signal transduction system BfmR/BfmS. We previously demonstrated, in antimicrobial sensitive clinical isolates, that BfmR conferred increased resistance to meropenem and polymyxin E. In this study, potential mechanisms were informed, in part, by a prior transcriptome analysis of the antimicrobial sensitive isolate AB307-0294, which identified the porins OprB and aquaporin (Omp33-36, MapA) as plausible mediators for resistance to hydrophilic antimicrobials such as meropenem. Studies were then performed in the XDR isolate HUMC1, since delineating resistance mechanisms in this genomic background would be more translationally relevant. In HUMC1 BfmR likewise increased meropenem and polymyxin E resistance and upregulated gene expression of OprB and aquaporin. However, the comparison of HUMC1 with isogenic mutant constructs demonstrated that neither OprB nor aquaporin affected meropenem resistance; polymyxin E susceptibility was also unaffected. Next, we determined whether BfmR-mediated biofilm production affected either meropenem or polymyxin E susceptibilities. Interestingly, biofilm formation increased resistance to polymyxin E, but had little, if any effect on meropenem activity. Additionally, BfmR mediated meropenem resistance, and perhaps polymyxin E resistance, was due to BfmR regulated factors that do not affect biofilm formation. These findings increase our understanding of the mechanisms by which BfmR mediates intrinsic antimicrobial resistance in a clinically relevant XDR isolate and suggest that the efficacy of different classes of antimicrobials may vary under biofilm inducing conditions.
Collapse
Affiliation(s)
- Candace M Marr
- Department of Medicine, University at Buffalo, Buffalo, NY, United States.,Erie County Medical Center, Buffalo, NY, United States
| | - Ulrike MacDonald
- Department of Medicine, University at Buffalo, Buffalo, NY, United States.,Veterans Affairs Western New York Healthcare System, Buffalo, NY, United States
| | - Grishma Trivedi
- Department of Medicine, University at Buffalo, Buffalo, NY, United States.,Veterans Affairs Western New York Healthcare System, Buffalo, NY, United States
| | | | - Thomas A Russo
- Department of Medicine, University at Buffalo, Buffalo, NY, United States.,Veterans Affairs Western New York Healthcare System, Buffalo, NY, United States.,Department of Microbiology and Immunology, University at Buffalo, Buffalo, NY, United States.,Witebsky Center for Microbial Pathogenesis, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
47
|
Munusamy K, Loke MF, Vadivelu J, Tay ST. LC-MS analysis reveals biological and metabolic processes essential for Candida albicans biofilm growth. Microb Pathog 2020; 152:104614. [PMID: 33202254 DOI: 10.1016/j.micpath.2020.104614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 12/22/2022]
Abstract
Candidiasis is the most common fungal infection associated with high morbidity and mortality among immunocompromised patients. The ability to form biofilm is essential for Candida albicans pathogenesis and drug resistance. In this study, the planktonic cell and biofilm proteomes of C. albicans SC5314 strain analyzed using Liquid Chromatography-Mass Spectrometry (LC-MS) were compared. In total, 280 and 449 proteins are annotated from the planktonic cell and biofilm proteomes, respectively. The biofilm proteome demonstrated significantly higher proportion of proteins associated with the endomembrane system, mitochondrion and cytoplasm than planktonic proteome. Among proteins detected, 143 and 207 biological processes are annotated, of which, 38 and 102 are specific to the planktonic cell and biofilm proteomes, respectively, while 105 are common biological processes. The specific biological processes of C. albicans planktonic cell proteome are associated with cell polarity, energy metabolism and nucleotide (purine) metabolism, oxido-reduction coenzyme metabolic process, monosaccharide and amino acid (methionine) biosynthesis, regulation of anatomical structure morphogenesis and cell cycling, and single organism reproduction. Meanwhile, regulation of cellular macromolecule biosynthesis and metabolism, transcription and gene expression are major biological processes specifically associated with C. albicans biofilm proteome. Biosynthesis of leucine, isoleucine, and thiocysteine are highlighted as planktonic-related pathways, whereas folate metabolism, fatty acid metabolism and biosynthesis of amino acids (lysine, serine and glycine) are highlighted as biofilm-related pathways. In summary, LC-MS-based proteomic analysis reveals different adaptative strategies of C. albicans via specific biological and metabolic processes for planktonic cell and biofilm lifestyles. The mass spectrometry data are available via ProteomeXchange with identifiers PXD007830 (for biofilm proteome) and PXD007831 (for planktonic cell proteome).
Collapse
Affiliation(s)
- Komathy Munusamy
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mun Fai Loke
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sun Tee Tay
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
48
|
Mayer C, Muras A, Parga A, Romero M, Rumbo-Feal S, Poza M, Ramos-Vivas J, Otero A. Quorum Sensing as a Target for Controlling Surface Associated Motility and Biofilm Formation in Acinetobacter baumannii ATCC ® 17978 TM. Front Microbiol 2020; 11:565548. [PMID: 33101239 PMCID: PMC7554515 DOI: 10.3389/fmicb.2020.565548] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
The important nosocomial pathogen Acinetobacter baumannii presents a quorum sensing (QS) system (abaI/abaR) mediated by acyl-homoserine-lactones (AHLs) and several quorum quenching (QQ) enzymes. However, the roles of this complex network in the control of the expression of important virulence-related phenotypes such as surface-associated motility and biofilm formation is not clear. Therefore, the effect of the mutation of the AHL synthase AbaI, and the exogenous addition of the QQ enzyme Aii20J on surface-associated motility and biofilm formation by A. baumannii ATCC® 17978TM was studied in detail. The effect of the enzyme on biofilm formation by several multidrug-resistant A. baumannii clinical isolates differing in their motility pattern was also tested. We provide evidence that a functional QS system is required for surface-associated motility and robust biofilm formation in A. baumannii ATCC® 17978TM. Important differences were found with the well-studied strain A. nosocomialis M2 regarding the relevance of the QS system depending on environmental conditions The in vitro biofilm-formation capacity of A. baumannii clinical strains was highly variable and was not related to the antibiotic resistance or surface-associated motility profiles. A high variability was also found in the sensitivity of the clinical strains to the action of the QQ enzyme, revealing important differences in virulence regulation between A. baumannii isolates and confirming that studies restricted to a single strain are not representative for the development of novel antimicrobial strategies. Extracellular DNA emerges as a key component of the extracellular matrix in A. baumannii biofilms since the combined action of the QQ enzyme Aii20J and DNase reduced biofilm formation in all tested strains. Results demonstrate that QQ strategies in combination with other enzymatic treatments such as DNase could represent an alternative approach for the prevention of A. baumannii colonization and survival on surfaces and the prevention and treatment of infections caused by this pathogen.
Collapse
Affiliation(s)
- Celia Mayer
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía, Edificio CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,National Biofilms Innovation Centre, Biodiscovery Institute and School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Andrea Muras
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía, Edificio CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Parga
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía, Edificio CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Romero
- National Biofilms Innovation Centre, Biodiscovery Institute and School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Soraya Rumbo-Feal
- Microbioloxía, Instituto de Investigación Biomédica da Coruña, Centro de Investigacións Científicas Avanzadas da Coruña, Universidade da Coruña, A Coruña, Spain
| | - Margarita Poza
- Microbioloxía, Instituto de Investigación Biomédica da Coruña, Centro de Investigacións Científicas Avanzadas da Coruña, Universidade da Coruña, A Coruña, Spain
| | - José Ramos-Vivas
- Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Valdecilla, Santander, Spain
| | - Ana Otero
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía, Edificio CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
49
|
AlMatar M, Albarri O, Makky EA, Köksal F. Efflux pump inhibitors: new updates. Pharmacol Rep 2020; 73:1-16. [PMID: 32946075 DOI: 10.1007/s43440-020-00160-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/17/2022]
Abstract
The discovery of antibiotics ought to have ended the issue of bacterial infections, but this was not the case as it has led to the evolution of various mechanisms of bacterial resistance against various antibiotics. The efflux pump remains one of the mechanisms through which organisms develop resistance against antibiotics; this is because organisms can extrude most of the clinically relevant antibiotics from the interior cell environment to the exterior environment via the efflux pumps. Efflux pumps are thought to contribute significantly to biofilm formation as highlighted by various studies. Therefore, the inhibition of these efflux pumps can be a potential way of improving the activity of antibiotics, particularly now that the discovery of novel antibiotics is becoming tedious. Efflux pump inhibitors (EPIs) are molecules that can inhibit efflux pumps; they have been considered potential therapeutic agents for rejuvenating the activity of antibiotics that have already lost their activity against bacteria. However, studies are yet to determine the specific substrates for such pumps; the effect of altered efflux activity of these pumps on biofilm formation is still being investigated. A clear knowledge of the involvement of efflux pumps in biofilm development could aid in developing new agents that can interfere with their function and help to prevent biofilms formation; thereby, improving the outcome of treatment strategies. This review focuses on the novel update of EPIs and discusses the evidence of the roles of efflux pumps in biofilm formation; the potential approaches towards overcoming the increasing problem of biofilm-based infections are also discussed.
Collapse
Affiliation(s)
- Manaf AlMatar
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), 26300, Gambang, Kuantan, Malaysia.
| | - Osman Albarri
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitüsü), Çukurova University, Adana, Turkey
| | - Essam A Makky
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), 26300, Gambang, Kuantan, Malaysia.
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
50
|
Martínez-Guitián M, Vázquez-Ucha JC, Álvarez-Fraga L, Conde-Pérez K, Vallejo JA, Perina A, Bou G, Poza M, Beceiro A. Global Transcriptomic Analysis During Murine Pneumonia Infection Reveals New Virulence Factors in Acinetobacter baumannii. J Infect Dis 2020; 223:1356-1366. [DOI: 10.1093/infdis/jiaa522] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
Infections caused by multidrug-resistant pathogens such as Acinetobacter baumannii constitute a major health problem worldwide. In this study we present a global in vivo transcriptomic analysis of A. baumannii isolated from the lungs of mice with pneumonia infection.
Methods
Mice were infected with A. baumannii ATCC 17978 and AbH12O-A2 strains and the total bacterial RNA were analyzed by RNA sequencing. Lists of differentially expressed genes were obtained and 14 of them were selected for gene deletion and further analysis.
Results
Transcriptomic analysis revealed a specific gene expression profile in A. baumannii during lung infection with upregulation of genes involved in iron acquisition and host invasion. Mutant strains lacking feoA, mtnN, yfgC, basB, hisF, oatA, and bfnL showed a significant loss of virulence in murine pneumonia. A decrease in biofilm formation, adherence to human epithelial cells, and growth rate was observed in selected mutants.
Conclusions
This study provides an insight into A. baumannii gene expression profile during murine pneumonia infection. Data revealed that 7 in vivo upregulated genes were involved in virulence and could be considered new therapeutic targets.
Collapse
Affiliation(s)
- Marta Martínez-Guitián
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| | - Juan C Vázquez-Ucha
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| | - Laura Álvarez-Fraga
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| | - Kelly Conde-Pérez
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| | - Juan A Vallejo
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| | | | - Germán Bou
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| | - Margarita Poza
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| | - Alejandro Beceiro
- Servicio de Microbiología del Complejo Hospitalario Universitario, Instituto de Investigación Biomédica, Centro de Investigaciones Científicas Avanzadas, Universidad de A Coruña, A Coruña, Spain
| |
Collapse
|