1
|
Kavanaugh LG, Hinrichsen ME, Dunham CM, Conn GL. Regulation, structure, and activity of the Pseudomonas aeruginosa MexXY efflux system. Antimicrob Agents Chemother 2025; 69:e0182524. [PMID: 40192483 PMCID: PMC12057347 DOI: 10.1128/aac.01825-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025] Open
Abstract
The current crisis in bacterial antibiotic resistance can be attributed to the overuse (or misuse) of these essential medicines in healthcare and agriculture, coupled with the slowed progression of new drug development. In the versatile, opportunistic pathogen Pseudomonas aeruginosa, the Resistance-Nodulation-Division (RND) efflux pump MexXY plays critical roles in both cell physiology and the acquisition of multidrug resistance. The mexXY operon is not constitutively expressed, but this process is instead controlled by a complex network of multiple interconnected regulatory mechanisms. These include induction by several of the pump's ribosome-targeting antibiotic substrates and transcriptional repression and anti-repression processes that are themselves influenced by various cellular factors, processes, or stresses. Although extensive studies of the MexXY complex are currently lacking as compared to other RND efflux pumps such as Escherichia coli AcrAB-TolC, recent studies have provided valuable insights into the MexXY architecture and substrate profiles, including its contribution to clinical resistance. Furthermore, while MexXY primarily associates with the outer membrane protein OprM, emerging evidence suggests that this transporter-periplasmic adaptor pair may also partner with other outer membrane proteins, potentially to alter the efflux substrate profile and activity under specific environmental conditions. In this minireview, we summarize current understanding of MexXY regulation, structure, and substrate selectivity within the context of clinical resistance and as a framework for future efflux pump inhibitor development.
Collapse
Affiliation(s)
- Logan G. Kavanaugh
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
- Graduate Program in Microbiology and Molecular Genetics, Emory University, Atlanta, Georgia, USA
| | - Megan E. Hinrichsen
- Department of Chemistry, Emory University, Atlanta, Georgia, USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, Georgia, USA
| | - Christine M. Dunham
- Department of Chemistry, Emory University, Atlanta, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Graeme L. Conn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Gomes VMS, Bulla ACS, Torres PHM, Leal da Silva M. RND/HAE-1 members in the Pseudomonadota phylum: exploring multidrug resistance. Biophys Rev 2025; 17:687-699. [PMID: 40376394 PMCID: PMC12075780 DOI: 10.1007/s12551-025-01297-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 02/24/2025] [Indexed: 05/18/2025] Open
Abstract
The hydrophobe/amphiphile efflux-1 (HAE-1) family, part of the Resistance-Nodulation-Division (RND) superfamily, plays a critical role in the development of multidrug resistance (MDR) in bacteria. Known for its broad substrate transport capacity, this family of efflux pumps can actively expel a wide range of molecules, including antibiotics, salts, and dyes, thereby reducing the intracellular concentration of toxic substances. These transporters, which form efflux systems, are primarily found in bacteria within the phylum Pseudomonadota (Proteobacteria), where they are strongly associated with increased resistance and enhanced virulence, thus contributing to bacterial survival in hostile environments. In addition, efflux systems are composed of two other protein components: Membrane Fusion Proteins (MFPs) and Outer Membrane Factors (OMFs). Notably, several bacterial species identified by the World Health Organization (WHO) as urgent priorities for new antibiotic development, such as Escherichia coli and Pseudomonas aeruginosa, have well-studied HAE-1 efflux systems, such as AcrAB-TolC and MexAB-OprM. These systems efficiently transport molecules from the periplasm to the extracellular space, facilitating bacterial persistence. In this review, we examined the current knowledge of HAE-1 efflux transporters and their roles in the physiology and survival of bacteria in the Pseudomonadota phylum.
Collapse
Affiliation(s)
- Vinnícius Machado Schelk Gomes
- Programa de Pós-Graduação Em Biologia Computacional E Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ 21040-900 Brazil
| | - Ana Carolina Silva Bulla
- Programa de Pós-Graduação Em Biologia Computacional E Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ 21040-900 Brazil
| | - Pedro Henrique Monteiro Torres
- Programa de Pós-Graduação em Ciências Biológicas – Biofísica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho 373 - CCS - Bloco G1-19, Cidade Universitária, Rio de Janeiro, RJ 21941-902 Brazil
| | - Manuela Leal da Silva
- Programa de Pós-Graduação Em Biologia Computacional E Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ 21040-900 Brazil
- Programa de Pós-Graduação Multicêntrico Em Ciências Fisiológicas, Instituto de Biodiversidade E Sustentabilidade NUPEM, Universidade Federal Do Rio de Janeiro, Avenida São José Do Barreto, 764. Centro, Macaé, RJ 27965-045 Brazil
| |
Collapse
|
3
|
Geremia N, Marino A, De Vito A, Giovagnorio F, Stracquadanio S, Colpani A, Di Bella S, Madeddu G, Parisi SG, Stefani S, Nunnari G. Rare or Unusual Non-Fermenting Gram-Negative Bacteria: Therapeutic Approach and Antibiotic Treatment Options. Antibiotics (Basel) 2025; 14:306. [PMID: 40149115 PMCID: PMC11939765 DOI: 10.3390/antibiotics14030306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Non-fermenting Gram-negative bacteria (NFGNB) are a heterogeneous group of opportunistic pathogens increasingly associated with healthcare-associated infections. While Pseudomonas aeruginosa, Acinetobacter baumannii, and Stenotrophomonas maltophilia are well known, rarer species such as Burkholderia cepacia complex, Achromobacter spp., Chryseobacterium spp., Elizabethkingia spp., Ralstonia spp., and others pose emerging therapeutic challenges. Their intrinsic and acquired resistance mechanisms limit effective treatment options, making targeted therapy essential. Objectives: This narrative review summarizes the current understanding of rare and unusual NFGNB, their clinical significance, resistance profiles, and evidence-based therapeutic strategies. Methods: A literature review was conducted using PubMed, Scopus, and Web of Science to identify relevant studies on the epidemiology, antimicrobial resistance, and treatment approaches to rare NFGNB. Results: Rare NFGNB exhibits diverse resistance mechanisms, including β-lactamase production, efflux pumps, and porin modifications. Treatment selection depends on species-specific susceptibility patterns, but some cornerstones can be individuated. Novel β-lactam/β-lactamase inhibitors and combination therapy approaches are being explored for multidrug-resistant isolates. However, clinical data remain limited. Conclusions: The increasing incidence of rare NFGNB requires heightened awareness and a tailored therapeutic approach. Given the paucity of clinical guidelines, antimicrobial stewardship and susceptibility-guided treatment are crucial in optimizing patient outcomes.
Collapse
Affiliation(s)
- Nicholas Geremia
- Unit of Infectious Diseases, Department of Clinical Medicine, Ospedale “dell’Angelo”, 30174 Venice, Italy;
- Unit of Infectious Diseases, Department of Clinical Medicine, Ospedale Civile “S.S. Giovanni e Paolo”, 30122 Venice, Italy
| | - Andrea Marino
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS Garibaldi Hospital, University of Catania, 95122 Catania, Italy;
| | - Andrea De Vito
- Unit of Infectious Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.D.V.); (A.C.); (G.M.)
| | - Federico Giovagnorio
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy; (F.G.); (S.G.P.)
| | - Stefano Stracquadanio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.S.); (S.S.)
| | - Agnese Colpani
- Unit of Infectious Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.D.V.); (A.C.); (G.M.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34129 Trieste, Italy;
| | - Giordano Madeddu
- Unit of Infectious Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.D.V.); (A.C.); (G.M.)
| | | | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.S.); (S.S.)
| | - Giuseppe Nunnari
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS Garibaldi Hospital, University of Catania, 95122 Catania, Italy;
| |
Collapse
|
4
|
Laklaeng SN, Songsri J, Wisessombat S, Mala W, Phothaworn P, Senghoi W, Nuinoon M, Tangphatsornruang S, Wongtawan T, Hayakijkosol O, Kerdsin A, Klangbud WK. Multi-locus sequence typing and genetic diversity of antibiotic-resistant genes and virulence-associated genes in Burkholderia pseudomallei: Insights from whole genome sequencing of animal and environmental isolates in Thailand. Vet Microbiol 2024; 298:110236. [PMID: 39216325 DOI: 10.1016/j.vetmic.2024.110236] [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: 01/21/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Burkholderia pseudomallei is a Gram-negative bacillus and the etiological agent of melioidosis in humans and animals. The disease is highly endemic in northern Australia and Southeast Asia. Comprehensive genomic data are essential for understanding the bacteria's dissemination and genetic relationships among strains from different geographical regions. In this study, we conducted antimicrobial susceptibility testing and whole-genome sequencing of 54 B. pseudomallei isolates obtained from environmental and animal sources in southern Thailand between 2011 and 2018. Their genomics were determined of antibiotic-resistant genes (ARGs), virulence-associated genes, mobile genetic elements (MGEs), sequence types (STs), and single nucleotide polymorphisms (SNPs) to evaluate their epidemiological relatedness. Remarkably, all 54 isolates displayed sensitivity to antimicrobial agents typically used for melioidosis treatment. We identified nine distinct sequence types: ST392, ST51, ST409, ST508, ST376, ST1721, ST389, ST395, and ST289. Oxacillinase genes and the resistance nodulation family of efflux pumps (RND) were identified as contributors to antimicrobial resistance. Phylogenetic analysis demonstrated close genetic relations with other strains isolated from Southeast Asia. Furthermore, 172 virulence-associated genes were identified among the isolates, suggesting variations in clinical presentations. These findings underscore the importance of ongoing molecular genetic surveillance of B. pseudomallei for effective healthcare management and reducing melioidosis mortality.
Collapse
Affiliation(s)
- Sa-Ngob Laklaeng
- Health Sciences (International Program), Collage of Graduate Studies, Walailak University, Nakhon Si Thammarat, Thailand
| | - Jirarat Songsri
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand; Center of Excellence Research for Melioidosis and Microorganisms, Walailak University, Nakhon Si Thammarat, Thailand
| | - Sueptrakool Wisessombat
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand; Center of Excellence Research for Melioidosis and Microorganisms, Walailak University, Nakhon Si Thammarat, Thailand
| | - Wanida Mala
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand; Center of Excellence Research for Melioidosis and Microorganisms, Walailak University, Nakhon Si Thammarat, Thailand
| | - Preeda Phothaworn
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand; Center of Excellence Research for Melioidosis and Microorganisms, Walailak University, Nakhon Si Thammarat, Thailand
| | - Wilaiwan Senghoi
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand; Center of Excellence Research for Melioidosis and Microorganisms, Walailak University, Nakhon Si Thammarat, Thailand
| | - Manit Nuinoon
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Sithichoke Tangphatsornruang
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Teumpong Wongtawan
- Akkhraratchakumari Veterinary College, Walailak University, Nakhon Si Thammarat, Thailand
| | - Orachun Hayakijkosol
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Australia
| | - Anusak Kerdsin
- Faculty of Public Health, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, Thailand
| | - Wiyada Kwanhian Klangbud
- Medical Technology Program, Faculty of Science, Nakhon Phanom University, Nakhon Phanom, Thailand.
| |
Collapse
|
5
|
Hussin A, Nathan S, Shahidan MA, Nor Rahim MY, Zainun MY, Khairuddin NAN, Ibrahim N. Identification and mechanism determination of the efflux pump subunit amrB gene mutations linked to gentamicin susceptibility in clinical Burkholderia pseudomallei from Malaysian Borneo. Mol Genet Genomics 2024; 299:12. [PMID: 38381232 DOI: 10.1007/s00438-024-02105-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/29/2023] [Indexed: 02/22/2024]
Abstract
The bacterium Burkholderia pseudomallei is typically resistant to gentamicin but rare susceptible strains have been isolated in certain regions, such as Thailand and Sarawak, Malaysia. Recently, several amino acid substitutions have been reported in the amrB gene (a subunit of the amrAB-oprA efflux pump gene) that confer gentamicin susceptibility. However, information regarding the mechanism of the substitutions conferring the susceptibility is lacking. To understand the mechanism of amino acid substitution that confers susceptibility, this study identifies the corresponding mutations in clinical gentamicin-susceptible B. pseudomallei isolates from the Malaysian Borneo (n = 46; Sarawak: 5; Sabah: 41). Three phenotypically confirmed gentamicin-susceptible (GENs) strains from Sarawak, Malaysia, were screened for mutations in the amrB gene using gene sequences of gentamicin-resistant (GENr) strains (QEH 56, QEH 57, QEH20, and QEH26) and publicly available sequences (AF072887.1 and BX571965.1) as the comparator. The effect of missense mutations on the stability of the AmrB protein was determined by calculating the average energy change value (ΔΔG). Mutagenesis analysis identified a polymorphism-associated mutation, g.1056 T > G, a possible susceptible-associated in-frame deletion, Delta V412, and a previously confirmed susceptible-associated amino acid substitution, T368R, in each of the three GENs isolates. The contribution of Delta V412 needs further confirmation by experimental mutagenesis analysis. The mechanism by which T368R confers susceptibility, as elucidated by in silico mutagenesis analysis using AmrB-modeled protein structures, is proposed to be due to the location of T368R in a highly conserved region, rather than destabilization of the AmrB protein structure.
Collapse
Affiliation(s)
- Ainulkhir Hussin
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
- Department of Pathology, Queen Elizabeth Hospital, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | - Sheila Nathan
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Muhammad Ashraf Shahidan
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Mohd Yusof Nor Rahim
- Department of Pathology, Queen Elizabeth Hospital, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | - Mohamad Yusof Zainun
- Department of Pathology, Queen Elizabeth Hospital, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | | | - Nazlina Ibrahim
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia.
| |
Collapse
|
6
|
Zhang SP, Ye YP, Hou J, Ye ZR, Wang ZS, Yu XQ, Guo DD, Wang Y, He YX. Antitoxin MqsA decreases antibiotic susceptibility through the global regulator AgtR in Pseudomonas fluorescens. Antimicrob Agents Chemother 2023; 67:e0081223. [PMID: 37877694 PMCID: PMC10649091 DOI: 10.1128/aac.00812-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/31/2023] [Indexed: 10/26/2023] Open
Abstract
Type II toxin-antitoxin systems are highly prevalent in bacterial genomes and play crucial roles in the general stress response. Previously, we demonstrated that the type II antitoxin PfMqsA regulates biofilm formation through the global regulator AgtR in Pseudomonas fluorescens. Here, we found that both the C-terminal DNA-binding domain of PfMqsA and AgtR are involved in bacterial antibiotic susceptibility. Electrophoretic mobility shift assay (EMSA) analyses revealed that AgtR, rather than PfMqsA, binds to the intergenic region of emhABC-emhR, in which emhABC encodes an resistance-nodulation-cell division efflux pump and emhR encodes a repressor. Through quantitative real-time reverse-transcription PCR and EMSA analysis, we showed that AgtR directly activates the expression of the emhR by binding to the DNA motif [5´-CTAAGAAATATACTTAC-3´], leading to repression of the emhABC. Furthermore, we demonstrated that PfMqsA modulates the expression of EmhABC and EmhR. These findings enhance our understanding of the mechanism by which antitoxin PfMqsA contributes to antibiotic susceptibility.
Collapse
Affiliation(s)
- Si-Ping Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yi-Ping Ye
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Jun Hou
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Zi-Rui Ye
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Zhi-Song Wang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xiao-Quan Yu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Ding-Ding Guo
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Yong Wang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Jenkins CH, Scott AE, O’Neill PA, Norville IH, Prior JL, Ireland PM. The Arabinose 5-Phosphate Isomerase KdsD Is Required for Virulence in Burkholderia pseudomallei. J Bacteriol 2023; 205:e0003423. [PMID: 37458584 PMCID: PMC10448790 DOI: 10.1128/jb.00034-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/06/2023] [Indexed: 08/25/2023] Open
Abstract
Burkholderia pseudomallei is the causative agent of melioidosis, which is endemic primarily in Southeast Asia and northern Australia but is increasingly being seen in other tropical and subtropical regions of the world. Melioidosis is associated with high morbidity and mortality rates, which is mediated by the wide range of virulence factors encoded by B. pseudomallei. These virulence determinants include surface polysaccharides such as lipopolysaccharide (LPS) and capsular polysaccharides (CPS). Here, we investigated a predicted arabinose-5-phosphate isomerase (API) similar to KdsD in B. pseudomallei strain K96243. KdsD is required for the production of the highly conserved 3-deoxy-d-manno-octulosonic acid (Kdo), a key sugar in the core region of LPS. Recombinant KdsD was expressed and purified, and API activity was determined. Although a putative API paralogue (KpsF) is also predicted to be encoded, the deletion of kdsD resulted in growth defects, loss of motility, reduced survival in RAW 264.7 murine macrophages, and attenuation in a BALB/c mouse model of melioidosis. Suppressor mutations were observed during a phenotypic screen for motility, revealing single nucleotide polymorphisms or indels located in the poorly understood CPS type IV cluster. Crucially, suppressor mutations did not result in reversion of attenuation in vivo. This study demonstrates the importance of KdsD for B. pseudomallei virulence and highlights further the complex nature of the polysaccharides it produces. IMPORTANCE The intrinsic resistance of B. pseudomallei to many antibiotics complicates treatment. This opportunistic pathogen possesses a wide range of virulence factors, resulting in severe and potentially fatal disease. Virulence factors as targets for drug development offer an alternative approach to combat pathogenic bacteria. Prior to initiating early drug discovery approaches, it is important to demonstrate that disruption of the target gene will prevent the development of disease. This study highlights the fact that KdsD is crucial for virulence of B. pseudomallei in an animal model of infection and provides supportive phenotypic characterization that builds a foundation for future therapeutic development.
Collapse
Affiliation(s)
- Christopher H. Jenkins
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Salisbury, Wiltshire, United Kingdom
| | - Andrew E. Scott
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Salisbury, Wiltshire, United Kingdom
| | - Paul A. O’Neill
- University of Exeter Sequencing Service, Exeter, United Kingdom
| | - Isobel H. Norville
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Salisbury, Wiltshire, United Kingdom
- Biosciences Department, University of Exeter, Exeter, United Kingdom
| | - Joann L. Prior
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Salisbury, Wiltshire, United Kingdom
- Biosciences Department, University of Exeter, Exeter, United Kingdom
- Southampton General Hospital, Southampton, United Kingdom
| | - Philip M. Ireland
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Salisbury, Wiltshire, United Kingdom
| |
Collapse
|
8
|
Hall CM, Somprasong N, Hagen JP, Nottingham R, Sahl JW, Webb JR, Mayo M, Currie BJ, Podin Y, Wagner DM, Keim P, Schweizer HP. Exploring Cefiderocol Resistance Mechanisms in Burkholderia pseudomallei. Antimicrob Agents Chemother 2023; 67:e0017123. [PMID: 37133377 PMCID: PMC10269091 DOI: 10.1128/aac.00171-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/04/2023] [Indexed: 05/04/2023] Open
Abstract
Cefiderocol is a siderophore cephalosporin designed mainly for treatment of infections caused by β-lactam and multidrug-resistant Gram-negative bacteria. Burkholderia pseudomallei clinical isolates are usually highly cefiderocol susceptible, with in vitro resistance found in a few isolates. Resistance in clinical B. pseudomallei isolates from Australia is caused by a hitherto uncharacterized mechanism. We show that, like in other Gram-negatives, the PiuA outer membrane receptor plays a major role in cefiderocol nonsusceptibility in isolates from Malaysia.
Collapse
Affiliation(s)
- Carina M. Hall
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - Nawarat Somprasong
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - Johannah P. Hagen
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - Roxanne Nottingham
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
| | - Jason W. Sahl
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Jessica R. Webb
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Mark Mayo
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Bart J. Currie
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Yuwana Podin
- Institute of Health and Community Medicine, Universiti Malaysia Sarawak, Sarawak, Malaysia
| | - David M. Wagner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Paul Keim
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| | - Herbert P. Schweizer
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, USA
| |
Collapse
|
9
|
Burkholderia pseudomallei JW270 Is Lethal in the Madagascar Hissing Cockroach Infection Model and Can Be Utilized at Biosafety Level 2 to Identify Putative Virulence Factors. Infect Immun 2022; 90:e0015922. [PMID: 35862734 PMCID: PMC9387215 DOI: 10.1128/iai.00159-22] [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: 01/13/2023] Open
Abstract
Burkholderia pseudomallei, the causative agent of melioidosis, is classified by the CDC as a tier 1 select agent, and work involving it must be performed in a biosafety level 3 (BSL-3) laboratory. Three BSL-2 surrogate strains derived from B. pseudomallei 1026b, a virulent clinical isolate, have been removed from the CDC select agent list. These strains, Bp82, B0011, and JW270, are highly attenuated in rodent models of melioidosis and cannot be utilized to identify virulence determinants because of their high 50% lethal dose (LD50). We previously demonstrated that the Madagascar hissing cockroach (MHC) is a tractable surrogate host to study the innate immune response against Burkholderia. In this study, we found that JW270 maintains its virulence in MHCs. This surprising result indicates that it may be possible to identify potential virulence genes in JW270 by using MHCs at BSL-2. We tested this hypothesis by constructing JW270 mutations in genes that are required (hcp1) or dispensable (hcp2) for B. pseudomallei virulence in rodents. JW270 Δhcp1 was avirulent in MHCs and JW270 Δhcp2 was virulent, suggesting that MHCs can be used at BSL-2 for the discovery of important virulence factors. JW270 ΔBPSS2185, a strain harboring a mutation in a type IV pilin locus (TFP8) required for full virulence in BALB/c mice, was also found to be attenuated in MHCs. Finally, we demonstrate that the hmqA-G locus, which encodes the production of a family of secondary metabolites called 4-hydroxy-3-methyl-2-alkylquinolines, is important for JW270 virulence in MHCs and may represent a novel virulence determinant.
Collapse
|
10
|
Klimko CP, Shoe JL, Rill NO, Hunter M, Dankmeyer JL, Talyansky Y, Schmidt LK, Orne CE, Fetterer DP, Biryukov SS, Burtnick MN, Brett PJ, DeShazer D, Cote CK. Layered and integrated medical countermeasures against Burkholderia pseudomallei infections in C57BL/6 mice. Front Microbiol 2022; 13:965572. [PMID: 36060756 PMCID: PMC9432870 DOI: 10.3389/fmicb.2022.965572] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Burkholderia pseudomallei, the gram-negative bacterium that causes melioidosis, is notoriously difficult to treat with antibiotics. A significant effort has focused on identifying protective vaccine strategies to prevent melioidosis. However, when used as individual medical countermeasures both antibiotic treatments (therapeutics or post-exposure prophylaxes) and experimental vaccine strategies remain partially protective. Here we demonstrate that when used in combination, current vaccine strategies (recombinant protein subunits AhpC and/or Hcp1 plus capsular polysaccharide conjugated to CRM197 or the live attenuated vaccine strain B. pseudomallei 668 ΔilvI) and co-trimoxazole regimens can result in near uniform protection in a mouse model of melioidosis due to apparent synergy associated with distinct medical countermeasures. Our results demonstrated significant improvement when examining several suboptimal antibiotic regimens (e.g., 7-day antibiotic course started early after infection or 21-day antibiotic course with delayed initiation). Importantly, this combinatorial strategy worked similarly when either protein subunit or live attenuated vaccines were evaluated. Layered and integrated medical countermeasures will provide novel treatment options for melioidosis as well as diseases caused by other pathogens that are refractory to individual strategies, particularly in the case of engineered, emerging, or re-emerging bacterial biothreat agents.
Collapse
Affiliation(s)
- Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jennifer L. Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Nathaniel O. Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Yuli Talyansky
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Lindsey K. Schmidt
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - Caitlyn E. Orne
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - David P. Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Sergei S. Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Mary N. Burtnick
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Paul J. Brett
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - David DeShazer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| |
Collapse
|
11
|
Sharma A, Yadav SP, Sarma D, Mukhopadhaya A. Modulation of host cellular responses by gram-negative bacterial porins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:35-77. [PMID: 35034723 DOI: 10.1016/bs.apcsb.2021.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The outer membrane of a gram-negative bacteria encapsulates the plasma membrane thereby protecting it from the harsh external environment. This membrane acts as a sieving barrier due to the presence of special membrane-spanning proteins called "porins." These porins are β-barrel channel proteins that allow the passive transport of hydrophilic molecules and are impermeable to large and charged molecules. Many porins form trimers in the outer membrane. They are abundantly present on the bacterial surface and therefore play various significant roles in the host-bacteria interactions. These include the roles of porins in the adhesion and virulence mechanisms necessary for the pathogenesis, along with providing resistance to the bacteria against the antimicrobial substances. They also act as the receptors for phage and complement proteins and are involved in modulating the host cellular responses. In addition, the potential use of porins as adjuvants, vaccine candidates, therapeutic targets, and biomarkers is now being exploited. In this review, we focus briefly on the structure of the porins along with their important functions and roles in the host-bacteria interactions.
Collapse
Affiliation(s)
- Arpita Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Shashi Prakash Yadav
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Dwipjyoti Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
| |
Collapse
|
12
|
Bugrysheva JV, Lascols C, McLaughlin HP, Gee JE, Elrod MG, Sue D. Antimicrobial Susceptibility of Western Hemisphere Isolates of Burkholderia pseudomallei: Phenotypic and Genomic Analyses. Microb Drug Resist 2021; 27:1176-1185. [PMID: 33570476 PMCID: PMC10910562 DOI: 10.1089/mdr.2020.0362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Current antimicrobial treatment recommendations for melioidosis, the disease caused by Burkholderia pseudomallei, are largely based on studies of strains isolated from the Eastern Hemisphere (EH), where most human cases are identified and reported. In this study, we evaluated the antimicrobial susceptibility of 26 strains in the CDC (Centers for Diseases Control and Prevention) collection from the Western Hemisphere (WH) isolated from 1960 to 2015. Minimal inhibitory concentration (MIC) values were measured by standard broth microdilution for 16 antimicrobials following Clinical and Laboratory Standards Institute (CLSI) guidelines. Twenty-four of the 26 WH strains were susceptible to the six antimicrobials with CLSI-defined MIC susceptibility interpretive criteria for B. pseudomallei: amoxicillin/clavulanate, ceftazidime, imipenem, doxycycline, tetracycline, and trimethoprim/sulfamethoxazole. One WH strain demonstrated intermediate amoxicillin/clavulanate resistance and another strain had intermediate resistance to tetracycline. For all antimicrobials tested, the susceptibility profiles of WH isolates were comparable with previously reported MIC results of EH strains. The overall similarities suggest that the same antimicrobials are useful for melioidosis treatment in both the WH and EH. Using in silico analyses of WH genomes, we identified a novel amino acid substitution P258S in the beta-lactamase PenA, which may contribute to decreased susceptibility to amoxicillin/clavulanate in B. pseudomallei.
Collapse
Affiliation(s)
| | | | | | - Jay E Gee
- Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - Mindy G Elrod
- Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| | - David Sue
- Centers for Diseases Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Conservation of Resistance-Nodulation-Cell Division Efflux Pump-Mediated Antibiotic Resistance in Burkholderia cepacia Complex and Burkholderia pseudomallei Complex Species. Antimicrob Agents Chemother 2021; 65:e0092021. [PMID: 34181473 DOI: 10.1128/aac.00920-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Burkholderia cepacia complex (Bcc) and Burkholderia pseudomallei complex (Bpc) species include pathogens that are typically multidrug resistant. Dominant intrinsic and acquired multidrug resistance mechanisms are efflux mediated by pumps of the resistance-nodulation-cell division (RND) family. From comparative bioinformatic and, in many instances, functional studies, we infer that RND pump-based resistance mechanisms are conserved in Burkholderia. We propose to use these findings as a foundation for adoption of a uniform RND efflux pump nomenclature.
Collapse
|
14
|
Chattagul S, Khan MM, Scott AJ, Nita-Lazar A, Ernst RK, Goodlett DR, Sermswan RW. Transcriptomics Analysis Uncovers Transient Ceftazidime Tolerance in Burkholderia Biofilms. ACS Infect Dis 2021; 7:2324-2336. [PMID: 34138549 DOI: 10.1021/acsinfecdis.1c00003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Burkholderia pseudomallei is an etiological agent of melioidosis, a severe community-acquired infectious disease. B. pseudomallei strain K96243 is sensitive to the drug ceftazidime (CAZ), but has been shown to exhibit transient CAZ tolerance when in a biofilm form. To investigate an observed shift in gene expression profile during CAZ tolerance condition and to better understand the mechanistic aspects of this transient tolerance, RNA-sequencing was performed on B. pseudomallei K96243 from the following three states: planktonic, biofilm, and planktonic shedding. Results indicated that the expression of 651 genes (10.97%) were significantly changed in both biofilm (resistant) and planktonic shedding (sensitive) cells in comparison to the planktonic state. The top four highly expressed genes identified in both states are associated with nitrosative stress response (BPSL2368), Fe-S homeostasis (BPSL2369), and nitrate respiration (BPSS1154 and BPSS1158). Additionally, five orthologous genes, BPSL2370-BPSL2374, implicated in Fe-S cluster biogenesis, and another gene, BPSL2863, involved in DNA-binding of the stress protein ferritin, were shown to increase expression by RT-qPCR. The shift in gene expression was especially prominent at the late stages of biofilm growth (72 and 96 h), specifically in the biofilm-challenged CAZ survivor cells. This suggested that in response to stress in a biofilm, differential expression of these genes may support development of the CAZ tolerance in Burkholderia. The application of iron chelator deferoxamine (DFO) to the biofilm caused a significant reduction in biofilm formation and associated CAZ tolerance. Therefore, the shift in Fe-S metabolism when B. pseudomallei is in a biofilm may help stabilize the levels of reactive oxygen species (ROS), thereby limiting tolerance to CAZ.
Collapse
Affiliation(s)
- Supaksorn Chattagul
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Melioidosis Research Center, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Mohd M. Khan
- University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Alison J. Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry,Baltimore, Maryland 21201, United States
| | - Aleksandra Nita-Lazar
- Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry,Baltimore, Maryland 21201, United States
| | - David R. Goodlett
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry,Baltimore, Maryland 21201, United States
| | - Rasana W. Sermswan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Melioidosis Research Center, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
15
|
Alav I, Kobylka J, Kuth MS, Pos KM, Picard M, Blair JMA, Bavro VN. Structure, Assembly, and Function of Tripartite Efflux and Type 1 Secretion Systems in Gram-Negative Bacteria. Chem Rev 2021; 121:5479-5596. [PMID: 33909410 PMCID: PMC8277102 DOI: 10.1021/acs.chemrev.1c00055] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Indexed: 12/11/2022]
Abstract
Tripartite efflux pumps and the related type 1 secretion systems (T1SSs) in Gram-negative organisms are diverse in function, energization, and structural organization. They form continuous conduits spanning both the inner and the outer membrane and are composed of three principal components-the energized inner membrane transporters (belonging to ABC, RND, and MFS families), the outer membrane factor channel-like proteins, and linking the two, the periplasmic adaptor proteins (PAPs), also known as the membrane fusion proteins (MFPs). In this review we summarize the recent advances in understanding of structural biology, function, and regulation of these systems, highlighting the previously undescribed role of PAPs in providing a common architectural scaffold across diverse families of transporters. Despite being built from a limited number of basic structural domains, these complexes present a staggering variety of architectures. While key insights have been derived from the RND transporter systems, a closer inspection of the operation and structural organization of different tripartite systems reveals unexpected analogies between them, including those formed around MFS- and ATP-driven transporters, suggesting that they operate around basic common principles. Based on that we are proposing a new integrated model of PAP-mediated communication within the conformational cycling of tripartite systems, which could be expanded to other types of assemblies.
Collapse
Affiliation(s)
- Ilyas Alav
- Institute
of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Jessica Kobylka
- Institute
of Biochemistry, Biocenter, Goethe Universität
Frankfurt, Max-von-Laue-Straße 9, D-60438 Frankfurt, Germany
| | - Miriam S. Kuth
- Institute
of Biochemistry, Biocenter, Goethe Universität
Frankfurt, Max-von-Laue-Straße 9, D-60438 Frankfurt, Germany
| | - Klaas M. Pos
- Institute
of Biochemistry, Biocenter, Goethe Universität
Frankfurt, Max-von-Laue-Straße 9, D-60438 Frankfurt, Germany
| | - Martin Picard
- Laboratoire
de Biologie Physico-Chimique des Protéines Membranaires, CNRS
UMR 7099, Université de Paris, 75005 Paris, France
- Fondation
Edmond de Rothschild pour le développement de la recherche
Scientifique, Institut de Biologie Physico-Chimique, 75005 Paris, France
| | - Jessica M. A. Blair
- Institute
of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Vassiliy N. Bavro
- School
of Life Sciences, University of Essex, Colchester, CO4 3SQ United Kingdom
| |
Collapse
|
16
|
Hobson C, Chan AN, Wright GD. The Antibiotic Resistome: A Guide for the Discovery of Natural Products as Antimicrobial Agents. Chem Rev 2021; 121:3464-3494. [PMID: 33606500 DOI: 10.1021/acs.chemrev.0c01214] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The use of life-saving antibiotics has long been plagued by the ability of pathogenic bacteria to acquire and develop an array of antibiotic resistance mechanisms. The sum of these resistance mechanisms, the antibiotic resistome, is a formidable threat to antibiotic discovery, development, and use. The study and understanding of the molecular mechanisms in the resistome provide the basis for traditional approaches to combat resistance, including semisynthetic modification of naturally occurring antibiotic scaffolds, the development of adjuvant therapies that overcome resistance mechanisms, and the total synthesis of new antibiotics and their analogues. Using two major classes of antibiotics, the aminoglycosides and tetracyclines as case studies, we review the success and limitations of these strategies when used to combat the many forms of resistance that have emerged toward natural product-based antibiotics specifically. Furthermore, we discuss the use of the resistome as a guide for the genomics-driven discovery of novel antimicrobials, which are essential to combat the growing number of emerging pathogens that are resistant to even the newest approved therapies.
Collapse
Affiliation(s)
- Christian Hobson
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Andrew N Chan
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Gerard D Wright
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
17
|
Burkholderia ubonensis High-Level Tetracycline Resistance Is Due to Efflux Pump Synergy Involving a Novel TetA(64) Resistance Determinant. Antimicrob Agents Chemother 2021; 65:AAC.01767-20. [PMID: 33318011 DOI: 10.1128/aac.01767-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Burkholderia ubonensis, a nonpathogenic soil bacterium belonging to the Burkholderia cepacia complex (Bcc), is highly resistant to some clinically significant antibiotics. The concern is that B. ubonensis may serve as a resistance reservoir for Bcc or B. pseudomallei complex (Bpc) organisms that are opportunistic human pathogens. Using a B. ubonensis strain highly resistant to tetracycline (MIC, ≥256 µg/ml), we identified and characterized tetA(64) that encodes a novel tetracycline-specific efflux pump of the major facilitator superfamily. TetA(64) and associated TetR(64) regulator expression are induced by tetracyclines. Although TetA(64) is the primary tetracycline and doxycycline resistance determinant, maximum tetracycline and doxycycline resistance requires synergy between TetA(64) and the nonspecific AmrAB-OprA resistance nodulation cell division efflux pump. TetA(64) does not efflux minocycline, tigecycline, and eravacycline. Comprehensive screening of genome sequences showed that TetA(64) is unequally distributed in the Bcc and absent from the Bpc. It is present in some major cystic fibrosis pathogens, like Burkholderia cenocepacia, but absent from others like Burkholderia multivorans The tetR(64)-tetA(64) genes are located in a region of chromosome 1 that is highly conserved in Burkholderia sp. Because there is no evidence for transposition, the tetR(64)-tetA(64) genes may have been acquired by homologous recombination after horizontal gene transfer. Although Burkholderia species contain a resident multicomponent efflux pump that allows them to respond to tetracyclines up to a certain concentration, the acquisition of the single-component TetA(64) by some species likely provides the synergy that these bacteria need to defend against high tetracycline concentrations in niche environments.
Collapse
|
18
|
Schnetterle M, Gorgé O, Nolent F, Boughammoura A, Sarilar V, Vigier C, Guillier S, Koch L, Degand N, Ramisse V, Tichadou X, Girleanu M, Favier AL, Valade E, Biot F, Neulat-Ripoll F. Genomic and RT-qPCR analysis of trimethoprim-sulfamethoxazole and meropenem resistance in Burkholderia pseudomallei clinical isolates. PLoS Negl Trop Dis 2021; 15:e0008913. [PMID: 33592059 PMCID: PMC7909661 DOI: 10.1371/journal.pntd.0008913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/26/2021] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Melioidosis is an endemic disease in southeast Asia and northern Australia caused by the saprophytic bacteria Burkholderia pseudomallei, with a high mortality rate. The clinical presentation is multifaceted, with symptoms ranging from acute septicemia to multiple chronic abscesses. Here, we report a chronic case of melioidosis in a patient who lived in Malaysia in the 70s and was suspected of contracting tuberculosis. Approximately 40 years later, in 2014, he was diagnosed with pauci-symptomatic melioidosis during a routine examination. Four strains were isolated from a single sample. They showed divergent morphotypes and divergent antibiotic susceptibility, with some strains showing resistance to trimethoprim-sulfamethoxazole and fluoroquinolones. In 2016, clinical samples were still positive for B. pseudomallei, and only one type of strain, showing atypical resistance to meropenem, was isolated. PRINCIPAL FINDINGS We performed whole genome sequencing and RT-qPCR analysis on the strains isolated during this study to gain further insights into their differences. We thus identified two types of resistance mechanisms in these clinical strains. The first one was an adaptive and transient mechanism that disappeared during the course of laboratory sub-cultures; the second was a mutation in the efflux pump regulator amrR, associated with the overexpression of the related transporter. CONCLUSION The development of such mechanisms may have a clinical impact on antibiotic treatment. Indeed, their transient nature could lead to an undiagnosed resistance. Efflux overexpression due to mutation leads to an important multiple resistance, reducing the effectiveness of antibiotics during treatment.
Collapse
Affiliation(s)
- Marine Schnetterle
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
- Ecole du Val de Grace, Paris, France
| | - Olivier Gorgé
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Flora Nolent
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Aïda Boughammoura
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Véronique Sarilar
- Molecular Biology Unit, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Cécile Vigier
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Sophie Guillier
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Lionel Koch
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
- Ecole du Val de Grace, Paris, France
| | - Nicolas Degand
- Laboratoire de bactériologie, Hôpital de l’Archet, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Vincent Ramisse
- DGA MNRBC- Le Bouchet, Division Biologie, ABIO, Vert-le-Petit, France
| | - Xavier Tichadou
- DGA MNRBC- Le Bouchet, Division Biologie, ABIO, Vert-le-Petit, France
| | - Maria Girleanu
- Imagery Unit, Departement of plateforms and technology research, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Anne-Laure Favier
- Imagery Unit, Departement of plateforms and technology research, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Eric Valade
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
- Ecole du Val de Grace, Paris, France
| | - Fabrice Biot
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| | - Fabienne Neulat-Ripoll
- Bacteriology Unit, UMR-MD1 INSERM 1261, French Armed Biomedical Research Institut, Brétigny-sur-Orge, France
| |
Collapse
|
19
|
Grund ME, Choi SJ, McNitt DH, Barbier M, Hu G, LaSala PR, Cote CK, Berisio R, Lukomski S. Burkholderia collagen-like protein 8, Bucl8, is a unique outer membrane component of a putative tetrapartite efflux pump in Burkholderia pseudomallei and Burkholderia mallei. PLoS One 2020; 15:e0242593. [PMID: 33227031 PMCID: PMC7682875 DOI: 10.1371/journal.pone.0242593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/06/2020] [Indexed: 12/19/2022] Open
Abstract
Bacterial efflux pumps are an important pathogenicity trait because they extrude a variety of xenobiotics. Our laboratory previously identified in silico Burkholderia collagen-like protein 8 (Bucl8) in the hazardous pathogens Burkholderia pseudomallei and Burkholderia mallei. We hypothesize that Bucl8, which contains two predicted tandem outer membrane efflux pump domains, is a component of a putative efflux pump. Unique to Bucl8, as compared to other outer membrane proteins, is the presence of an extended extracellular region containing a collagen-like (CL) domain and a non-collagenous C-terminus (Ct). Molecular modeling and circular dichroism spectroscopy with a recombinant protein, corresponding to this extracellular CL-Ct portion of Bucl8, demonstrated that it adopts a collagen triple helix, whereas functional assays screening for Bucl8 ligands identified binding to fibrinogen. Bioinformatic analysis of the bucl8 gene locus revealed it resembles a classical efflux-pump operon. The bucl8 gene is co-localized with downstream fusCDE genes encoding fusaric acid (FA) resistance, and with an upstream gene, designated as fusR, encoding a LysR-type transcriptional regulator. Using reverse transcriptase (RT)-qPCR, we defined the boundaries and transcriptional organization of the fusR-bucl8-fusCDE operon. We found exogenous FA induced bucl8 transcription over 80-fold in B. pseudomallei, while deletion of the entire bucl8 locus decreased the minimum inhibitory concentration of FA 4-fold in its isogenic mutant. We furthermore showed that the putative Bucl8-associated pump expressed in the heterologous Escherichia coli host confers FA resistance. On the contrary, the Bucl8-associated pump did not confer resistance to a panel of clinically-relevant antimicrobials in Burkholderia and E. coli. We finally demonstrated that deletion of the bucl8-locus drastically affects the growth of the mutant in L-broth. We determined that Bucl8 is a component of a novel tetrapartite efflux pump, which confers FA resistance, fibrinogen binding, and optimal growth.
Collapse
Affiliation(s)
- Megan E. Grund
- Department of Microbiology, Immunology and Cell Biology, School of
Medicine, West Virginia University, Morgantown, WV, United States of
America
| | - Soo J. Choi
- Department of Microbiology, Immunology and Cell Biology, School of
Medicine, West Virginia University, Morgantown, WV, United States of
America
| | - Dudley H. McNitt
- Department of Microbiology, Immunology and Cell Biology, School of
Medicine, West Virginia University, Morgantown, WV, United States of
America
| | - Mariette Barbier
- Department of Microbiology, Immunology and Cell Biology, School of
Medicine, West Virginia University, Morgantown, WV, United States of
America
| | - Gangqing Hu
- Department of Microbiology, Immunology and Cell Biology, School of
Medicine, West Virginia University, Morgantown, WV, United States of
America
- Cancer Center, West Virginia University, Morgantown, WV, United States of
America
- Bioinformatics Core, West Virginia University, Morgantown, WV, United
States of America
| | - P. Rocco LaSala
- Department of Pathology, West Virginia University, Morgantown, WV, United
States of America
| | - Christopher K. Cote
- Bacteriology Division, The United States Army Medical Research Institute
of Infectious Diseases (USAMRIID), Frederick, MD, United States of
America
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, National Research Council,
Naples, Italy
| | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, School of
Medicine, West Virginia University, Morgantown, WV, United States of
America
- Cancer Center, West Virginia University, Morgantown, WV, United States of
America
| |
Collapse
|
20
|
Puja H, Comment G, Chassagne S, Plésiat P, Jeannot K. Coordinate overexpression of two
RND
efflux systems,
ParXY
and
TtgABC
, is responsible for multidrug resistance in
Pseudomonas putida. Environ Microbiol 2020; 22:5222-5231. [DOI: 10.1111/1462-2920.15200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Hélène Puja
- UMR 6249 Chrono‐environnement UFR Santé, Université de Bourgogne‐Franche Comté Besançon France
| | - Gwendoline Comment
- UMR 6249 Chrono‐environnement UFR Santé, Université de Bourgogne‐Franche Comté Besançon France
| | - Sophie Chassagne
- UMR 6249 Chrono‐environnement UFR Santé, Université de Bourgogne‐Franche Comté Besançon France
| | - Patrick Plésiat
- UMR 6249 Chrono‐environnement UFR Santé, Université de Bourgogne‐Franche Comté Besançon France
- Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire de Besançon Besançon France
| | - Katy Jeannot
- UMR 6249 Chrono‐environnement UFR Santé, Université de Bourgogne‐Franche Comté Besançon France
- Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire de Besançon Besançon France
| |
Collapse
|
21
|
|
22
|
Fu J, Zhong C, Zhang P, Zong G, Liu M, Cao G. Novel Mobilizable Genomic Island GEI-D18A Mediates Conjugational Transfer of Antibiotic Resistance Genes in the Multidrug-Resistant Strain Rheinheimera sp. D18. Front Microbiol 2020; 11:627. [PMID: 32318052 PMCID: PMC7155750 DOI: 10.3389/fmicb.2020.00627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/20/2020] [Indexed: 12/22/2022] Open
Abstract
Aquatic environments act as reservoirs of antimicrobial-resistant bacteria and antimicrobial resistance (AMR) genes, and the dissemination of antibiotic resistance from these environments is of increasing concern. In this study, a multidrug-resistant bacterial strain, identified as Rheinheimera sp. D18, was isolated from the sea water of an industrial maricultural system in the Yellow Sea, China. Whole-genome sequencing of D18 revealed the presence of a novel 25.8 kb antibiotic resistance island, designated GEI-D18A, which carries several antibiotic resistance genes (ARGs), including aadA1, aacA3, tetR, tet(B), catA, dfrA37, and three sul1 genes. Besides, integrase, transposase, resolvase, and recombinase encoding genes were also identified in GEI-D18A. The transferability of GEI-D18A was confirmed by mating experiments between Rheinheimera sp. D18 and Escherichia coli 25DN, and efflux pump inhibitor assays also suggested that tet(B) in GEI-D18A was responsible for tetracycline resistance in both D18 and the transconjugant. This study represents the first characterization of a mobilizable antibiotic resistance island in a species of Rheinheimera and provides evidence that Rheinheimera spp. could be important reservoirs and vehicles for ARGs in the Yellow Sea area.
Collapse
Affiliation(s)
- Jiafang Fu
- Department of Epidemiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Shandong Medicinal Biotechnology Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Chuanqing Zhong
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, China
| | - Peipei Zhang
- Department of Epidemiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Shandong Medicinal Biotechnology Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Gongli Zong
- Department of Epidemiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Key Laboratory for Biotech-Drugs of National Health Commission, Jinan, China
| | - Meng Liu
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, China
| | - Guangxiang Cao
- Department of Epidemiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Shandong Medicinal Biotechnology Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
23
|
Abstract
The causative agent of melioidosis, Burkholderia pseudomallei, a tier 1 select agent, is endemic in Southeast Asia and northern Australia, with increased incidence associated with high levels of rainfall. Increasing reports of this condition have occurred worldwide, with estimates of up to 165,000 cases and 89,000 deaths per year. The ecological niche of the organism has yet to be clearly defined, although the organism is associated with soil and water. The culture of appropriate clinical material remains the mainstay of laboratory diagnosis. Identification is best done by phenotypic methods, although mass spectrometric methods have been described. Serology has a limited diagnostic role. Direct molecular and antigen detection methods have limited availability and sensitivity. Clinical presentations of melioidosis range from acute bacteremic pneumonia to disseminated visceral abscesses and localized infections. Transmission is by direct inoculation, inhalation, or ingestion. Risk factors for melioidosis include male sex, diabetes mellitus, alcohol abuse, and immunosuppression. The organism is well adapted to intracellular survival, with numerous virulence mechanisms. Immunity likely requires innate and adaptive responses. The principles of management of this condition are drainage and debridement of infected material and appropriate antimicrobial therapy. Global mortality rates vary between 9% and 70%. Research into vaccine development is ongoing.
Collapse
Affiliation(s)
- I Gassiep
- Pathology Queensland, Townsville Hospital, Townsville, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - M Armstrong
- Pathology Queensland, Townsville Hospital, Townsville, Queensland, Australia
| | - R Norton
- Pathology Queensland, Townsville Hospital, Townsville, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
24
|
Shang Q, Gao Y, Qin T, Wang S, Shi Y, Chen T. Interaction of Oral and Toothbrush Microbiota Affects Oral Cavity Health. Front Cell Infect Microbiol 2020; 10:17. [PMID: 32117797 PMCID: PMC7011102 DOI: 10.3389/fcimb.2020.00017] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
Tooth brushing is necessary to maintain oral health. Little research has been carried out to explore microbial diversity in toothbrushes and to study the potential impact of these bacteria on human health. In the present study, 20 participants were enrolled, and the microbial diversity in their oral cavity and toothbrushes was investigated using high-throughput sequencing. Our results indicate that 1,136 and 976 operational taxonomic units (OTUs) were obtained from groups CB (samples from toothbrushes of participants using traditional Chinese medicinal toothpaste) and AB (samples from toothbrushes of those using antibacterial toothpaste), respectively. The pathogens Acinetobacter baumannii, Staphylococcus aureus, and Candida albicans were identified on toothbrushes. The presence of these pathogens increases the chance for the host to get infectious diseases, neurodegenerative diseases, cardiovascular diseases, and cancers. Moreover, our in vitro results indicate that traditional Chinese medicinal toothpaste and antibacterial toothpaste can not only inhibit the growth of pathogens but also markedly inhibit the growth of probiotics Lactobacillus salivarius and Streptococcus salivarius. Therefore, the inhibitory effect of toothpaste on probiotics, together with the existence of pathogens in toothbrushes, indicates a potential risk of tooth brushing for people in a sub-healthy state.
Collapse
Affiliation(s)
- Qingyao Shang
- The Key Laboratory of Oral Biomedicine, Department of Conservative Dentistry and Endodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang, China.,School of Stomatology, Nanchang University, Nanchang, China
| | - Yuan Gao
- The Key Laboratory of Oral Biomedicine, Department of Conservative Dentistry and Endodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang, China
| | - Ting Qin
- The Key Laboratory of Oral Biomedicine, Department of Conservative Dentistry and Endodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang, China
| | - Shuai Wang
- The Key Laboratory of Oral Biomedicine, Department of Conservative Dentistry and Endodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang, China
| | - Yan Shi
- The Key Laboratory of Oral Biomedicine, Department of Conservative Dentistry and Endodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang, China.,School of Stomatology, Nanchang University, Nanchang, China.,National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Tingtao Chen
- The Key Laboratory of Oral Biomedicine, Department of Conservative Dentistry and Endodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang, China.,National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Duplantier AJ, Shurtleff AC, Miller C, Chiang CY, Panchal RG, Sunay M. Combating biothreat pathogens: ongoing efforts for countermeasure development and unique challenges. DRUG DISCOVERY TARGETING DRUG-RESISTANT BACTERIA 2020. [PMCID: PMC7258707 DOI: 10.1016/b978-0-12-818480-6.00007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Research to discover and develop antibacterial and antiviral drugs with potent activity against pathogens of biothreat concern presents unique methodological and process-driven challenges. Herein, we review laboratory approaches for finding new antibodies, antibiotics, and antiviral molecules for pathogens of biothreat concern. Using high-throughput screening techniques, molecules that directly inhibit a pathogen’s entry, replication, or growth can be identified. Alternatively, molecules that target host proteins can be interesting targets for development when countering biothreat pathogens, due to the modulation of the host immune response or targeting proteins that interfere with the pathways required by the pathogen for replication. Monoclonal and cocktail antibody therapies approved by the Food and Drug Administration for countering anthrax and under development for treatment of Ebola virus infection are discussed. A comprehensive tabular review of current in vitro, in vivo, pharmacokinetic and efficacy datasets has been presented for biothreat pathogens of greatest concern. Finally, clinical trials and animal rule or traditional drug approval pathways are also reviewed. Opinions; interpretations; conclusions; and recommendations are those of the authors and are not necessarily endorsed by the US Army.
Collapse
|
26
|
Sarovich DS, Webb JR, Pitman MC, Viberg LT, Mayo M, Baird RW, Robson JM, Currie BJ, Price EP. Raising the Stakes: Loss of Efflux Pump Regulation Decreases Meropenem Susceptibility in Burkholderia pseudomallei. Clin Infect Dis 2019; 67:243-250. [PMID: 29394337 DOI: 10.1093/cid/ciy069] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/29/2018] [Indexed: 02/06/2023] Open
Abstract
Background Burkholderia pseudomallei, the causative agent of the high-mortality disease melioidosis, is a gram-negative bacterium that is naturally resistant to many antibiotics. There is no vaccine for melioidosis, and effective eradication is reliant on biphasic and prolonged antibiotic administration. The carbapenem drug meropenem is the current gold standard option for treating severe melioidosis. Intrinsic B. pseudomallei resistance toward meropenem has not yet been documented; however, resistance could conceivably develop over the course of infection, leading to prolonged sepsis and treatment failure. Methods We examined our 30-year clinical collection of melioidosis cases to identify B. pseudomallei isolates with reduced meropenem susceptibility. Isolates were subjected to minimum inhibitory concentration (MIC) testing toward meropenem. Paired isolates from patients who had evolved decreased susceptibility were subjected to whole-genome sequencing. Select agent-compliant genetic manipulation was carried out to confirm the molecular mechanisms conferring resistance. Results We identified 11 melioidosis cases where B. pseudomallei isolates developed decreased susceptibility toward meropenem during treatment, including 2 cases not treated with this antibiotic. Meropenem MICs increased from 0.5-0.75 µg/mL to 3-8 µg/mL. Comparative genomics identified multiple mutations affecting multidrug resistance-nodulation-division (RND) efflux pump regulators, with concomitant overexpression of their corresponding pumps. All cases were refractory to treatment despite aggressive, targeted therapy, and 2 were associated with a fatal outcome. Conclusions This study confirms the role of RND efflux pumps in decreased meropenem susceptibility in B. pseudomallei. These findings have important ramifications for the diagnosis, treatment, and management of life-threatening melioidosis cases.
Collapse
Affiliation(s)
- Derek S Sarovich
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Northern Territory.,Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland
| | - Jessica R Webb
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Northern Territory
| | - Matthew C Pitman
- Department of Territory Pathology, Royal Darwin Hospital, Tiwi, Northern Territory.,Infectious Diseases, Royal Darwin Hospital, Tiwi, Northern Territory
| | - Linda T Viberg
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Northern Territory
| | - Mark Mayo
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Northern Territory
| | - Robert W Baird
- Department of Territory Pathology, Royal Darwin Hospital, Tiwi, Northern Territory.,Infectious Diseases, Royal Darwin Hospital, Tiwi, Northern Territory
| | | | - Bart J Currie
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Northern Territory.,Infectious Diseases, Royal Darwin Hospital, Tiwi, Northern Territory.,Northern Territory Medical Program, Royal Darwin Hospital, Tiwi, Australia
| | - Erin P Price
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Northern Territory.,Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland
| |
Collapse
|
27
|
Demonstration of the broad spectrum in vitro activity of finafloxacin against pathogens of biodefence interest. Antimicrob Agents Chemother 2019:AAC.01470-19. [PMID: 31570393 PMCID: PMC6879258 DOI: 10.1128/aac.01470-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study investigated the in vitro activity of finafloxacin against bacterial strain panels of the biodefense pathogens. Broth microdilution assays were performed at neutral and acidic pH to determine the effectiveness of the antibiotics under conditions typical of an intracellular environment. In all instances, finafloxacin demonstrated superior activity at low pH. This study investigated the in vitro activity of finafloxacin against bacterial strain panels of the biodefense pathogens. Broth microdilution assays were performed at neutral and acidic pH to determine the effectiveness of the antibiotics under conditions typical of an intracellular environment. In all instances, finafloxacin demonstrated superior activity at low pH. These results highlight the importance of evaluating antimicrobial efficacy under conditions relevant to those encountered in vivo.
Collapse
|
28
|
Shearer JD, Saylor ML, Butler CM, Treston AM, Heine HS, Chirakul S, Schweizer HP, Louie A, Drusano GL, Zumbrun SD, Warfield KL. GC-072: A Novel Therapeutic Candidate for Oral Treatment of Melioidosis and Infections Caused by Select Biothreat Pathogens. Antimicrob Agents Chemother 2019; 63:AAC.00834-19. [PMID: 31548183 PMCID: PMC6879241 DOI: 10.1128/aac.00834-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/13/2019] [Indexed: 12/03/2022] Open
Abstract
Burkholderia pseudomallei (B. pseudomallei), the etiological agent of melioidosis, is a Gram-negative bacterium with additional concern as a biothreat pathogen. The mortality rate from B. pseudomallei varies depending on the type of infection and extent of available health care, but in the case of septicemia left untreated it can range from 50 - 90%. Current therapy for melioidosis is biphasic, consisting of parenteral acute-phase treatment for two weeks or longer, followed by oral eradication-phase treatment lasting several months. An effective oral therapeutic for outpatient treatment of acute-phase melioidosis is needed. GC-072 is a potent, 4-oxoquinolizine antibiotic with selective inhibitory activity against bacterial topoisomerases. GC-072 has demonstrated in vitro potency against susceptible and drug-resistant strains of B. pseudomallei and is also active against Burkholderia mallei, Bacillus anthracis, Yersinia pestis, and Francisella tularensis GC-072 is bactericidal both extra- and intracellularly, with rapid killing noted within a few hours and reduced development of resistance compared to ceftazidime. GC-072, delivered intragastrically to mimic oral administration, promoted dose-dependent survival in mice using lethal inhalational models of B. pseudomallei infection following exposure to a 24 or 339 LD50 challenge with B. pseudomallei strain 1026b. Overall, GC-072 appears to be a strong candidate for first-line, oral treatment of melioidosis.
Collapse
Affiliation(s)
| | | | | | | | - Henry S Heine
- Institute for Therapeutic Innovation, University of Florida, College of Medicine, Orlando, FL
| | - Sunisa Chirakul
- Emerging Pathogens Institute, University of Florida, College of Medicine, Gainesville, FL
| | - Herbert P Schweizer
- Institute for Therapeutic Innovation, University of Florida, College of Medicine, Orlando, FL
- Emerging Pathogens Institute, University of Florida, College of Medicine, Gainesville, FL
| | - Arnold Louie
- Institute for Therapeutic Innovation, University of Florida, College of Medicine, Orlando, FL
| | - George L Drusano
- Institute for Therapeutic Innovation, University of Florida, College of Medicine, Orlando, FL
| | - Steven D Zumbrun
- United States Army Medical Research Institute of Infectious Diseases, Frederick MD
| | | |
Collapse
|
29
|
Serio AW, Keepers T, Andrews L, Krause KM. Aminoglycoside Revival: Review of a Historically Important Class of Antimicrobials Undergoing Rejuvenation. EcoSal Plus 2018; 8. [PMID: 30447062 PMCID: PMC11575671 DOI: 10.1128/ecosalplus.esp-0002-2018] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Indexed: 01/04/2023]
Abstract
Aminoglycosides are cidal inhibitors of bacterial protein synthesis that have been utilized for the treatment of serious bacterial infections for almost 80 years. There have been approximately 15 members of this class approved worldwide for the treatment of a variety of infections, many serious and life threatening. While aminoglycoside use declined due to the introduction of other antibiotic classes such as cephalosporins, fluoroquinolones, and carbapenems, there has been a resurgence of interest in the class as multidrug-resistant pathogens have spread globally. Furthermore, aminoglycosides are recommended as part of combination therapy for empiric treatment of certain difficult-to-treat infections. The development of semisynthetic aminoglycosides designed to overcome common aminoglycoside resistance mechanisms, and the shift to once-daily dosing, has spurred renewed interest in the class. Plazomicin is the first new aminoglycoside to be approved by the FDA in nearly 40 years, marking the successful start of a new campaign to rejuvenate the class.
Collapse
|
30
|
Webb JR, Price EP, Somprasong N, Schweizer HP, Baird RW, Currie BJ, Sarovich DS. Development and validation of a triplex quantitative real-time PCR assay to detect efflux pump-mediated antibiotic resistance in Burkholderia pseudomallei. Future Microbiol 2018; 13:1403-1418. [PMID: 30256166 PMCID: PMC6190177 DOI: 10.2217/fmb-2018-0155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/23/2018] [Indexed: 01/12/2023] Open
Abstract
AIM To develop a probe-based triplex quantitative real-time PCR assay to simultaneously detect the upregulation of the efflux pumps AmrAB-OprA, BpeAB-OprB and BpeEF-OprC in Burkholderia pseudomallei strains exhibiting increased minimum inhibitory concentrations toward meropenem, doxycycline or trimethoprim-sulfamethoxazole. METHODS The triplex assay was developed and subsequently tested on RNA isolated from eight clinical and eight laboratory-generated B. pseudomallei mutants harboring efflux pump regulator mutations. RESULTS The triplex assay accurately detected efflux pump upregulation in all clinical and laboratory mutants, which corresponded with decreased antibiotic susceptibility or antibiotic resistance. CONCLUSION Rapid detection of antibiotic resistance provides clinicians with a tool to identify potential treatment failure in near real time, enabling informed alteration of treatment during an infection and improved patient outcomes.
Collapse
Affiliation(s)
- Jessica R Webb
- Global & Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Erin P Price
- Global & Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Faculty of Science, Health, Education & Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Nawarat Somprasong
- Department of Molecular Genetics & Microbiology, College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Herbert P Schweizer
- Department of Molecular Genetics & Microbiology, College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Robert W Baird
- Departments of Infectious Diseases & Pathology & Northern Territory Medical Program, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Bart J Currie
- Global & Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Departments of Infectious Diseases & Pathology & Northern Territory Medical Program, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Derek S Sarovich
- Global & Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Faculty of Science, Health, Education & Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
31
|
Rhodes KA, Somprasong N, Podnecky NL, Mima T, Chirakul S, Schweizer HP. Molecular determinants of Burkholderia pseudomallei BpeEF-OprC efflux pump expression. MICROBIOLOGY-SGM 2018; 164:1156-1167. [PMID: 30024368 DOI: 10.1099/mic.0.000691] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Burkholderia pseudomallei, the cause of melioidosis, is intrinsically resistant to many antibiotics. Acquired multidrug resistance, including resistance to doxycycline and co-trimoxazole used for melioidosis eradication phase therapy, is mainly attributed to constitutive expression of the BpeEF-OprC efflux pump. Constitutive expression of this pump is caused by mutations affecting two highly similar LysR-type transcriptional regulators (LTTR), BpeT and BpeS, but their interaction with the regulatory region governing BpeEF-OprC expression has not yet been studied. The bpeE-bpeF-oprC genes are distally located in the llpE-bpeE-bpeF-oprC operon. The llpE gene encodes a putative lipase/esterase of unknown function. We show that in a bpeT mutant llpE is constitutively co-transcribed with bpeE-bpeF-oprC. As expected from previous studies with B. cenocepacia, deletion of llpE does not affect antibiotic efflux. Using transcriptional bpeE'-lacZ fusions, we demonstrate that the 188 bp bpeT-llpE intergenic region located between bpeT and the llpE-bpeE-bpeF-oprC operon contains regulatory elements needed for control of bpeT and llpE-bpeE-bpeF-oprC operon expression. By native polyacrylamide gel electrophoresis and electrophoretic mobility shift assays with purified recombinant BpeT and BpeS proteins, we show BpeT and BpeS form oligomers that share a 14 bp binding site overlapping the essential region required for llpE-bpeE-bpeF-oprC expression. The binding site contains the conserved T-N11-A LTTR box motif involved in binding of LysR proteins, which in concert with two other possible LTTR boxes may mediate BpeT and BpeS regulation of BpeEF-OprC expression. These studies form the basis for further investigation of BpeEF-OprC expression and regulation at the molecular level by yet unknown external stimuli.
Collapse
Affiliation(s)
- Katherine A Rhodes
- 1Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,3Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.,2Department of Molecular Genetics and Microbiology, College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.,†Present address: University of Arizona BIO5 Institute, Tucson, AZ 85721, USA
| | - Nawarat Somprasong
- 1Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,2Department of Molecular Genetics and Microbiology, College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.,3Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Nicole L Podnecky
- 1Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,‡Present address: Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Tromsø, 9037 Tromsø, Norway
| | - Takehiko Mima
- 1Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,§Present address: Department of Bacteriology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Sunisa Chirakul
- 2Department of Molecular Genetics and Microbiology, College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.,3Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Herbert P Schweizer
- 3Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.,2Department of Molecular Genetics and Microbiology, College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.,1Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
32
|
Price EP, Viberg LT, Kidd TJ, Bell SC, Currie BJ, Sarovich DS. Transcriptomic analysis of longitudinal Burkholderia pseudomallei infecting the cystic fibrosis lung. Microb Genom 2018; 4. [PMID: 29989529 PMCID: PMC6159556 DOI: 10.1099/mgen.0.000194] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The melioidosis bacterium, Burkholderia pseudomallei, is increasingly being recognised as a pathogen in patients with cystic fibrosis (CF). We have recently catalogued genome-wide variation of paired, isogenic B. pseudomallei isolates from seven Australasian CF cases, which were collected between 4 and 55 months apart. Here, we extend this investigation by documenting the transcriptomic changes in B. pseudomallei in five cases. Following growth in an artificial CF sputum medium, four of the five paired isolates exhibited significant differential gene expression (DE) that affected between 32 and 792 genes. The greatest number of DE events was observed between the strains from patient CF9, consistent with the hypermutator status of the latter strain, which is deficient in the DNA mismatch repair protein MutS. Two patient isolates harboured duplications that concomitantly increased expression of the β-lactamase-encoding gene penA, and a 35 kb deletion in another abolished expression of 29 genes. Convergent expression profiles in the chronically-adapted isolates identified two significantly downregulated and 17 significantly upregulated loci, including the resistance-nodulation-division (RND) efflux pump BpeEF-OprC, the quorum-sensing hhqABCDE operon, and a cyanide- and pyocyanin-insensitive cytochrome bd quinol oxidase. These convergent pathoadaptations lead to increased expression of pathways that may suppress competing bacterial and fungal pathogens, and that enhance survival in oxygen-restricted environments, the latter of which may render conventional antibiotics less effective in vivo. Treating chronically adapted B. pseudomallei infections with antibiotics designed to target anaerobic infections, such as the nitroimidazole class of antibiotics, may significantly improve pathogen eradication attempts by exploiting this Achilles heel.
Collapse
Affiliation(s)
- Erin P Price
- 1Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia.,2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Linda T Viberg
- 2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Timothy J Kidd
- 3Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,4School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Scott C Bell
- 3Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,5QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.,6Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Bart J Currie
- 2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia.,7Department of Infectious Diseases and Northern Territory Medical Program, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Derek S Sarovich
- 1Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia.,2Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| |
Collapse
|
33
|
Study of the Role of Efflux Pumps in Amikacin-Resistant Acinetobacter Isolates from Teaching Hospitals of Mashhad, Iran. Jundishapur J Microbiol 2018. [DOI: 10.5812/jjm.12754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
34
|
Dhar S, Kumari H, Balasubramanian D, Mathee K. Cell-wall recycling and synthesis in Escherichia coli and Pseudomonas aeruginosa – their role in the development of resistance. J Med Microbiol 2018; 67:1-21. [DOI: 10.1099/jmm.0.000636] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Supurna Dhar
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Hansi Kumari
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | | | - Kalai Mathee
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
35
|
Verma A, Bhandari V, Deep DK, Sundar S, Dujardin JC, Singh R, Salotra P. Transcriptome profiling identifies genes/pathways associated with experimental resistance to paromomycin in Leishmania donovani. Int J Parasitol Drugs Drug Resist 2017; 7:370-377. [PMID: 29035735 PMCID: PMC5645162 DOI: 10.1016/j.ijpddr.2017.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 01/19/2023]
Abstract
Widespread resistance towards antimony and reports of relapses following miltefosine treatment has severely affected the management of visceral leishmaniasis (VL) in the Indian subcontinent. Paromomycin (PMM), an aminoglycoside antibiotic, has been licensed for VL treatment in India in 2007. Although its use is still restricted in the field, unraveling the molecular mechanism of resistance towards PMM is the key to preserve the drug. In this study, PMM resistant lines were selected up to 100 μM of PMM in three distinct field isolates of Leishmania donovani at promastigote stage. The resistance induced at promastigote level was also evident in amastigotes which showed 6 fold decreases in PMM susceptibility. Comparative transcriptome profiling of PMM resistant (PMM-R) and the corresponding PMM sensitive (PMM-S) parasites revealed modulated expression of 500 genes (1.5 fold cut off) in PMM-R parasites. Selected genes were validated for their modulated expression by quantitative real-time PCR. Functional classification and pathway analysis of modulated genes indicated probable adaptations in drug resistant lines which included a) reduced oxidative phosphorylation; b) increased glycosomal succinate fermentation and substrate level phosphorylation; c) dependency on lipids and amino acids for energy generation; d) reduced DNA synthesis and increased DNA damage repair and e) decreased protein synthesis and degradation. Interestingly, PMM-R parasites showed a marked increase in PMM susceptibility in presence of verapamil and amlodipine, antagonists of Ca2+ channel that are also modulators of ABC transporters. Moreover, infection of macrophages by PMM-R parasites led to modulated nitric oxide (NO) levels while reactive oxygen species (ROS) level remained unaltered. The present study highlights the putative mechanisms of PMM resistance in Leishmania.
Collapse
Affiliation(s)
- Aditya Verma
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India; Symbiosis School of Biomedical Sciences, Symbiosis International University, Pune, India
| | - Vasundhra Bhandari
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Deepak Kumar Deep
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jean Claude Dujardin
- Unit of Molecular Parasitology, Department of Parasitology, Institute of Tropical Medicine, Antwerp, Belgium
| | - Ruchi Singh
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Poonam Salotra
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.
| |
Collapse
|
36
|
Sandhu P, Akhter Y. Evolution of structural fitness and multifunctional aspects of mycobacterial RND family transporters. Arch Microbiol 2017; 200:19-31. [PMID: 28951954 DOI: 10.1007/s00203-017-1434-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/07/2017] [Accepted: 09/20/2017] [Indexed: 12/11/2022]
Abstract
Drug resistance is a major concern due to the evolution and emergence of pathogenic bacterial strains with novel strategies to resist the antibiotics in use. Mycobacterium tuberculosis (Mtb) is one of such pathogens with reported strains, which are not treatable with any of the available anti-TB drugs. This scenario has led to the need to look for some novel drug targets in Mtb, which may be exploited to design effective treatment strategies against the infection. The goal of this review is to discuss one such class of emerging drug targets in Mtb. MmpL (mycobacterial membrane protein large) proteins from Mtb are reported to be involved in multi-substrate transport including drug efflux and considered as one of the contributing factors for the emergence of multidrug-resistant strains. MmpL proteins belong to resistance nodulation division permeases superfamily of membrane transporters, which are viably and pathogenetically important and their inhibition could be lethal for the bacteria.
Collapse
Affiliation(s)
- Padmani Sandhu
- Structural Bioinformatics Group, Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Shahpur District, Kangra, Himachal Pradesh, 176206, India
| | - Yusuf Akhter
- Structural Bioinformatics Group, Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Shahpur District, Kangra, Himachal Pradesh, 176206, India.
| |
Collapse
|
37
|
Relationships Between Resistance and Virulence in Burkholderia pseudomallei. CURRENT TROPICAL MEDICINE REPORTS 2017. [DOI: 10.1007/s40475-017-0119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
O’Rourke A, Yee N, Nierman WC, Beyhan S. Environmental and Genetic Factors Controlling Burkholderia pseudomallei Persister Phenotypes. CURRENT TROPICAL MEDICINE REPORTS 2017. [DOI: 10.1007/s40475-017-0116-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
39
|
Role of AxyZ Transcriptional Regulator in Overproduction of AxyXY-OprZ Multidrug Efflux System in Achromobacter Species Mutants Selected by Tobramycin. Antimicrob Agents Chemother 2017; 61:AAC.00290-17. [PMID: 28584156 DOI: 10.1128/aac.00290-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/31/2017] [Indexed: 11/20/2022] Open
Abstract
AxyXY-OprZ is an RND-type efflux system that confers innate aminoglycoside resistance to Achromobacter spp. We investigated here a putative TetR family transcriptional regulator encoded by the axyZ gene located upstream of axyXY-oprZ An in-frame axyZ gene deletion assay led to increased MICs of antibiotic substrates of the efflux system, including aminoglycosides, cefepime, fluoroquinolones, tetracyclines, and erythromycin, indicating that the product of axyZ negatively regulates expression of axyXY-oprZ Moreover, we identified an amino acid substitution at position 29 of AxyZ (V29G) in a clinical Achromobacter strain that occurred during the course of chronic respiratory tract colonization in a cystic fibrosis (CF) patient. This substitution, also detected in three other strains exposed in vitro to tobramycin, led to an increase in the axyY transcription level (5- to 17-fold) together with an increase in antibiotic resistance level. This overproduction of AxyXY-OprZ is the first description of antibiotic resistance acquisition due to modification of a chromosomally encoded mechanism in Achromobacter and might have an impact on the management of infected CF patients. Indeed, tobramycin is widely used for aerosol therapy within this population, and we have demonstrated that it easily selects mutants with increased MICs of not only aminoglycosides but also fluoroquinolones, cefepime, and tetracyclines.
Collapse
|
40
|
Dutta S, Haq S, Hasan MR, Haq JA. Antimicrobial susceptibility pattern of clinical isolates of Burkholderia pseudomallei in Bangladesh. BMC Res Notes 2017; 10:299. [PMID: 28728591 PMCID: PMC5520358 DOI: 10.1186/s13104-017-2626-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/13/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Melioidosis an infectious disease, caused by a Gram negative bacterium called Burkholderia pseudomallei, is endemic in Bangladesh. This organism is sensitive to limited number of antimicrobial agents and need prolonged treatment. There is no comprehensive data on the antimicrobial susceptibility profile of B. pseudomallei isolated in Bangladesh over last several years. The present study aimed to determine the antimicrobial susceptibility pattern of B. pseudomallei isolated in a tertiary care hospital of Dhaka city from 2009 to 2015. METHODS All B. pseudomallei isolated from melioidosis patients over a period of 7 years (2009-2015) in the Department of Microbiology of a 725-bed tertiary care referral hospital in Dhaka city, Bangladesh were included in the study. B. pseudomallei was identified by Gram stain, culture, specific biochemical tests, serology and PCR using specific primers constructed from 16s rRNA region of B. pseudomallei. Antimicrobial susceptibility to specific agents was determined by disk diffusion and minimum inhibitory concentration methods. RESULTS A total of 20 isolates of B. pseudomallei which were isolated from patients coming from different geographic locations of Bangladesh were included in the study. All the isolates were uniformly sensitive (100%) to ceftazidime, imipenem, piperacillin-tazobactam, amoxicillin-clavulanic acid and tetracycline by both disk diffusion and MIC methods. Two strains were resistant to trimethoprim-sulfamethoxazole by disk diffusion method but were sensitive by MIC method. The MIC50 and MIC90 values of the above antimicrobial agents were almost similar. All the isolates were resistant to amikacin by both MIC and disk diffusion methods. CONCLUSION The results of the study suggest that B. pseudomallei prevalent in Bangladesh were still susceptible to all recommended antimicrobial agents used for the treatment of melioidosis. However, regular monitoring is needed to detect any emergence of resistance and shifting of MIC50 and MIC90 values.
Collapse
Affiliation(s)
- Subarna Dutta
- Department of Microbiology, Bangladesh Institute of Research and Rehabilitation in Diabetes, Endocrine and Metabolic Disorders, 122 Kazi Nazrul Islam Avenue, Dhaka, 1000 Bangladesh
| | - Sabah Haq
- Department of Microbiology, Ibrahim Medical College, 122 Kazi Nazrul Islam Avenue, Dhaka, 1000 Bangladesh
| | - Mohammad Rokibul Hasan
- Department of Microbiology, Bangladesh Institute of Research and Rehabilitation in Diabetes, Endocrine and Metabolic Disorders, 122 Kazi Nazrul Islam Avenue, Dhaka, 1000 Bangladesh
| | - Jalaluddin Ashraful Haq
- Department of Microbiology, Ibrahim Medical College, 122 Kazi Nazrul Islam Avenue, Dhaka, 1000 Bangladesh
| |
Collapse
|
41
|
Micheva-Viteva SN, Shou Y, Ganguly K, Wu TH, Hong-Geller E. PKC-η-MARCKS Signaling Promotes Intracellular Survival of Unopsonized Burkholderia thailandensis. Front Cell Infect Microbiol 2017. [PMID: 28638804 PMCID: PMC5461351 DOI: 10.3389/fcimb.2017.00231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pathogenic Burkholderia rely on host factors for efficient intracellular replication and are highly refractory to antibiotic treatment. To identify host genes that are required by Burkholderia spp. during infection, we performed a RNA interference (RNAi) screen of the human kinome and identified 35 host kinases that facilitated Burkholderia thailandensis intracellular survival in human monocytic THP-1 cells. We validated a selection of host kinases using imaging flow cytometry to assess efficiency of B. thailandensis survival in the host upon siRNA-mediated knockdown. We focused on the role of the novel protein kinase C isoform, PKC-η, in Burkholderia infection and characterized PKC-η/MARCKS signaling as a key event that promotes the survival of unopsonized B. thailandensis CDC2721121 within host cells. While infection of lung epithelial cells with unopsonized Gram-negative bacteria stimulated phosphorylation of Ser175/160 in the MARCKS effector domain, siRNA-mediated knockdown of PKC-η expression reduced the levels of phosphorylated MARCKS by >3-fold in response to infection with Bt CDC2721121. We compared the effect of the conventional PKC-α and novel PKC-η isoforms on the growth of B. thailandensis CDC2721121 within monocytic THP-1 cells and found that ≥75% knock-down of PRKCH transcript levels reduced intracellular bacterial load 100% more efficiently when compared to growth in cells siRNA-depleted of the classical PKC-α, suggesting that the PKC-η isoform can specifically mediate Burkholderia intracellular survival. Based on imaging studies of intracellular B. thailandensis, we found that PKC-η function stimulates phagocytic pathways that promote B. thailandensis escape into the cytoplasm leading to activation of autophagosome flux. Identification of host kinases that are targeted by Burkholderia during infection provides valuable molecular insights in understanding Burkholderia pathogenesis, and ultimately, in designing effective host-targeted therapies against infectious disease caused by intracellular pathogens.
Collapse
Affiliation(s)
| | - Yulin Shou
- Bioscience Division, Los Alamos National LaboratoryLos Alamos, NM, United States
| | - Kumkum Ganguly
- Bioscience Division, Los Alamos National LaboratoryLos Alamos, NM, United States
| | - Terry H Wu
- Center for Infectious Disease and Immunity and Department of Internal Medicine, University of New Mexico Health Sciences CenterAlbuquerque, NM, United States
| | | |
Collapse
|
42
|
Bugrysheva JV, Sue D, Gee JE, Elrod MG, Hoffmaster AR, Randall LB, Chirakul S, Tuanyok A, Schweizer HP, Weigel LM. Antibiotic Resistance Markers in Burkholderia pseudomallei Strain Bp1651 Identified by Genome Sequence Analysis. Antimicrob Agents Chemother 2017; 61:e00010-17. [PMID: 28396541 PMCID: PMC5444168 DOI: 10.1128/aac.00010-17] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 03/31/2017] [Indexed: 12/30/2022] Open
Abstract
Burkholderia pseudomallei Bp1651 is resistant to several classes of antibiotics that are usually effective for treatment of melioidosis, including tetracyclines, sulfonamides, and β-lactams such as penicillins (amoxicillin-clavulanic acid), cephalosporins (ceftazidime), and carbapenems (imipenem and meropenem). We sequenced, assembled, and annotated the Bp1651 genome and analyzed the sequence using comparative genomic analyses with susceptible strains, keyword searches of the annotation, publicly available antimicrobial resistance prediction tools, and published reports. More than 100 genes in the Bp1651 sequence were identified as potentially contributing to antimicrobial resistance. Most notably, we identified three previously uncharacterized point mutations in penA, which codes for a class A β-lactamase and was previously implicated in resistance to β-lactam antibiotics. The mutations result in amino acid changes T147A, D240G, and V261I. When individually introduced into select agent-excluded B. pseudomallei strain Bp82, D240G was found to contribute to ceftazidime resistance and T147A contributed to amoxicillin-clavulanic acid and imipenem resistance. This study provides the first evidence that mutations in penA may alter susceptibility to carbapenems in B. pseudomallei Another mutation of interest was a point mutation affecting the dihydrofolate reductase gene folA, which likely explains the trimethoprim resistance of this strain. Bp1651 was susceptible to aminoglycosides likely because of a frameshift in the amrB gene, the transporter subunit of the AmrAB-OprA efflux pump. These findings expand the role of penA to include resistance to carbapenems and may assist in the development of molecular diagnostics that predict antimicrobial resistance and provide guidance for treatment of melioidosis.
Collapse
Affiliation(s)
| | - David Sue
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jay E Gee
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mindy G Elrod
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Linnell B Randall
- Department of Molecular Genetics and Microbiology, Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Sunisa Chirakul
- Department of Molecular Genetics and Microbiology, Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Apichai Tuanyok
- Department of Molecular Genetics and Microbiology, Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Herbert P Schweizer
- Department of Molecular Genetics and Microbiology, Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Linda M Weigel
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
43
|
Loss of Methyltransferase Function and Increased Efflux Activity Leads to Doxycycline Resistance in Burkholderia pseudomallei. Antimicrob Agents Chemother 2017; 61:AAC.00268-17. [PMID: 28348161 DOI: 10.1128/aac.00268-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/18/2017] [Indexed: 01/15/2023] Open
Abstract
The soil-dwelling bacterium Burkholderia pseudomallei is the causative agent of the potentially fatal disease melioidosis. The lack of a vaccine toward B. pseudomallei means that melioidosis treatment relies on prolonged antibiotic therapy, which can last up to 6 months in duration or longer. Due to intrinsic resistance, few antibiotics are effective against B. pseudomallei The lengthy treatment regimen required increases the likelihood of resistance development, with subsequent potentially fatal relapse. Doxycycline (DOX) has historically played an important role in the eradication phase of melioidosis treatment. Both primary and acquired DOX resistances have been documented in B. pseudomallei; however, the molecular mechanisms underpinning DOX resistance have remained elusive. Here, we identify and functionally characterize the molecular mechanisms conferring acquired DOX resistance in an isogenic B. pseudomallei pair. Two synergistic mechanisms were identified. The first mutation occurred in a putative S-adenosyl-l-methionine-dependent methyltransferase (encoded by BPSL3085), which we propose leads to altered ribosomal methylation, thereby decreasing DOX binding efficiency. The second mutation altered the function of the efflux pump repressor gene, amrR, resulting in increased expression of the resistance-nodulation-division efflux pump, AmrAB-OprA. Our findings highlight the diverse mechanisms by which B. pseudomallei can become resistant to antibiotics used in melioidosis therapy and the need for resistance monitoring during treatment regimens, especially in patients with prolonged or recrudesced positive cultures for B. pseudomallei.
Collapse
|
44
|
Viberg LT, Sarovich DS, Kidd TJ, Geake JB, Bell SC, Currie BJ, Price EP. Within-Host Evolution of Burkholderia pseudomallei during Chronic Infection of Seven Australasian Cystic Fibrosis Patients. mBio 2017; 8:e00356-17. [PMID: 28400528 PMCID: PMC5388805 DOI: 10.1128/mbio.00356-17] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/15/2017] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder characterized by progressive lung function decline. CF patients are at an increased risk of respiratory infections, including those by the environmental bacterium Burkholderia pseudomallei, the causative agent of melioidosis. Here, we compared the genomes of B. pseudomallei isolates collected between ~4 and 55 months apart from seven chronically infected CF patients. Overall, the B. pseudomallei strains showed evolutionary patterns similar to those of other chronic infections, including emergence of antibiotic resistance, genome reduction, and deleterious mutations in genes involved in virulence, metabolism, environmental survival, and cell wall components. We documented the first reported B. pseudomallei hypermutators, which were likely caused by defective MutS. Further, our study identified both known and novel molecular mechanisms conferring resistance to three of the five clinically important antibiotics for melioidosis treatment. Our report highlights the exquisite adaptability of microorganisms to long-term persistence in their environment and the ongoing challenges of antibiotic treatment in eradicating pathogens in the CF lung. Convergent evolution with other CF pathogens hints at a degree of predictability in bacterial evolution in the CF lung and potential targeted eradication of chronic CF infections in the future.IMPORTANCEBurkholderia pseudomallei, the causative agent of melioidosis, is an environmental opportunistic bacterium that typically infects immunocompromised people and those with certain risk factors such as cystic fibrosis (CF). Patients with CF tend to develop chronic melioidosis infections, for reasons that are not well understood. This report is the first to describe B. pseudomallei evolution within the CF lung during chronic infection. We show that the pathways by which B. pseudomallei adapts to the CF lung are similar to those seen in better-studied CF pathogens such as Pseudomonas aeruginosa, Staphylococcus aureus, and Burkholderia cepacia complex species. Adaptations include the accumulation of antibiotic resistance, loss of nonessential genes, metabolic alterations, and virulence factor attenuation. Known and novel mechanisms of resistance to three of the five antibiotics used in melioidosis treatment were identified. Similar pathways of evolution in CF pathogens, including B. pseudomallei, provide exciting avenues for more-targeted treatment of chronic, recalcitrant infections.
Collapse
Affiliation(s)
- Linda T Viberg
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Derek S Sarovich
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Centre for Animal Health Innovation, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Timothy J Kidd
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - James B Geake
- Department of Respiratory Medicine, The Lyell McEwin Hospital, Elizabeth Vale, South Australia, Australia
| | - Scott C Bell
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Bart J Currie
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Infectious Diseases and Northern Territory Medical Program, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Erin P Price
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Centre for Animal Health Innovation, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
45
|
Bond TEH, Sorenson AE, Schaeffer PM. Functional characterisation of Burkholderia pseudomallei biotin protein ligase: A toolkit for anti-melioidosis drug development. Microbiol Res 2017; 199:40-48. [PMID: 28454708 DOI: 10.1016/j.micres.2017.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/06/2017] [Accepted: 03/15/2017] [Indexed: 01/17/2023]
Abstract
Burkholderia pseudomallei (Bp) is the causative agent of melioidosis. The bacterium is responsible for 20% of community-acquired sepsis cases and 40% of sepsis-related mortalities in northeast Thailand, and is intrinsically resistant to aminoglycosides, macrolides, rifamycins, cephalosporins, and nonureidopenicillins. There is no vaccine and its diagnosis is problematic. Biotin protein ligase (BirA) which is essential for fatty acid synthesis has been proposed as a drug target in bacteria. Very few bacterial BirA have been characterized, and a better understanding of these enzymes is necessary to further assess their value as drug targets. BirA within the Burkholderia genus have not yet been investigated. We present for the first time the cloning, expression, purification and functional characterisation of the putative Bp BirA and orthologous B. thailandensis (Bt) biotin carboxyl carrier protein (BCCP) substrate. A GFP-tagged Bp BirA was produced and applied for the development of a high-throughput (HT) assay based on our differential scanning fluorimetry of GFP-tagged proteins (DSF-GTP) principle as well as an electrophoretic mobility shift assay. Our biochemical data in combination with the new HT DSF-GTP and biotinylation activity assay could facilitate future drug screening efforts against this drug-resistant organism.
Collapse
Affiliation(s)
- Thomas E H Bond
- Comparative Genomics Centre, James Cook University, DB21, James Cook Drive, Townsville, QLD 4811, Australia
| | - Alanna E Sorenson
- Comparative Genomics Centre, James Cook University, DB21, James Cook Drive, Townsville, QLD 4811, Australia
| | - Patrick M Schaeffer
- Comparative Genomics Centre, James Cook University, DB21, James Cook Drive, Townsville, QLD 4811, Australia.
| |
Collapse
|
46
|
Spengler G, Kincses A, Gajdács M, Amaral L. New Roads Leading to Old Destinations: Efflux Pumps as Targets to Reverse Multidrug Resistance in Bacteria. Molecules 2017; 22:molecules22030468. [PMID: 28294992 PMCID: PMC6155429 DOI: 10.3390/molecules22030468] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 01/05/2023] Open
Abstract
Multidrug resistance (MDR) has appeared in response to selective pressures resulting from the incorrect use of antibiotics and other antimicrobials. This inappropriate application and mismanagement of antibiotics have led to serious problems in the therapy of infectious diseases. Bacteria can develop resistance by various mechanisms and one of the most important factors resulting in MDR is efflux pump-mediated resistance. Because of the importance of the efflux-related multidrug resistance the development of new therapeutic approaches aiming to inhibit bacterial efflux pumps is a promising way to combat bacteria having over-expressed MDR efflux systems. The definition of an efflux pump inhibitor (EPI) includes the ability to render the bacterium increasingly more sensitive to a given antibiotic or even reverse the multidrug resistant phenotype. In the recent years numerous EPIs have been developed, although so far their clinical application has not yet been achieved due to their in vivo toxicity and side effects. In this review, we aim to give a short overview of efflux mediated resistance in bacteria, EPI compounds of plant and synthetic origin, and the possible methods to investigate and screen EPI compounds in bacterial systems.
Collapse
Affiliation(s)
- Gabriella Spengler
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
| | - Annamária Kincses
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
| | - Márió Gajdács
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
| | - Leonard Amaral
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
- Travel Medicine, Institute of Hygiene and Tropical Medicine, Universidade Nova de Lisboa, 1349-008 Lisbon, Portugal.
| |
Collapse
|
47
|
Sidrim JJC, Vasconcelos DC, Riello GB, Guedes GMDM, Serpa R, Bandeira TDJPG, Monteiro AJ, Cordeiro RDA, Castelo-Branco DDSCM, Rocha MFG, Brilhante RSN. Promethazine improves antibiotic efficacy and disrupts biofilms of Burkholderia pseudomallei. BIOFOULING 2017; 33:88-97. [PMID: 27936915 DOI: 10.1080/08927014.2016.1262846] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/11/2016] [Indexed: 06/06/2023]
Abstract
Efflux pumps are important defense mechanisms against antimicrobial drugs and maintenance of Burkholderia pseudomallei biofilms. This study evaluated the effect of the efflux pump inhibitor promethazine on the structure and antimicrobial susceptibility of B. pseudomallei biofilms. Susceptibility of planktonic cells and biofilms to promethazine alone and combined with antimicrobials was assessed by the broth microdilution test and biofilm metabolic activity was determined with resazurin. The effect of promethazine on 48 h-grown biofilms was also evaluated through confocal and electronic microscopy. The minimum inhibitory concentration (MIC) of promethazine was 780 mg l-1, while the minimum biofilm elimination concentration (MBEC) was 780-3,120 mg l-1. Promethazine reduced the MIC values for erythromycin, trimethoprim/sulfamethoxazole, gentamicin and ciprofloxacin and reduced the MBEC values for all tested drugs (p<0.05). Microscopic analyses demonstrated that promethazine altered the biofilm structure of B. pseudomallei, even at subinhibitory concentrations, possibly facilitating antibiotic penetration. Promethazine improves antibiotics efficacy against B. pseudomallei biofilms, by disrupting biofilm structure.
Collapse
Affiliation(s)
| | | | | | | | - Rosana Serpa
- a Specialized Center of Medical Micology , Federal University of Ceará , Fortaleza , Brazil
| | | | - André Jalles Monteiro
- b Department of Statistics and Applied Mathematics , Federal University of Ceará , Fortaleza , Brazil
| | | | | | - Marcos Fábio Gadelha Rocha
- a Specialized Center of Medical Micology , Federal University of Ceará , Fortaleza , Brazil
- c Postgraduate Program in Veterinary Science , State University of Ceará , Fortaleza , Brazil
| | | |
Collapse
|
48
|
Phenotypic and genomic characterization of the antimicrobial producer Rheinheimera sp. EpRS3 isolated from the medicinal plant Echinacea purpurea: insights into its biotechnological relevance. Res Microbiol 2016; 168:293-305. [PMID: 27884784 DOI: 10.1016/j.resmic.2016.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 12/23/2022]
Abstract
In recent years, there has been increasing interest in plant microbiota; however, despite medicinal plant relevance, very little is known about their highly complex endophytic communities. In this work, we report on the genomic and phenotypic characterization of the antimicrobial compound producer Rheinheimera sp. EpRS3, a bacterial strain isolated from the rhizospheric soil of the medicinal plant Echinacea purpurea. In particular, EpRS3 is able to inhibit growth of different bacterial pathogens (Bcc, Acinetobacter baumannii, and Klebsiella pneumoniae) which might be related to the presence of gene clusters involved in the biosynthesis of different types of secondary metabolites. The outcomes presented in this work highlight the fact that the strain possesses huge biotechnological potential; indeed, it also shows antimicrobial effects upon well-described multidrug-resistant (MDR) human pathogens, and it affects plant root elongation and morphology, mimicking indole acetic acid (IAA) action.
Collapse
|
49
|
Burkholderia pseudomallei resistance to antibiotics in biofilm-induced conditions is related to efflux pumps. J Med Microbiol 2016; 65:1296-1306. [DOI: 10.1099/jmm.0.000358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
50
|
Lehman SS, Mladinich KM, Boonyakanog A, Mima T, Karkhoff-Schweizer RR, Schweizer HP. Versatile nourseothricin and streptomycin/spectinomycin resistance gene cassettes and their use in chromosome integration vectors. J Microbiol Methods 2016; 129:8-13. [PMID: 27457407 PMCID: PMC5018448 DOI: 10.1016/j.mimet.2016.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 11/24/2022]
Abstract
An obstacle for the development of genetic systems for many bacteria is the limited number of antibiotic selection markers, especially for bacteria that are intrinsically antibiotic resistant or where utilization of such markers is strictly regulated. Here we describe the development of versatile cassettes containing nourseothricin, streptomycin/spectinomycin, and spectinomycin selection markers. The antibiotic resistance genes contained on these cassettes are flanked by loxP sites with allow their in vivo excision from the chromosome of target bacteria using Cre recombinase. The respective selection marker cassettes were used to derive mini-Tn7 elements that can be used for single-copy insertion of genes into bacterial chromosomes. The utility of the selection markers was tested by insertion of the resulting mini-Tn7 elements into the genomes of Burkholderia thailandensis and B. pseudomallei efflux pump mutants susceptible to aminoglycosides, aminocyclitols, and streptothricins, followed by Cre-mediated antibiotic resistance marker excision. The versatile nourseothricin, streptomycin/spectinomycin and spectinomycin resistance loxP cassette vectors described here extend the repertoire of antibiotic selection markers for genetic manipulation of diverse bacteria that are susceptible to aminoglycosides and aminocyclitols.
Collapse
Affiliation(s)
- Stephanie S. Lehman
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Katherine M. Mladinich
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Angkana Boonyakanog
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Takehiko Mima
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Herbert P. Schweizer
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
- Department of Molecular Genetics and Microbiology, Emerging Pathogens Institute, University of Florida, Gainesville, FL 30610, USA
| |
Collapse
|