1
|
He H, Liou SY, Shimabuku KK, Zhou P, Choi Y, Meschke JS, Roberts MC, Lee Y, Dodd MC. Degradation and Deactivation of Intracellular Bacterial Antibiotic Resistance Genes by Commonly Used Healthcare and Personal Care Disinfectants. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:8317-8328. [PMID: 40257592 DOI: 10.1021/acs.est.4c10223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
This work investigated efficacies of commonly used healthcare and personal care disinfectants, including glutaraldehyde, chlorhexidine, ethanol, povidone-iodine, benzalkonium chloride, phenol, free chlorine, hydrogen peroxide (H2O2), and 254 nm UV light, in degrading (as measured by qPCR analyses of ∼1000 bp amplicon loss) and deactivating (as measured by transforming activity loss) bacterial antibiotic resistance genes (ARGs) during inactivation of antibiotic-resistant bacteria (ARB) on inanimate surfaces or in aqueous suspension. Intracellular ARGs (iARGs) blt, mecA, and ampC, within vegetative cells of Bacillus subtilis, Staphylococcus aureus, and Pseudomonas aeruginosa, respectively, were treated on PTFE and/or stainless-steel surfaces or in aqueous phosphate buffer (PB; H2O2 only), to simulate potential healthcare and personal care cleaning applications under representative disinfectant exposure conditions. No chemical disinfectant yielded more than limited (≤1.9log10) iARG degradation/deactivation under the conditions investigated, even when ARB cells were extensively inactivated (at levels from 3.1log10 to ≥6log10). In contrast, UV irradiation yielded up to ∼2.8-3.2log10 iARG degradation/deactivation at corresponding ARB inactivation levels up to ∼4log10 in the case of the blt gene within B. subtilis cells on PTFE surfaces, though levels of iARG degradation/deactivation and ARB inactivation were generally lower than expected based on prior aqueous-phase results, likely due to light-shielding effects at the typical ∼108-109 CFU/mL cell inoculum densities used for surface disinfection tests. During exposure to H2O2 in PB, iARG deactivation and ARB inactivation reached up to 1.7log10 and >3.5log10, respectively, while iARG degradation was minimal (≤0.2log10); this appears to be driven by DNA-strand fragmentation (as observed by pulsed-field gel electrophoresis analysis) likely resulting from reaction with endogenous HO• (or Fe(IV)) generated via intracellular iron-catalyzed H2O2 decomposition. While all investigated disinfectants were able to effectively inactivate ARB cells themselves, these results demonstrate that most are ineffective in simultaneously degrading and deactivating iARGs, highlighting the potential benefits of employing disinfectants such as 254 nm UV light, that selectively target bacterial DNA, to improve mitigation of antibiotic resistance dissemination.
Collapse
Affiliation(s)
- Huan He
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, P.R. China
- Department of Civil and Environmental Engineering, University of Washington, Box 352700, Seattle, Washington 98195, United States
| | - Sin-Yi Liou
- Department of Civil and Environmental Engineering, University of Washington, Box 352700, Seattle, Washington 98195, United States
| | - Kyle K Shimabuku
- Department of Civil and Environmental Engineering, University of Washington, Box 352700, Seattle, Washington 98195, United States
| | - Peiran Zhou
- Department of Civil and Environmental Engineering, University of Washington, Box 352700, Seattle, Washington 98195, United States
| | - Yegyun Choi
- School of Environment and Energy Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - John S Meschke
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105, United States
| | - Marilyn C Roberts
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105, United States
| | - Yunho Lee
- School of Environment and Energy Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Michael C Dodd
- Department of Civil and Environmental Engineering, University of Washington, Box 352700, Seattle, Washington 98195, United States
| |
Collapse
|
2
|
Kerek Á, Román I, Szabó Á, Pézsa NP, Jerzsele Á. Antibiotic Resistance Gene Expression in Veterinary Probiotics: Two Sides of the Coin. Vet Sci 2025; 12:217. [PMID: 40266902 PMCID: PMC11945515 DOI: 10.3390/vetsci12030217] [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: 01/26/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 04/25/2025] Open
Abstract
The rapid proliferation of antimicrobial resistance has emerged as one of the most pressing animal and public health challenges of our time. Probiotics, extensively employed in human and veterinary medicine, are instrumental in maintaining a balanced microbiome and mitigating its disruption during antibiotic therapy. While their numerous benefits are well documented, probiotics also present potential risks, notably the capacity to harbor antimicrobial resistance genes. This genetic reservoir could contribute to the emergence and spread of antimicrobial resistance by facilitating the horizontal transfer of resistance genes to pathogenic bacteria within the gut. This review critically examines the presence of antimicrobial resistance genes in commonly used probiotic strains, explores the underlying mechanisms of resistance, and provides a balanced analysis of the benefits and risks associated with their use. By addressing these dual aspects, this paper highlights the need for vigilant evaluation of probiotics to preserve their therapeutic potential while minimizing public health risks.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - István Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Nikolett Palkovicsné Pézsa
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| |
Collapse
|
3
|
Mayer-Harnisch CE, Figueroa Paniagua D, Maltseva N, Kim Y, Le VTB, Joachimiak A, Kuhn ML. N-terminal domain swapping: A new paradigm for spermidine/spermine N-acetyltransferase (SSAT) protein structures? Biochem Biophys Res Commun 2025; 748:151302. [PMID: 39823891 PMCID: PMC11808394 DOI: 10.1016/j.bbrc.2025.151302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/24/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
Enterococcus faecalis is a multi-drug-resistant human pathogen that is found in a variety of environments and is challenging to treat. Under stress conditions, some bacteria regulate intracellular polyamine concentrations via polyamine acetyltransferases to reduce their toxicity. The E. faecalis genome encodes two polyamine acetyltransferases: PmvE and BltD. Both of these proteins belong to the Gcn5-related N-acetyltransferase (GNAT) superfamily. It is unclear why there are two enzymes with similar substrate specificities in this organism. To better understand the structure/function relationship of the E. faecalis BltD enzyme, we determined its crystal structure and performed additional assays to explore its oligomeric state and enzymatic activity. The goal was to determine whether there were structural or catalytic differences between this enzyme and other polyamine acetyltransferases that could explain this redundancy and be exploited for future development of targeted inhibitors for this important human pathogen. We found the BltD enzyme was structurally unique due to its N-terminal domain swapped dimer. However, this enzyme adopts a catalytically active monomer rather than dimer in solution. This indicates the crystal structure we obtained may represent a state that forms at high protein and salt concentrations and at low pH used during crystallization. The BltD dimer found in the crystal may represent a unique view of how an inhibitory peptide or molecule could be designed to occupy its active site. Additionally, this structure shows the extensive flexibility of the N-terminal portion of the E. faecalis BltD enzyme.
Collapse
Affiliation(s)
- Claudia E Mayer-Harnisch
- San Francisco State University, Department of Chemistry and Biochemistry, San Francisco, CA, 94132, USA
| | - Daniel Figueroa Paniagua
- San Francisco State University, Department of Chemistry and Biochemistry, San Francisco, CA, 94132, USA
| | - Natalia Maltseva
- Center for Structural Biology of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL, 60667, USA; Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Youngchang Kim
- Center for Structural Biology of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL, 60667, USA; Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Van Thi Bich Le
- San Francisco State University, Department of Chemistry and Biochemistry, San Francisco, CA, 94132, USA
| | - Andrzej Joachimiak
- Center for Structural Biology of Infectious Diseases, Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL, 60667, USA; Structural Biology Center, X-ray Science Division, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Misty L Kuhn
- San Francisco State University, Department of Chemistry and Biochemistry, San Francisco, CA, 94132, USA.
| |
Collapse
|
4
|
Sun H, Levenfors JJ, Brandt C, Schnürer A. Assessing phenotypic and genotypic antibiotic resistance in bacillus-related bacteria isolated from biogas digestates. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117859. [PMID: 39947064 DOI: 10.1016/j.ecoenv.2025.117859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/18/2025] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
Antibiotic resistance poses a significant public health challenge, with biogas digestate, a byproduct of anaerobic digestion (AD), presenting potential risks when applied as a biofertilizer. Understanding the actual resistance levels in digestate is crucial for its safe application. While many studies have investigated antibiotic resistance in AD processes using culture-independent molecular methods, these approaches are limited by their reliance on reference databases and inability to account for gene expression, leading to potential inaccuracies in resistance assessment. This study addresses these limitations by combining culture-independent whole-genome sequencing (WGS) with culture-dependent phenotypic testing to provide a more accurate understanding of antibiotic resistance in digestate. We investigated the phenotypic and genotypic resistance profiles of 18 antibiotic-resistant bacteria (ARB) isolated from digestates produced from food waste and animal manure. Resistance was assessed using WGS and Estrip testing across 12 antibiotics from multiple classes. This is the first study to directly compare phenotypic and genotypic resistance in bacteria isolated from digestate, revealing significant discrepancies between the two methods. Approximately 30 % of resistance levels were misinterpreted when relying solely on culture-independent methods, with both over- and underestimation observed. These findings highlight the necessity of integrating both methods for reliable resistance assessments. Additionally, our WGS analysis indicated low potential for transferability of detected ARGs among the isolated ARB, suggesting a limited risk of environmental dissemination. This study provides new insights into antibiotic resistance in digestate and underscores the importance of integrating methodological approaches to achieve accurate evaluations of resistance risks.
Collapse
Affiliation(s)
- He Sun
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jolanta J Levenfors
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden; Ultupharma AB, Uppsala, Sweden
| | - Christian Brandt
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Anna Schnürer
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| |
Collapse
|
5
|
Méndez A, Maisto F, Pavlović J, Rusková M, Pangallo D, Sanmartín P. Microbiome shifts elicited by ornamental lighting of granite facades identified by MinION sequencing. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 261:113065. [PMID: 39549663 DOI: 10.1016/j.jphotobiol.2024.113065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/07/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Night-time outdoor illumination in combination with natural sunlight can influence the visible phototrophic colonizers (mainly algae) growing on stone facades; however, the effects on the microbiome (invisible to the naked eye) are not clear. The presence of stone-dwelling microbes, such as bacteria, diatoms, fungi, viruses and archaea, drives further biological colonization, which may exacerbate the biodeterioration of substrates. Considering the microbiome is therefore important for conservation of the built heritage. The impact of the following types of lighting on the relative abundance and diversity of the microbiome on granite ashlars was evaluated in a year-long outdoor pilot study: no lighting; lighting with a metal halide lamp (a traditional lighting system currently used to illuminate monuments); and lighting with a novel LED lamp (an environmentally sound prototype lamp with a biostatic effect, halting biological colonization by phototrophs, currently under trial). Culturable fractions of microbiome and whole-genome sequencing by metabarcoding with Oxford Nanopore Sequencing (MinION) was conducted for bacteria and fungi in order to complement both community characterization strategies. In addition, the possible biodeteriorative profiles of the isolated strains, relative to calcium carbonate precipitation/solubilisation and iron oxidation/reduction, were investigated by plate assays. Alpha and beta diversity indexes were also determined, along with the abundance of biocide and antibiotic resistance genes. Culture-dependent microbiological analysis failed to properly show changes in community composition, for which metagenomic approaches like MinION are better suited. Thus, MinION analysis identified shifts in the granite microbiome elicited by ornamental lighting. The novel LED lamp with the biostatic effect on phototrophs caused an increase in the diversity of bacteria and fungi. In this case, the microbiome was more similar to that in the unlit samples. In the samples illuminated by the metal halide lamp, dominance of bacteria was favoured and the presence of fungi was negligible.
Collapse
Affiliation(s)
- Anxo Méndez
- CRETUS. Gemap (GI-1243), Departamento de Edafoloxía e Química Agrícola, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Francesca Maisto
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 845 51 Bratislava, Slovakia.
| | - Jelena Pavlović
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 845 51 Bratislava, Slovakia.
| | - Magdaléna Rusková
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 845 51 Bratislava, Slovakia.
| | - Domenico Pangallo
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 845 51 Bratislava, Slovakia; Caravella, s.r.o., Tupolevova 2, 851 01 Bratislava, Slovakia.
| | - Patricia Sanmartín
- CRETUS. Gemap (GI-1243), Departamento de Edafoloxía e Química Agrícola, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
6
|
Kerek Á, Román IL, Szabó Á, Papp M, Bányai K, Kardos G, Kaszab E, Bali K, Makrai L, Jerzsele Á. Comprehensive Metagenomic Analysis of Veterinary Probiotics in Broiler Chickens. Animals (Basel) 2024; 14:1927. [PMID: 38998039 PMCID: PMC11240415 DOI: 10.3390/ani14131927] [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: 05/26/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Probiotics are widely used in broiler chickens to support the gut microbiome, gut health, and to reduce the amount of antibiotics used. Despite their benefits, there is concern over their ability to carry and spread antimicrobial resistance genes (ARGs), posing a significant public health risk. This study utilized next-generation sequencing to investigate ARGs in probiotics approved for poultry, focusing on their potential to be transferred via mobile genetic elements such as plasmids and phages. We examined the gut microbiome and resistome changes in 60 broiler chickens over their rearing period, correlating these changes with different probiotic treatments. Specific resistance mechanisms against critically important antibiotics were identified, including genes related to fluoroquinolone resistance and peptide antibiotic resistance. We also found genes with significant relevance to public health (aadK, AAC(6')-Ii) and multiple drug-resistance genes (vmlR, ykkC, ykkD, msrC, clbA, eatAv). Only one phage-encoded gene (dfrA43) was detected, with no evidence of plasmid or mobile genetic element transmission. Additionally, metagenomic analysis of fecal samples showed no significant changes corresponding to time or diet across groups. Our findings highlight the potential risks associated with the use of probiotics in poultry, particularly regarding the carriage of ARGs. It is crucial to conduct further research into the molecular genetics of probiotics to develop strategies that mitigate the risk of resistance gene transfer in agriculture, ensuring the safe and effective use of probiotics in animal husbandry.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
| | - István László Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
| | - Márton Papp
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- Centre for Bioinformatics, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Krisztián Bányai
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- Veterinary Medical Research Institute, Hungária krt. 21, H-1143 Budapest, Hungary
| | - Gábor Kardos
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- One Health Institute, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
- National Public Health Center, Albert Flórián út 2-6, H-1097 Budapest, Hungary
- Department of Gerontology, Faculty of Health Sciences, University of Debrecen, Sóstói út 2-4, H-4400 Nyíregyháza, Hungary
| | - Eszter Kaszab
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- One Health Institute, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Krisztina Bali
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - László Makrai
- Autovakcina Kft., Szabadság sgrt. 57, H-1171 Budapest, Hungary;
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
| |
Collapse
|
7
|
Gomes AFF, de Almeida LG, Cônsoli FL. Comparative Genomics of Pesticide-Degrading Enterococcus Symbionts of Spodoptera frugiperda (Lepidoptera: Noctuidae) Leads to the Identification of Two New Species and the Reappraisal of Insect-Associated Enterococcus Species. MICROBIAL ECOLOGY 2023; 86:2583-2605. [PMID: 37433981 DOI: 10.1007/s00248-023-02264-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023]
Abstract
Enterococcus species have been described as core members of the microbial community of Spodoptera frugiperda (Lepidoptera: Noctuidae) and have been previously reported as insecticide degrading agents. This study aimed to investigate the molecular composition of these microbial symbionts of S. frugiperda to better understand their association with the host and their potential for insecticide metabolization. Through phenotypic assays and comparative genomic analyses of several pesticide-degrading Enterococcus isolated from the gut of S. frugiperda larvae, we identified two new species: Enterococcus entomosocium n. sp. and Enterococcus spodopteracolus n. sp. Their identities as new species were confirmed by whole genome alignment, utilizing cut-offs of 95-96% for the average nucleotide identity (ANI) and 70% for the digital DNA: DNA hybridization (dDDH) values. The systematic positioning of these new species within the genus Enterococcus was resolved using genome-based analysis, revealing Enterococcus casseliflavus as a sister group of E. entomosocium n. sp., and Enterococcus mundtii as a sister group of E. spodopteracolus n. sp. Comparative genomic analyses of several isolates of E. entomosocium n. sp. and E. spodopteracolus n. sp. provided a better assessment of the interactions established in the symbiotic association with S. frugiperda and led to the discovery of misidentified new species of Enterococcus associated with insects. Our analyses indicated that the potential of E. entomosocium n. sp. and E. spodopteracolus n. sp. to metabolize different pesticides arises from molecular mechanisms that result in rapid evolution of new phenotypes in response to environmental stressors, in this case, the pesticides their host insect is exposed to.
Collapse
Affiliation(s)
- Ana Flávia Freitas Gomes
- Luiz de Queiroz College of Agriculture, Department of Entomology and Acarology, Insect Interactions Laboratory, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Luís Gustavo de Almeida
- Luiz de Queiroz College of Agriculture, Department of Entomology and Acarology, Insect Interactions Laboratory, University of São Paulo, Piracicaba, São Paulo, Brazil
| | - Fernando Luis Cônsoli
- Luiz de Queiroz College of Agriculture, Department of Entomology and Acarology, Insect Interactions Laboratory, University of São Paulo, Piracicaba, São Paulo, Brazil.
| |
Collapse
|
8
|
Sharon BM, Hulyalkar NV, Zimmern PE, Palmer KL, De Nisco NJ. Inter-species diversity and functional genomic analyses of closed genome assemblies of clinically isolated, megaplasmid-containing Enterococcus raffinosus Er676 and ATCC49464. Access Microbiol 2023; 5:acmi000508.v3. [PMID: 37424546 PMCID: PMC10323788 DOI: 10.1099/acmi.0.000508.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/10/2023] [Indexed: 07/11/2023] Open
Abstract
Enterococcus raffinosus is an understudied member of its genus possessing a characteristic megaplasmid contributing to a large genome size. Although less commonly associated with human infection compared to other enterococci, this species can cause disease and persist in diverse niches such as the gut, urinary tract, blood and environment. Few complete genome assemblies have been published to date for E. raffinosus . In this study, we report the complete assembly of the first clinical urinary E. raffinosus strain, Er676, isolated from a postmenopausal woman with history of recurrent urinary tract infection. We additionally completed the assembly of clinical type strain ATCC49464. Comparative genomic analyses reveal inter-species diversity driven by large accessory genomes. The presence of a conserved megaplasmid indicates it is a ubiquitous and vital genetic feature of E. raffinosus . We find that the E. raffinosus chromosome is enriched for DNA replication and protein biosynthesis genes while the megaplasmid is enriched for transcription and carbohydrate metabolism genes. Prophage analysis suggests that diversity in the chromosome and megaplasmid sequences arises, in part, from horizontal gene transfer. Er676 demonstrated the largest genome size reported to date for E. raffinosus and the highest probability of human pathogenicity. Er676 also possesses multiple antimicrobial resistance genes, of which all but one are encoded on the chromosome, and has the most complete prophage sequences. Complete assembly and comparative analyses of the Er676 and ATCC49464 genomes provide important insight into the inter-species diversity of E. raffinosus that gives it its ability to colonize and persist in the human body. Investigating genetic factors that contribute to the pathogenicity of this species will provide valuable tools to combat diseases caused by this opportunistic pathogen.
Collapse
Affiliation(s)
- Belle M. Sharon
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Neha V. Hulyalkar
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Philippe E. Zimmern
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kelli L. Palmer
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Nicole J. De Nisco
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
9
|
Pellizza L, Bialer MG, Sieira R, Aran M. MliR, a novel MerR-like regulator of iron homeostasis, impacts metabolism, membrane remodeling, and cell adhesion in the marine Bacteroidetes Bizionia argentinensis. Front Microbiol 2022; 13:987756. [PMID: 36118216 PMCID: PMC9478572 DOI: 10.3389/fmicb.2022.987756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The MerR family is a group of transcriptional activators with conserved N-terminal helix-turn-helix DNA binding domains and variable C-terminal effector binding regions. In most MerR proteins the effector binding domain (EBD) contains a cysteine center suited for metal binding and mediates the response to environmental stimuli, such as oxidative stress, heavy metals or antibiotics. We here present a novel transcriptional regulator classified in the MerR superfamily that lacks an EBD domain and has neither conserved metal binding sites nor cysteine residues. This regulator from the psychrotolerant bacteria Bizionia argentinensis JUB59 is involved in iron homeostasis and was named MliR (MerR-like iron responsive Regulator). In silico analysis revealed that homologs of the MliR protein are widely distributed among different bacterial species. Deletion of the mliR gene led to decreased cell growth, increased cell adhesion and filamentation. Genome-wide transcriptomic analysis showed that genes associated with iron homeostasis were downregulated in mliR-deletion mutant. Through nuclear magnetic resonance-based metabolomics, ICP-MS, fluorescence microscopy and biochemical analysis we evaluated metabolic and phenotypic changes associated with mliR deletion. This work provides the first evidence of a MerR-family regulator involved in iron homeostasis and contributes to expanding our current knowledge on relevant metabolic pathways and cell remodeling mechanisms underlying in the adaptive response to iron availability in bacteria.
Collapse
|
10
|
Atas B, Aksoy CS, Avci FG, Sayar NA, Ulgen K, Ozkirimli E, Akbulut BS. Carvacrol Enhances the Antimicrobial Potency of Berberine in Bacillus subtilis. Curr Microbiol 2022; 79:135. [PMID: 35303184 DOI: 10.1007/s00284-022-02823-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/21/2022] [Indexed: 11/26/2022]
Abstract
The essential oil carvacrol from oregano displays a wide range of biological activities among which is found the inhibition of efflux pumps. Thus, using carvacrol, the current work undertook the effort to potentiate the antimicrobial activity of berberine, a natural product with limited antimicrobial efficacy due to its efflux. Following the selection of concentrations for the combinatorial treatments, guided by checkerboard microtiter plate assay and growth experiments, ethidium bromide accumulation assay was used to find that 25 μg mL-1 carvacrol displayed a weak efflux pump inhibitor character in Bacillus subtilis. Scanning electron microscopy images and cellular material leakage assays showed that carvacrol at this concentration neither altered the morphology nor the permeability of the membrane alone but when combined with 75 μg mL-1 berberine. Among the efflux pumps of different families found in B. subtilis, except for BmrA and Mdr, the increase in the expressional changes was striking, with Blt displaying ~ 4500-fold increase in expression under the combination treatment. Overall, the findings demonstrated that carvacrol potentiated the effect of berberine; however, not only multiple pumps but also different targets may be responsible for the observed activity.
Collapse
Affiliation(s)
- Basak Atas
- Bioengineering Department, Marmara University, Kadikoy, 34722, Istanbul, Turkey
| | - Cemile Selin Aksoy
- Bioengineering Department, Marmara University, Kadikoy, 34722, Istanbul, Turkey
| | - Fatma Gizem Avci
- Bioengineering Department, Uskudar University, Uskudar, 34662, Istanbul, Turkey
| | - Nihat Alpagu Sayar
- Bioengineering Department, Marmara University, Kadikoy, 34722, Istanbul, Turkey
| | - Kutlu Ulgen
- Chemical Engineering Department, Bogazici University, Bebek, 34342, Istanbul, Turkey
| | - Elif Ozkirimli
- Chemical Engineering Department, Bogazici University, Bebek, 34342, Istanbul, Turkey
| | | |
Collapse
|
11
|
Chaptal V, Zampieri V, Wiseman B, Orelle C, Martin J, Nguyen KA, Gobet A, Di Cesare M, Magnard S, Javed W, Eid J, Kilburg A, Peuchmaur M, Marcoux J, Monticelli L, Hogbom M, Schoehn G, Jault JM, Boumendjel A, Falson P. Substrate-bound and substrate-free outward-facing structures of a multidrug ABC exporter. SCIENCE ADVANCES 2022; 8:eabg9215. [PMID: 35080979 DOI: 10.1101/2021.03.12.435132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Multidrug ABC transporters translocate drugs across membranes by a mechanism for which the molecular features of drug release are so far unknown. Here, we resolved three ATP-Mg2+-bound outward-facing conformations of the Bacillus subtilis (homodimeric) BmrA by x-ray crystallography and single-particle cryo-electron microscopy (EM) in detergent solution, one of them with rhodamine 6G (R6G), a substrate exported by BmrA when overexpressed in B. subtilis. Two R6G molecules bind to the drug-binding cavity at the level of the outer leaflet, between transmembrane (TM) helices 1-2 of one monomer and TM5'-6' of the other. They induce a rearrangement of TM1-2, highlighting a local flexibility that we confirmed by hydrogen/deuterium exchange and molecular dynamics simulations. In the absence of R6G, simulations show a fast postrelease occlusion of the cavity driven by hydrophobicity, while when present, R6G can move within the cavity, maintaining it open.
Collapse
Affiliation(s)
- Vincent Chaptal
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Veronica Zampieri
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Benjamin Wiseman
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
- Department of Biochemistry and Biophysics, Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Cédric Orelle
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Juliette Martin
- Modeling Biological Macromolecules Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Kim-Anh Nguyen
- University of Grenoble Alpes, INSERM, LRB, 38000 Grenoble, France
| | - Alexia Gobet
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Margot Di Cesare
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Sandrine Magnard
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Waqas Javed
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Jad Eid
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Arnaud Kilburg
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Marine Peuchmaur
- University of Grenoble Alpes, CNRS, DPM UMR 5063, 38041 Grenoble, France
| | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR 5089, Université de Toulouse, CNRS, UPS, 31000 Toulouse, France
| | - Luca Monticelli
- Modeling Biological Macromolecules Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Martin Hogbom
- Department of Biochemistry and Biophysics, Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Guy Schoehn
- University of Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Jean-Michel Jault
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | | | - Pierre Falson
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| |
Collapse
|
12
|
Chaptal V, Zampieri V, Wiseman B, Orelle C, Martin J, Nguyen KA, Gobet A, Di Cesare M, Magnard S, Javed W, Eid J, Kilburg A, Peuchmaur M, Marcoux J, Monticelli L, Hogbom M, Schoehn G, Jault JM, Boumendjel A, Falson P. Substrate-bound and substrate-free outward-facing structures of a multidrug ABC exporter. SCIENCE ADVANCES 2022; 8:eabg9215. [PMID: 35080979 PMCID: PMC8791611 DOI: 10.1126/sciadv.abg9215] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Multidrug ABC transporters translocate drugs across membranes by a mechanism for which the molecular features of drug release are so far unknown. Here, we resolved three ATP-Mg2+-bound outward-facing conformations of the Bacillus subtilis (homodimeric) BmrA by x-ray crystallography and single-particle cryo-electron microscopy (EM) in detergent solution, one of them with rhodamine 6G (R6G), a substrate exported by BmrA when overexpressed in B. subtilis. Two R6G molecules bind to the drug-binding cavity at the level of the outer leaflet, between transmembrane (TM) helices 1-2 of one monomer and TM5'-6' of the other. They induce a rearrangement of TM1-2, highlighting a local flexibility that we confirmed by hydrogen/deuterium exchange and molecular dynamics simulations. In the absence of R6G, simulations show a fast postrelease occlusion of the cavity driven by hydrophobicity, while when present, R6G can move within the cavity, maintaining it open.
Collapse
Affiliation(s)
- Vincent Chaptal
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Veronica Zampieri
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Benjamin Wiseman
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
- Department of Biochemistry and Biophysics, Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Cédric Orelle
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Juliette Martin
- Modeling Biological Macromolecules Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Kim-Anh Nguyen
- University of Grenoble Alpes, INSERM, LRB, 38000 Grenoble, France
| | - Alexia Gobet
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Margot Di Cesare
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Sandrine Magnard
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Waqas Javed
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Jad Eid
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Arnaud Kilburg
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Marine Peuchmaur
- University of Grenoble Alpes, CNRS, DPM UMR 5063, 38041 Grenoble, France
| | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR 5089, Université de Toulouse, CNRS, UPS, 31000 Toulouse, France
| | - Luca Monticelli
- Modeling Biological Macromolecules Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | - Martin Hogbom
- Department of Biochemistry and Biophysics, Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Guy Schoehn
- University of Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Jean-Michel Jault
- Bacterial Nucleotide-Binding Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
| | | | - Pierre Falson
- Drug Resistance and Membrane Proteins Group, Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, IBCP, 7, passage du Vercors, 69367 Lyon, France
- Corresponding author.
| |
Collapse
|
13
|
Fang C, Zhang Y. Bacterial MerR family transcription regulators: activationby distortion. Acta Biochim Biophys Sin (Shanghai) 2021; 54:25-36. [PMID: 35130613 PMCID: PMC9909328 DOI: 10.3724/abbs.2021003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Transcription factors (TFs) modulate gene expression by regulating the accessibility of promoter DNA to RNA polymerases (RNAPs) in bacteria. The MerR family TFs are a large class of bacterial proteins unique in their physiological functions and molecular action: they function as transcription repressors under normal circumstances, but rapidly transform to transcription activators under various cellular triggers, including oxidative stress, imbalance of cellular metal ions, and antibiotic challenge. The promoters regulated by MerR TFs typically contain an abnormal long spacer between the -35 and -10 elements, where MerR TFs bind and regulate transcription activity through unique mechanisms. In this review, we summarize the function, ligand reception, DNA recognition, and molecular mechanism of transcription regulation of MerR-family TFs.
Collapse
Affiliation(s)
- Chengli Fang
- Key Laboratory of Synthetic BiologyCAS Center for Excellence in Molecular Plant SciencesShanghai Institute of Plant Physiology and EcologyChinese Academy of SciencesShanghai200032China
| | - Yu Zhang
- Key Laboratory of Synthetic BiologyCAS Center for Excellence in Molecular Plant SciencesShanghai Institute of Plant Physiology and EcologyChinese Academy of SciencesShanghai200032China
| |
Collapse
|
14
|
Microbial Efflux Pump Inhibitors: A Journey around Quinoline and Indole Derivatives. Molecules 2021; 26:molecules26226996. [PMID: 34834098 PMCID: PMC8618814 DOI: 10.3390/molecules26226996] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/29/2022] Open
Abstract
Antimicrobial resistance (AMR) is a complex threat to human health and, to date, it represents a hot topic in drug discovery. The use of non-antibiotic molecules to block resistance mechanisms is a powerful alternative to the identification of new antibiotics. Bacterial efflux pumps exert the early step of AMR development, allowing the bacteria to grow in presence of sub-inhibitory drug concentration and develop more specific resistance mechanisms. Thus, efflux pump inhibitors (EPIs) offer a great opportunity to fight AMR, potentially restoring antibiotic activity. Based on our experience in designing and synthesizing novel EPIs, herein, we retrieved information around quinoline and indole derivatives reported in literature on this topic. Thus, our aim was to collect all data around these promising classes of EPIs in order to delineate a comprehensive structure–activity relationship (SAR) around each core for different microbes. With this review article, we aim to help future research in the field in the discovery of new microbial EPIs with improved activity and a better safety profile.
Collapse
|
15
|
Short FL, Liu Q, Shah B, Clift HE, Naidu V, Li L, Prity FT, Mabbutt BC, Hassan KA, Paulsen IT. The Acinetobacter baumannii disinfectant resistance protein, AmvA, is a spermidine and spermine efflux pump. Commun Biol 2021; 4:1114. [PMID: 34552198 PMCID: PMC8458285 DOI: 10.1038/s42003-021-02629-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 09/01/2021] [Indexed: 02/08/2023] Open
Abstract
Antimicrobial resistance genes, including multidrug efflux pumps, evolved long before the ubiquitous use of antimicrobials in medicine and infection control. Multidrug efflux pumps often transport metabolites, signals and host-derived molecules in addition to antibiotics or biocides. Understanding their ancestral physiological roles could inform the development of strategies to subvert their activity. In this study, we investigated the response of Acinetobacter baumannii to polyamines, a widespread, abundant class of amino acid-derived metabolites, which led us to identify long-chain polyamines as natural substrates of the disinfectant efflux pump AmvA. Loss of amvA dramatically reduced tolerance to long-chain polyamines, and these molecules induce expression of amvA through binding to its cognate regulator AmvR. A second clinically-important efflux pump, AdeABC, also contributed to polyamine tolerance. Our results suggest that the disinfectant resistance capability that allows A. baumannii to survive in hospitals may have evolutionary origins in the transport of polyamine metabolites.
Collapse
Affiliation(s)
- Francesca L. Short
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia ,grid.1002.30000 0004 1936 7857Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC Australia
| | - Qi Liu
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia
| | - Bhumika Shah
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia
| | - Heather E. Clift
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia ,grid.280427.b0000 0004 0434 015XPresent Address: Versiti Blood Research Institute, Milwaukee, WI USA
| | - Varsha Naidu
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia
| | - Liping Li
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia
| | - Farzana T. Prity
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia
| | - Bridget C. Mabbutt
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia
| | - Karl A. Hassan
- grid.266842.c0000 0000 8831 109XSchool of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW Australia
| | - Ian T. Paulsen
- grid.1004.50000 0001 2158 5405Department of Molecular Sciences, Macquarie University, North Ryde, NSW Australia
| |
Collapse
|
16
|
Drew D, North RA, Nagarathinam K, Tanabe M. Structures and General Transport Mechanisms by the Major Facilitator Superfamily (MFS). Chem Rev 2021; 121:5289-5335. [PMID: 33886296 PMCID: PMC8154325 DOI: 10.1021/acs.chemrev.0c00983] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/12/2022]
Abstract
The major facilitator superfamily (MFS) is the largest known superfamily of secondary active transporters. MFS transporters are responsible for transporting a broad spectrum of substrates, either down their concentration gradient or uphill using the energy stored in the electrochemical gradients. Over the last 10 years, more than a hundred different MFS transporter structures covering close to 40 members have provided an atomic framework for piecing together the molecular basis of their transport cycles. Here, we summarize the remarkable promiscuity of MFS members in terms of substrate recognition and proton coupling as well as the intricate gating mechanisms undergone in achieving substrate translocation. We outline studies that show how residues far from the substrate binding site can be just as important for fine-tuning substrate recognition and specificity as those residues directly coordinating the substrate, and how a number of MFS transporters have evolved to form unique complexes with chaperone and signaling functions. Through a deeper mechanistic description of glucose (GLUT) transporters and multidrug resistance (MDR) antiporters, we outline novel refinements to the rocker-switch alternating-access model, such as a latch mechanism for proton-coupled monosaccharide transport. We emphasize that a full understanding of transport requires an elucidation of MFS transporter dynamics, energy landscapes, and the determination of how rate transitions are modulated by lipids.
Collapse
Affiliation(s)
- David Drew
- Department
of Biochemistry and Biophysics, Stockholm
University, SE 106 91 Stockholm, Sweden
| | - Rachel A. North
- Department
of Biochemistry and Biophysics, Stockholm
University, SE 106 91 Stockholm, Sweden
| | - Kumar Nagarathinam
- Center
of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Lübeck, D-23538, Lübeck, Germany
| | - Mikio Tanabe
- Structural
Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Oho 1-1, Tsukuba, Ibaraki 305-0801, Japan
| |
Collapse
|
17
|
ALTINIŞIK F, ATAŞ B, AVCI FG. Evaluation of silibinin as an efflux pump inhibitor in Bacillus subtilis. INTERNATIONAL JOURNAL OF SECONDARY METABOLITE 2021. [DOI: 10.21448/ijsm.865031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
18
|
Ferrándiz MJ, Hernández P, de la Campa AG. Genome-wide proximity between RNA polymerase and DNA topoisomerase I supports transcription in Streptococcus pneumoniae. PLoS Genet 2021; 17:e1009542. [PMID: 33930020 PMCID: PMC8115823 DOI: 10.1371/journal.pgen.1009542] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/12/2021] [Accepted: 04/10/2021] [Indexed: 02/02/2023] Open
Abstract
Streptococcus pneumoniae is a major cause of disease and death that develops resistance to multiple antibiotics. DNA topoisomerase I (TopoI) is a novel pneumococcal drug target. TopoI is the sole type-I pneumococcal topoisomerase that regulates supercoiling homeostasis in this bacterium. In this study, a direct in vitro interaction between TopoI and RNA polymerase (RNAP) was detected by surface plasmon resonance. To understand the interplay between transcription and supercoiling regulation in vivo, genome-wide association of RNAP and TopoI was studied by ChIP-Seq. RNAP and TopoI were enriched at the promoters of 435 and 356 genes, respectively. Higher levels of expression were consistently measured in those genes whose promoters recruit both RNAP and TopoI, in contrast with those enriched in only one of them. Both enzymes occupied a narrow region close to the ATG codon. In addition, RNAP displayed a regular distribution throughout the coding regions. Likewise, the summits of peaks called with MACS tool, mapped around the ATG codon in both cases. However, RNAP showed a broader distribution towards ATG-downstream positions. Remarkably, inhibition of RNAP with rifampicin prevented the localization of TopoI at promoters and, vice versa, inhibition of TopoI with seconeolitsine prevented the binding of RNAP to promoters. This indicates a functional interplay between RNAP and TopoI. To determine the molecular factors responsible for RNAP and TopoI co-recruitment, we looked for DNA sequence motifs. We identified a motif corresponding to a -10-extended promoter for TopoI and for RNAP. Furthermore, RNAP was preferentially recruited to genes co-directionally oriented with replication, while TopoI was more abundant in head-on genes. TopoI was located in the intergenic regions of divergent genes pairs, near the promoter of the head-on gene of the pair. These results suggest a role for TopoI in the formation/stability of the RNAP-DNA complex at the promoter and during transcript elongation. Streptococcus pneumoniae is a main cause of pneumonia, meningitis and sepsis. Antibiotic resistance in this bacterium has spread worldwide, compromising medical treatment. Therefore, the development of new drugs directed to novel targets is necessary. DNA topology is essential for the regulation of replication and gene expression. Topology is regulated and maintained by DNA topoisomerases, carrying out nicking-closing reactions. Type I and type II topoisomerases act on single-stranded and double-stranded DNA, respectively. Although type II topoisomerases are the target of clinically used antibiotics, there are no clinical antibiotics directed against type I topoisomerases. Seconeolitsine, a new drug targeting topoisomerase I, is effective against bacteria that have a single type I topoisomerase, such as Streptococcus pneumoniae and Mycobacterium tuberculosis. In this report, we studied the role of topoisomerase I in transcription. We found that topoisomerase I and RNA polymerase physically interact in vitro and co-localize at gene promoters in vivo. Binding of each of these enzymes to promoters was prevented by the specific inhibition of the other enzyme, supporting a role for topoisomerase I in RNA polymerase transcription.
Collapse
Affiliation(s)
- María-José Ferrándiz
- Unidad de Genética Bacteriana, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Pablo Hernández
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Adela G. de la Campa
- Unidad de Genética Bacteriana, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
19
|
Henderson PJF, Maher C, Elbourne LDH, Eijkelkamp BA, Paulsen IT, Hassan KA. Physiological Functions of Bacterial "Multidrug" Efflux Pumps. Chem Rev 2021; 121:5417-5478. [PMID: 33761243 DOI: 10.1021/acs.chemrev.0c01226] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial multidrug efflux pumps have come to prominence in human and veterinary pathogenesis because they help bacteria protect themselves against the antimicrobials used to overcome their infections. However, it is increasingly realized that many, probably most, such pumps have physiological roles that are distinct from protection of bacteria against antimicrobials administered by humans. Here we undertake a broad survey of the proteins involved, allied to detailed examples of their evolution, energetics, structures, chemical recognition, and molecular mechanisms, together with the experimental strategies that enable rapid and economical progress in understanding their true physiological roles. Once these roles are established, the knowledge can be harnessed to design more effective drugs, improve existing microbial production of drugs for clinical practice and of feedstocks for commercial exploitation, and even develop more sustainable biological processes that avoid, for example, utilization of petroleum.
Collapse
Affiliation(s)
- Peter J F Henderson
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Claire Maher
- School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, New South Wales, Australia
| | - Liam D H Elbourne
- Department of Biomolecular Sciences, Macquarie University, Sydney 2109, New South Wales, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney 2019, New South Wales, Australia
| | - Bart A Eijkelkamp
- College of Science and Engineering, Flinders University, Bedford Park 5042, South Australia, Australia
| | - Ian T Paulsen
- Department of Biomolecular Sciences, Macquarie University, Sydney 2109, New South Wales, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney 2019, New South Wales, Australia
| | - Karl A Hassan
- School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, New South Wales, Australia.,ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney 2019, New South Wales, Australia
| |
Collapse
|
20
|
A Major Facilitator Superfamily (MFS) Efflux Pump, SCO4121, from Streptomyces coelicolor with Roles in Multidrug Resistance and Oxidative Stress Tolerance and Its Regulation by a MarR Regulator. Appl Environ Microbiol 2021; 87:AEM.02238-20. [PMID: 33483304 DOI: 10.1128/aem.02238-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Overexpression of efflux pumps is one of the major determinants of resistance in bacteria. Streptomyces species harbor a large array of efflux pumps that are transcriptionally silenced under laboratory conditions. However, their dissemination results in multidrug resistance in different clinical pathogens. In this study, we have identified an efflux pump from Streptomyces coelicolor, SCO4121, belonging to the major facilitator superfamily (MFS) family of transporters and characterized its role in antibiotic resistance. SCO4121 provided resistance to multiple dissimilar drugs upon overexpression in both native and heterologous hosts. Further, deletion of SCO4121 resulted in increased sensitivity toward ciprofloxacin and chloramphenicol, suggesting the pump to be a major transporter of these substrates. Apart from providing multidrug resistance, SCO4121 imparted increased tolerance against the strong oxidant HOCl. In wild-type Streptomyces coelicolor cells, these drugs were found to transcriptionally regulate the pump in a concentration-dependent manner. Additionally, we identified SCO4122, a MarR regulator that positively regulates SCO4121 in response to various drugs and the oxidant HOCl. Thus, through these studies we present the multiple roles of SCO4121 in S. coelicolor and highlight the intricate mechanisms via which it is regulated in response to antibiotics and oxidative stress.IMPORTANCE One of the key mechanisms of drug resistance in bacteria is overexpression of efflux pumps. Streptomyces species are a reservoir of a large number of efflux pumps, potentially to provide resistance to both endogenous and nonendogenous antibiotics. While many of these pumps are not expressed under standard laboratory conditions, they result in resistance to multiple drugs when spread to other bacterial pathogens through horizontal gene transfer. In this study, we have identified a widely conserved efflux pump SCO4121 from Streptomyces coelicolor with roles in both multidrug resistance and oxidative stress tolerance. We also report the presence of an adjacent MarR regulator, SCO4122, which positively regulates SCO4121 in the presence of diverse substrates in a redox-responsive manner. This study highlights that soil bacteria such as Streptomyces can reveal novel mechanisms of antibiotic resistance that may potentially emerge in clinically important bacteria.
Collapse
|
21
|
Fang C, Li L, Zhao Y, Wu X, Philips SJ, You L, Zhong M, Shi X, O'Halloran TV, Li Q, Zhang Y. The bacterial multidrug resistance regulator BmrR distorts promoter DNA to activate transcription. Nat Commun 2020; 11:6284. [PMID: 33293519 PMCID: PMC7722741 DOI: 10.1038/s41467-020-20134-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/10/2020] [Indexed: 01/25/2023] Open
Abstract
The MerR-family proteins represent a unique family of bacteria transcription factors (TFs), which activate transcription in a manner distinct from canonical ones. Here, we report a cryo-EM structure of a B. subtilis transcription activation complex comprising B. subtilis six-subunit (2αββ'ωε) RNA Polymerase (RNAP) core enzyme, σA, a promoter DNA, and the ligand-bound B. subtilis BmrR, a prototype of MerR-family TFs. The structure reveals that RNAP and BmrR recognize the upstream promoter DNA from opposite faces and induce four significant kinks from the -35 element to the -10 element of the promoter DNA in a cooperative manner, which restores otherwise inactive promoter activity by shortening the length of promoter non-optimal -35/-10 spacer. Our structure supports a DNA-distortion and RNAP-non-contact paradigm of transcriptional activation by MerR TFs.
Collapse
Affiliation(s)
- Chengli Fang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, 200032, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Linyu Li
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Yihan Zhao
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, 475004, Kaifeng, China
| | - Xiaoxian Wu
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, 200032, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Steven J Philips
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Linlin You
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, 200032, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Mingkang Zhong
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Xiaojin Shi
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Thomas V O'Halloran
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
- The Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Qunyi Li
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China.
| | - Yu Zhang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, 200032, Shanghai, China.
| |
Collapse
|
22
|
Serçinoğlu O, Senturk D, Altinisik Kaya FE, Avci FG, Frlan R, Tomašič T, Ozbek P, Orelle C, Jault JM, Sariyar Akbulut B. Identification of novel inhibitors of the ABC transporter BmrA. Bioorg Chem 2020; 105:104452. [PMID: 33212311 DOI: 10.1016/j.bioorg.2020.104452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/23/2020] [Accepted: 11/01/2020] [Indexed: 01/02/2023]
Abstract
The resistance of microbes to commonly used antibiotics has become a worldwide health problem. A major underlying mechanism of microbial antibiotic resistance is the export of drugs from bacterial cells. Drug efflux is mediated through the action of multidrug resistance efflux pumps located in the bacterial cell membranes. The critical role of bacterial efflux pumps in antibiotic resistance has directed research efforts to the identification of novel efflux pump inhibitors that can be used alongside antibiotics in clinical settings. Here, we aimed to find potential inhibitors of the archetypical ATP-binding cassette (ABC) efflux pump BmrA of Bacillus subtilis via virtual screening of the Mu.Ta.Lig. Chemotheca small molecule library. Molecular docking calculations targeting the nucleotide-binding domain of BmrA were performed using AutoDock Vina. Following a further drug-likeness filtering step based on Lipinski's Rule of Five, top 25 scorers were identified. These ligands were then clustered into separate groups based on their contact patterns with the BmrA nucleotide-binding domain. Six ligands with distinct contact patterns were used for further in vitro inhibition assays based on intracellular ethidium bromide accumulation. Using this methodology, we identified two novel inhibitors of BmrA from the Chemotheca small molecule library.
Collapse
Affiliation(s)
- Onur Serçinoğlu
- Department of Bioengineering, Recep Tayyip Erdogan University, Fener 53100, Rize, Turkey
| | - Duygu Senturk
- Department of Bioengineering, Marmara University, Kadikoy 34722, Istanbul, Turkey
| | | | - Fatma Gizem Avci
- Department of Bioengineering, Uskudar University, Uskudar 34662, Istanbul, Turkey
| | - Rok Frlan
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Pemra Ozbek
- Department of Bioengineering, Marmara University, Kadikoy 34722, Istanbul, Turkey
| | - Cédric Orelle
- University of Lyon, CNRS, UMR5086 "Molecular Microbiology and Structural Biochemistry", 7 passage du Vercors, 69367 Lyon Cedex 7, France
| | - Jean-Michel Jault
- University of Lyon, CNRS, UMR5086 "Molecular Microbiology and Structural Biochemistry", 7 passage du Vercors, 69367 Lyon Cedex 7, France
| | | |
Collapse
|
23
|
Jonkers TJH, Steenhuis M, Schalkwijk L, Luirink J, Bald D, Houtman CJ, Kool J, Lamoree MH, Hamers T. Development of a high-throughput bioassay for screening of antibiotics in aquatic environmental samples. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 729:139028. [PMID: 32498177 DOI: 10.1016/j.scitotenv.2020.139028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 05/26/2023]
Abstract
The goal of the present study was to select a Gram-positive (Gram+) and Gram-negative (Gram-) strain to measure antimicrobial activity in environmental samples, allowing high-throughput environmental screening. The sensitivity of eight pre-selected bacterial strains were tested to a training set of ten antibiotics, i.e. three Gram+ Bacillus subtilis strains with different read-outs, and five Gram- strains. The latter group consisted of a bioluminescent Allivibrio fischeri strain and four Escherichia coli strains, i.e. a wild type (WT) and three strains with a modified cell envelope to increase their sensitivity. The WT B. subtilis and an E. coli strain newly developed in this study, were most sensitive to the training set. This E. coli strain carries an open variant of an outer membrane protein combined with an inactivated multidrug efflux transport system. The assay conditions of these two strains were optimized and validated by exposure to a validation set of thirteen antibiotics with clinical and environmental relevance. The assay sensitivity ranged from the ng/mL to μg/mL range. The applicability of the assays for toxicological characterization of aquatic environmental samples was demonstrated for hospital effluent extract. A future application includes effect-directed analysis to identify yet unknown antibiotic contaminants or their transformation products.
Collapse
Affiliation(s)
- Tim J H Jonkers
- Department of Environment & Health, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands.
| | - Maurice Steenhuis
- Department of Molecular Microbiology, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Louis Schalkwijk
- Department of Environment & Health, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Joen Luirink
- Department of Molecular Microbiology, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Dirk Bald
- Department of Molecular Cell Biology, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Corine J Houtman
- The Water Laboratory, J.W. Lucasweg 2, 2031 BE Haarlem, the Netherlands
| | - Jeroen Kool
- Biomolecular Analysis Group, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Marja H Lamoree
- Department of Environment & Health, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands; KWR Watercycle Research Institute, P.O. Box 1072, 3430 BB Nieuwegein, the Netherlands
| | - Timo Hamers
- Department of Environment & Health, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
24
|
Shrestha P, Ni J, Wong TY. Synergistic and antagonistic interactions of triclosan with various antibiotics in bacteria. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2020; 38:187-203. [PMID: 32648520 DOI: 10.1080/26896583.2020.1781494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Triclosan (TCS), a well-studied antimicrobial compound and an environmental pollutant, is present in many household products. A systematic survey of TCS-antibiotic-bacteria interactions is lacking. We wish to understand the origin of such interactions by testing 16 phylogenetically well-characterized bacteria for their sensitivities to 6 different classes of antibiotics with or without the presence of TCS. Our results show that TCS interacts synergistically with some antibiotics against some Bacilli species. TCS could also interact antagonistically with other antibiotics against certain bacteria, including pathogens such as Pseudomonas aeruginosa and Stenotrophomonas maltophilia. Antagonism between drugs often coincided with the concomitant enhanced removal of Ethidium bromide (EtBr) from the cells. Enterococcus faecalis shows a unique response to TCS. High levels of TCS inhibits E. faecalis. Cells survive at lower TCS concentrations, and these cells can remove EtBr more readily than unexposed cells. At even lower TCS concentration, cell-growth is inhibited again, causing the culture to exhibit a unique extra inhibition zone around the TCS-disk. The TCS-antibiotic-bacteria interaction profiles of some bacteria do not follow their bacterial phylogenetic relations. This suggests that such interactions may be related to horizontal gene transfer among different bacteria.
Collapse
Affiliation(s)
- Prabin Shrestha
- Biological Sciences Department, University of Memphis, Memphis, Tennessee, USA
| | - Jie Ni
- Biological Sciences Department, University of Memphis, Memphis, Tennessee, USA
| | - Tit-Yee Wong
- Biological Sciences Department, University of Memphis, Memphis, Tennessee, USA
| |
Collapse
|
25
|
Avci FG, Atas B, Aksoy CS, Kurpejovic E, Gulsoy Toplan G, Gurer C, Guillerminet M, Orelle C, Jault JM, Sariyar Akbulut B. Repurposing bioactive aporphine alkaloids as efflux pump inhibitors. Fitoterapia 2019; 139:104371. [PMID: 31629051 DOI: 10.1016/j.fitote.2019.104371] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 01/13/2023]
Abstract
Extrusion of drugs or drug-like compounds through bacterial efflux pumps is a serious health issue that leads to loss in drug efficacy. Combinatorial therapies of low-efficacy drugs with efflux pump inhibitors may help to restore the activities of such drugs. In this quest, natural products are attractive molecules, since in addition to their wide range of bioactivities they may inhibit efflux pumps. The current work repurposed the bioactive alkaloid roemerine as a potential efflux pump inhibitor. In Bacillus subtilis, both Bmr and BmrA, belonging to the major facilitator and the ATP-binding cassette superfamilies, respectively, were found to be inhibited by roemerine. Scanning electron microscopy and RNA-Seq analyses showed that it potentiated the effect of berberine. Growth rates and checkerboard assays confirmed the synergy of roemerine and berberine and that roemerine prevented berberine efflux by inhibiting Bmr. Transport assays with inverted membrane vesicles prepared from Escherichia coli overexpressing BmrA showed that increasing roemerine concentration decreased the transport of doxorubicin, the BmrA substrate, confirming that roemerine may also be considered as an inhibitor of BmrA. Thus, these findings suggest that conjugation of roemerine to substrates of efflux pumps, Bmr and BmrA, may help to potentiate the activity of their drug substrates.
Collapse
Affiliation(s)
- Fatma Gizem Avci
- Department of Bioengineering, Marmara University, 34722 Kadikoy, Istanbul, Turkey.
| | - Basak Atas
- Department of Bioengineering, Marmara University, 34722 Kadikoy, Istanbul, Turkey.
| | - Cemile Selin Aksoy
- Department of Bioengineering, Marmara University, 34722 Kadikoy, Istanbul, Turkey
| | - Eldin Kurpejovic
- Department of Bioengineering, Marmara University, 34722 Kadikoy, Istanbul, Turkey
| | - Gizem Gulsoy Toplan
- Department of Pharmacognosy, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey.
| | - Caglayan Gurer
- Department of Pharmacognosy, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey.
| | - Maxime Guillerminet
- University of Lyon, CNRS, UMR5086 "Molecular Microbiology and Structural Biochemistry", IBCP, 7 Passage du Vercors, F-69367 Lyon, France.
| | - Cedric Orelle
- University of Lyon, CNRS, UMR5086 "Molecular Microbiology and Structural Biochemistry", IBCP, 7 Passage du Vercors, F-69367 Lyon, France.
| | - Jean-Michel Jault
- University of Lyon, CNRS, UMR5086 "Molecular Microbiology and Structural Biochemistry", IBCP, 7 Passage du Vercors, F-69367 Lyon, France.
| | | |
Collapse
|
26
|
Pasqua M, Grossi M, Zennaro A, Fanelli G, Micheli G, Barras F, Colonna B, Prosseda G. The Varied Role of Efflux Pumps of the MFS Family in the Interplay of Bacteria with Animal and Plant Cells. Microorganisms 2019; 7:microorganisms7090285. [PMID: 31443538 PMCID: PMC6780985 DOI: 10.3390/microorganisms7090285] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Efflux pumps represent an important and large group of transporter proteins found in all organisms. The importance of efflux pumps resides in their ability to extrude a wide range of antibiotics, resulting in the emergence of multidrug resistance in many bacteria. Besides antibiotics, multidrug efflux pumps can also extrude a large variety of compounds: Bacterial metabolites, plant-produced compounds, quorum-sensing molecules, and virulence factors. This versatility makes efflux pumps relevant players in interactions not only with other bacteria, but also with plant or animal cells. The multidrug efflux pumps belonging to the major facilitator superfamily (MFS) are widely distributed in microbial genomes and exhibit a large spectrum of substrate specificities. Multidrug MFS efflux pumps are present either as single-component transporters or as tripartite complexes. In this review, we will summarize how the multidrug MFS efflux pumps contribute to the interplay between bacteria and targeted host cells, with emphasis on their role in bacterial virulence, in the colonization of plant and animal host cells and in biofilm formation. We will also address the complexity of these interactions in the light of the underlying regulatory networks required for the effective activation of efflux pump genes.
Collapse
Affiliation(s)
- Martina Pasqua
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Milena Grossi
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Alessandro Zennaro
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Giulia Fanelli
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Gioacchino Micheli
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche (CNR), P.le A. Moro 5, 00185 Roma, Italy
| | - Frederic Barras
- Département de Microbiologie, Institut Pasteur, 75015 Paris, France
- Équipe de Recherche Labellisée (ERL) Microbiology, Centre National de la Recherche Scientifique (CNRS), 13009 Marseille, France
| | - Bianca Colonna
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy
| | - Gianni Prosseda
- Istituto Pasteur Italia, Dipartimento di Biologia e Biotecnologie "C. Darwin", Sapienza Università di Roma, Via dei Sardi 70, 00185 Rome, Italy.
| |
Collapse
|
27
|
He H, Zhou P, Shimabuku KK, Fang X, Li S, Lee Y, Dodd MC. Degradation and Deactivation of Bacterial Antibiotic Resistance Genes during Exposure to Free Chlorine, Monochloramine, Chlorine Dioxide, Ozone, Ultraviolet Light, and Hydroxyl Radical. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:2013-2026. [PMID: 30712343 DOI: 10.1021/acs.est.8b04393] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
This work investigated degradation (measured by qPCR) and biological deactivation (measured by culture-based natural transformation) of extra- and intracellular antibiotic resistance genes (eARGs and iARGs) by free available chlorine (FAC), NH2Cl, O3, ClO2, and UV light (254 nm), and of eARGs by •OH, using a chromosomal ARG ( blt) of multidrug-resistant Bacillus subtilis 1A189. Rate constants for degradation of four 266-1017 bp amplicons adjacent to or encompassing the acfA mutation enabling blt overexpression increased in proportion to #AT+GC bps/amplicon, or in proportion to #5'-GG-3' or 5'-TT-3' doublets/amplicon, with respective values ranging from 0.59 to 2.3 (×1011 M-1 s-1) for •OH, 1.8-6.9 (×104 M-1 s-1) for O3, 3.9-9.2 (×103 M-1 s-1) for FAC, 0.35-1.2(×101 M-1 s-1) for ClO2, and 2.0-8.8 (×10-2 cm2/mJ) for UV at pH 7, and from 1.7-4.4 M-1 s-1 for NH2Cl at pH 8. For FAC, NH2Cl, O3, ClO2, and UV, ARG deactivation paralleled degradation of amplicons approximating a ∼800-1000 bp acfA-flanking sequence required for natural transformation in B. subtilis, whereas deactivation outpaced degradation for •OH. At practical disinfectant exposures, eARGs and iARGs were ≥90% degraded/deactivated by FAC, O3, and UV, but recalcitrant to NH2Cl and ClO2. iARG degradation/ deactivation always lagged cell inactivation. These findings provide a quantitative framework for evaluating ARG fate during disinfection/oxidation, and support using qPCR as a proxy for tracking ARG deactivation under carefully selected circumstances.
Collapse
Affiliation(s)
- Huan He
- Department of Civil and Environmental Engineering , University of Washington (UW) , Seattle , Washington 98195-2700 , United States
| | - Peiran Zhou
- Department of Civil and Environmental Engineering , University of Washington (UW) , Seattle , Washington 98195-2700 , United States
| | - Kyle K Shimabuku
- Department of Civil and Environmental Engineering , University of Washington (UW) , Seattle , Washington 98195-2700 , United States
| | - Xuzhi Fang
- Department of Civil and Environmental Engineering , University of Washington (UW) , Seattle , Washington 98195-2700 , United States
| | - Shu Li
- Department of Civil and Environmental Engineering , University of Washington (UW) , Seattle , Washington 98195-2700 , United States
| | - Yunho Lee
- School of Earth Sciences and Environmental Engineering , Gwangju Institute of Science and Technology (GIST) , Gwangju 61005 , Republic of Korea
| | - Michael C Dodd
- Department of Civil and Environmental Engineering , University of Washington (UW) , Seattle , Washington 98195-2700 , United States
| |
Collapse
|
28
|
Guillén Y, Noguera-Julian M, Rivera J, Casadellà M, Zevin AS, Rocafort M, Parera M, Rodríguez C, Arumí M, Carrillo J, Mothe B, Estany C, Coll J, Bravo I, Herrero C, Saz J, Sirera G, Torrella A, Navarro J, Crespo M, Negredo E, Brander C, Blanco J, Calle ML, Klatt NR, Clotet B, Paredes R. Low nadir CD4+ T-cell counts predict gut dysbiosis in HIV-1 infection. Mucosal Immunol 2019; 12:232-246. [PMID: 30171206 DOI: 10.1038/s41385-018-0083-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 02/04/2023]
Abstract
Human immunodeficiency virus (HIV)-1 infection causes severe gut and systemic immune damage, but its effects on the gut microbiome remain unclear. Previous shotgun metagenomic studies in HIV-negative subjects linked low-microbial gene counts (LGC) to gut dysbiosis in diseases featuring intestinal inflammation. Using a similar approach in 156 subjects with different HIV-1 phenotypes, we found a strong, independent, dose-effect association between nadir CD4+ T-cell counts and LGC. As in other diseases involving intestinal inflammation, the gut microbiomes of subjects with LGC were enriched in gram-negative Bacteroides, acetogenic bacteria and Proteobacteria, which are able to metabolize reactive oxygen and nitrogen species; and were depleted in oxygen-sensitive methanogenic archaea and sulfate-reducing bacteria. Interestingly, subjects with LGC also showed increased butyrate levels in direct fecal measurements, consistent with enrichment in Roseburia intestinalis despite reductions in other butyrate producers. The microbiomes of subjects with LGC were also enriched in bacterial virulence factors, as well as in genes associated with beta-lactam, lincosamide, tetracycline, and macrolide resistance. Thus, low nadir CD4+ T-cell counts, rather than HIV-1 serostatus per se, predict the presence of gut dysbiosis in HIV-1 infected subjects. Such dysbiosis does not display obvious HIV-specific features; instead, it shares many similarities with other diseases featuring gut inflammation.
Collapse
Affiliation(s)
- Yolanda Guillén
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Marc Noguera-Julian
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain
| | - Javier Rivera
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain
| | - Maria Casadellà
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Alexander S Zevin
- University of Washington, 3018 Western Avenue, Seattle, WA, 98121, USA
| | - Muntsa Rocafort
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Mariona Parera
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Cristina Rodríguez
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Marçal Arumí
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Jorge Carrillo
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Beatriz Mothe
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain.,Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Carla Estany
- Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Josep Coll
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Isabel Bravo
- Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Cristina Herrero
- Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Jorge Saz
- BCN Checkpoint, Carrer del Comte Borrell, 164, Barcelona, 08015, Catalonia, Spain
| | - Guillem Sirera
- Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Ariadna Torrella
- Infectious Diseases Unit, Hospital Universitari Vall d'Hebrón, Passeig de la Vall d'Hebrón, 119-129, Barcelona, 08035, Catalonia, Spain
| | - Jordi Navarro
- Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.,Infectious Diseases Unit, Hospital Universitari Vall d'Hebrón, Passeig de la Vall d'Hebrón, 119-129, Barcelona, 08035, Catalonia, Spain
| | - Manuel Crespo
- Infectious Diseases Unit, Internal Medicine Department, Complexo Hospitalario Universitario, Vigo. IIS Galicia Sur, Estrada de Clara Campoamor, 341, Vigo, 36312, Pontevedra, Spain
| | - Eugènia Negredo
- Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain.,Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Christian Brander
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, Barcelona, 08010, Catalonia, Spain
| | - Julià Blanco
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain
| | - Maria Luz Calle
- Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain
| | - Nichole R Klatt
- University of Washington, 3018 Western Avenue, Seattle, WA, 98121, USA
| | - Bonaventura Clotet
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain.,Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain
| | - Roger Paredes
- irsiCaixa AIDS Research Institute, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain. .,Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain. .,Universitat de Vic-Universitat Central de Catalunya, C. Sagrada Família 7, Vic, 08500, Catalonia, Spain. .,Infectious Diseases Service & Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Ctra de Canyet s/n, Badalona, 08916, Catalonia, Spain.
| |
Collapse
|
29
|
Castillo H, Li X, Schilkey F, Smith GB. Transcriptome analysis reveals a stress response of Shewanella oneidensis deprived of background levels of ionizing radiation. PLoS One 2018; 13:e0196472. [PMID: 29768440 PMCID: PMC5955497 DOI: 10.1371/journal.pone.0196472] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/13/2018] [Indexed: 11/21/2022] Open
Abstract
Natural ionizing background radiation has exerted a constant pressure on organisms since the first forms of life appeared on Earth, so that cells have developed molecular mechanisms to avoid or repair damages caused directly by radiation or indirectly by radiation-induced reactive oxygen species (ROS). In the present study, we investigated the transcriptional effect of depriving Shewanella oneidensis cultures of background levels of radiation by growing the cells in a mine 655 m underground, thus reducing the dose rate from 72.1 to 0.9 nGy h-1 from control to treatment, respectively. RNASeq transcriptome analysis showed the differential expression of 4.6 and 7.6% of the S. oneidensis genome during early- and late-exponential phases of growth, respectively. The greatest change observed in the treatment was the downregulation of ribosomal proteins (21% of all annotated ribosomal protein genes during early- and 14% during late-exponential) and tRNA genes (14% of all annotated tRNA genes in early-exponential), indicating a marked decrease in protein translation. Other significant changes were the upregulation of membrane transporters, implying an increase in the traffic of substrates across the cell membrane, as well as the up and downregulation of genes related to respiration, which could be interpreted as a response to insufficient oxidants in the cells. In other reports, there is evidence in multiple species that some ROS not just lead to oxidative stress, but act as signaling molecules to control cellular metabolism at the transcriptional level. Consistent with these reports, several genes involved in the metabolism of carbon and biosynthesis of amino acids were also regulated, lending support to the idea of a wide metabolic response. Our results indicate that S. oneidensis is sensitive to the withdrawal of background levels of ionizing radiation and suggest that a transcriptional response is required to maintain homeostasis and retain normal growth.
Collapse
Affiliation(s)
- Hugo Castillo
- Department of Biology, New Mexico State University, Las Cruces, NM, United States of America
| | - Xiaoping Li
- Department of Botany and Plant Pathology, Oregon State University, Hermiston, OR, United States of America
| | - Faye Schilkey
- National Center for Genome Resources, Santa Fe, NM, United States of America
| | - Geoffrey B Smith
- Department of Biology, New Mexico State University, Las Cruces, NM, United States of America
| |
Collapse
|
30
|
Hassan KA, Fagerlund A, Elbourne LDH, Vörös A, Kroeger JK, Simm R, Tourasse NJ, Finke S, Henderson PJF, Økstad OA, Paulsen IT, Kolstø AB. The putative drug efflux systems of the Bacillus cereus group. PLoS One 2017; 12:e0176188. [PMID: 28472044 PMCID: PMC5417439 DOI: 10.1371/journal.pone.0176188] [Citation(s) in RCA: 10] [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: 10/20/2016] [Accepted: 04/06/2017] [Indexed: 12/19/2022] Open
Abstract
The Bacillus cereus group of bacteria includes seven closely related species, three of which, B. anthracis, B. cereus and B. thuringiensis, are pathogens of humans, animals and/or insects. Preliminary investigations into the transport capabilities of different bacterial lineages suggested that genes encoding putative efflux systems were unusually abundant in the B. cereus group compared to other bacteria. To explore the drug efflux potential of the B. cereus group all putative efflux systems were identified in the genomes of prototypical strains of B. cereus, B. anthracis and B. thuringiensis using our Transporter Automated Annotation Pipeline. More than 90 putative drug efflux systems were found within each of these strains, accounting for up to 2.7% of their protein coding potential. Comparative analyses demonstrated that the efflux systems are highly conserved between these species; 70-80% of the putative efflux pumps were shared between all three strains studied. Furthermore, 82% of the putative efflux system proteins encoded by the prototypical B. cereus strain ATCC 14579 (type strain) were found to be conserved in at least 80% of 169 B. cereus group strains that have high quality genome sequences available. However, only a handful of these efflux pumps have been functionally characterized. Deletion of individual efflux pump genes from B. cereus typically had little impact to drug resistance phenotypes or the general fitness of the strains, possibly because of the large numbers of alternative efflux systems that may have overlapping substrate specificities. Therefore, to gain insight into the possible transport functions of efflux systems in B. cereus, we undertook large-scale qRT-PCR analyses of efflux pump gene expression following drug shocks and other stress treatments. Clustering of gene expression changes identified several groups of similarly regulated systems that may have overlapping drug resistance functions. In this article we review current knowledge of the small molecule efflux pumps encoded by the B. cereus group and suggest the likely functions of numerous uncharacterised pumps.
Collapse
Affiliation(s)
- Karl A. Hassan
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW, Australia
- School of BioMedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Annette Fagerlund
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Liam D. H. Elbourne
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW, Australia
| | - Aniko Vörös
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Jasmin K. Kroeger
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- Institut für Pharmazeutische Biologie und Biotechnologie, Albert-Ludwigs Universität, Freiburg, Germany
| | - Roger Simm
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Nicolas J. Tourasse
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Sarah Finke
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- Centre for Integrative Microbial Evolution (CIME), Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
| | - Peter J. F. Henderson
- School of BioMedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Ole Andreas Økstad
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- Centre for Integrative Microbial Evolution (CIME), Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
| | - Ian T. Paulsen
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW, Australia
- * E-mail: (ABK); (ITP)
| | - Anne-Brit Kolstø
- Laboratory for Microbial Dynamics (LaMDa), Section for Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- Centre for Integrative Microbial Evolution (CIME), Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
- * E-mail: (ABK); (ITP)
| |
Collapse
|
31
|
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
|
32
|
Abstract
The treatment of microbial infections has suffered greatly in this present century of pathogen dominance. Inspite of extensive research efforts and scientific advancements, the worldwide emergence of microbial tolerance continues to plague survivability. The innate property of microbe to resist any antibiotic due to evolution is the virtue of intrinsic resistance. However, the classical genetic mutations and extrachromosomal segments causing gene exchange attribute to acquired tolerance development. Rampant use of antimicrobials causes certain selection pressure which increases the resistance frequency. Genomic duplication, enzymatic site modification, target alteration, modulation in membrane permeability, and the efflux pump mechanism are the major contributors of multidrug resistance (MDR), specifically antibiotic tolerance development. MDRs will lead to clinical failures for treatment and pose health crisis. The molecular mechanisms of antimicrobial resistance are diverse as well as complex and still are exploited for new discoveries in order to prevent the surfacing of “superbugs.” Antimicrobial chemotherapy has diminished the threat of infectious diseases to some extent. To avoid the indiscriminate use of antibiotics, the new ones licensed for use have decreased with time. Additionally, in vitro assays and genomics for anti-infectives are novel approaches used in resolving the issues of microbial resistance. Proper use of drugs can keep it under check and minimize the risk of MDR spread.
Collapse
|
33
|
Korostelev YD, Zharov IA, Mironov AA, Rakhmaininova AB, Gelfand MS. Identification of Position-Specific Correlations between DNA-Binding Domains and Their Binding Sites. Application to the MerR Family of Transcription Factors. PLoS One 2016; 11:e0162681. [PMID: 27690309 PMCID: PMC5045206 DOI: 10.1371/journal.pone.0162681] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 08/26/2016] [Indexed: 11/25/2022] Open
Abstract
The large and increasing volume of genomic data analyzed by comparative methods provides information about transcription factors and their binding sites that, in turn, enables statistical analysis of correlations between factors and sites, uncovering mechanisms and evolution of specific protein-DNA recognition. Here we present an online tool, Prot-DNA-Korr, designed to identify and analyze crucial protein-DNA pairs of positions in a family of transcription factors. Correlations are identified by analysis of mutual information between columns of protein and DNA alignments. The algorithm reduces the effects of common phylogenetic history and of abundance of closely related proteins and binding sites. We apply it to five closely related subfamilies of the MerR family of bacterial transcription factors that regulate heavy metal resistance systems. We validate the approach using known 3D structures of MerR-family proteins in complexes with their cognate DNA binding sites and demonstrate that a significant fraction of correlated positions indeed form specific side-chain-to-base contacts. The joint distribution of amino acids and nucleotides hence may be used to predict changes of specificity for point mutations in transcription factors.
Collapse
Affiliation(s)
- Yuriy D. Korostelev
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, 19-1 Bolshoy Karetny pereulok, Moscow, Russia, 127994
- Department of Bioengineering and Bioinformatics, Moscow State University, 1-73 Vorobievy Gory, Moscow, Russia, 119991
| | - Ilya A. Zharov
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, 19-1 Bolshoy Karetny pereulok, Moscow, Russia, 127994
| | - Andrey A. Mironov
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, 19-1 Bolshoy Karetny pereulok, Moscow, Russia, 127994
- Department of Bioengineering and Bioinformatics, Moscow State University, 1-73 Vorobievy Gory, Moscow, Russia, 119991
| | - Alexandra B. Rakhmaininova
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, 19-1 Bolshoy Karetny pereulok, Moscow, Russia, 127994
| | - Mikhail S. Gelfand
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, 19-1 Bolshoy Karetny pereulok, Moscow, Russia, 127994
- Department of Bioengineering and Bioinformatics, Moscow State University, 1-73 Vorobievy Gory, Moscow, Russia, 119991
- * E-mail:
| |
Collapse
|
34
|
Jaing CJ, McLoughlin KS, Thissen JB, Zemla A, Gardner SN, Vergez LM, Bourguet F, Mabery S, Fofanov VY, Koshinsky H, Jackson PJ. Identification of Genome-Wide Mutations in Ciprofloxacin-Resistant F. tularensis LVS Using Whole Genome Tiling Arrays and Next Generation Sequencing. PLoS One 2016; 11:e0163458. [PMID: 27668749 PMCID: PMC5036845 DOI: 10.1371/journal.pone.0163458] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/08/2016] [Indexed: 11/19/2022] Open
Abstract
Francisella tularensis is classified as a Class A bioterrorism agent by the U.S. government due to its high virulence and the ease with which it can be spread as an aerosol. It is a facultative intracellular pathogen and the causative agent of tularemia. Ciprofloxacin (Cipro) is a broad spectrum antibiotic effective against Gram-positive and Gram-negative bacteria. Increased Cipro resistance in pathogenic microbes is of serious concern when considering options for medical treatment of bacterial infections. Identification of genes and loci that are associated with Ciprofloxacin resistance will help advance the understanding of resistance mechanisms and may, in the future, provide better treatment options for patients. It may also provide information for development of assays that can rapidly identify Cipro-resistant isolates of this pathogen. In this study, we selected a large number of F. tularensis live vaccine strain (LVS) isolates that survived in progressively higher Ciprofloxacin concentrations, screened the isolates using a whole genome F. tularensis LVS tiling microarray and Illumina sequencing, and identified both known and novel mutations associated with resistance. Genes containing mutations encode DNA gyrase subunit A, a hypothetical protein, an asparagine synthase, a sugar transamine/perosamine synthetase and others. Structural modeling performed on these proteins provides insights into the potential function of these proteins and how they might contribute to Cipro resistance mechanisms.
Collapse
Affiliation(s)
- Crystal J. Jaing
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
- * E-mail:
| | - Kevin S. McLoughlin
- Computations Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| | - James B. Thissen
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| | - Adam Zemla
- Computations Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| | - Shea N. Gardner
- Computations Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| | - Lisa M. Vergez
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| | - Feliza Bourguet
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| | - Shalini Mabery
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| | | | | | - Paul J. Jackson
- Physical Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, United States of America
| |
Collapse
|
35
|
Disruption of Transporters Affiliated with Enantio-Pyochelin Biosynthesis Gene Cluster of Pseudomonas protegens Pf-5 Has Pleiotropic Effects. PLoS One 2016; 11:e0159884. [PMID: 27442435 PMCID: PMC4956303 DOI: 10.1371/journal.pone.0159884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/08/2016] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas protegens Pf-5 (formerly Pseudomonas fluorescens) is a biocontrol bacterium that produces the siderophore enantio-pyochelin under conditions of iron starvation in a process that is often accompanied by the secretion of its biosynthesis intermediates, salicylic acid and dihydroaeruginoic acid. In this study, we investigated whether several transporters that are encoded by genes within or adjacent to the enantio-pyochelin biosynthetic cluster, serve as efflux systems for enantio-pyochelin and/or its intermediates. In addition, we determined whether these transporters have broad substrates range specificity using a Phenotype Microarray system. Intriguingly, knockouts of the pchH and fetF transporter genes resulted in mutant strains that secrete higher levels of enantio-pyochelin as well as its intermediates salicylic acid and dihydroaeruginoic acid. Analyses of these mutants did not indicate significant change in transcription of biosynthetic genes involved in enantio-pyochelin production. In contrast, the deletion mutant of PFL_3504 resulted in reduced transcription of the biosynthetic genes as well as decreased dihydroaeruginoic acid concentrations in the culture supernatant, which could either point to regulation of gene expression by the transporter or its role in dihydroaeruginoic acid transport. Disruption of each of the transporters resulted in altered stress and/or chemical resistance profile of Pf-5, which may reflect that these transporters could have specificity for rather a broad range of substrates.
Collapse
|
36
|
Schindler BD, Kaatz GW. Multidrug efflux pumps of Gram-positive bacteria. Drug Resist Updat 2016; 27:1-13. [DOI: 10.1016/j.drup.2016.04.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/28/2016] [Accepted: 04/22/2016] [Indexed: 11/16/2022]
|
37
|
Chen NH, Djoko KY, Veyrier FJ, McEwan AG. Formaldehyde Stress Responses in Bacterial Pathogens. Front Microbiol 2016; 7:257. [PMID: 26973631 PMCID: PMC4776306 DOI: 10.3389/fmicb.2016.00257] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/16/2016] [Indexed: 12/18/2022] Open
Abstract
Formaldehyde is the simplest of all aldehydes and is highly cytotoxic. Its use and associated dangers from environmental exposure have been well documented. Detoxification systems for formaldehyde are found throughout the biological world and they are especially important in methylotrophic bacteria, which generate this compound as part of their metabolism of methanol. Formaldehyde metabolizing systems can be divided into those dependent upon pterin cofactors, sugar phosphates and those dependent upon glutathione. The more prevalent thiol-dependent formaldehyde detoxification system is found in many bacterial pathogens, almost all of which do not metabolize methane or methanol. This review describes the endogenous and exogenous sources of formaldehyde, its toxic effects and mechanisms of detoxification. The methods of formaldehyde sensing are also described with a focus on the formaldehyde responsive transcription factors HxlR, FrmR, and NmlR. Finally, the physiological relevance of detoxification systems for formaldehyde in bacterial pathogens is discussed.
Collapse
Affiliation(s)
- Nathan H Chen
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia QLD, Australia
| | - Karrera Y Djoko
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia QLD, Australia
| | - Frédéric J Veyrier
- INRS-Institut Armand-Frappier, Institut National de la Recherche Scientifique, Université du Québec, Laval QC, Canada
| | - Alastair G McEwan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia QLD, Australia
| |
Collapse
|
38
|
Mousa JJ, Bruner SD. Structural and mechanistic diversity of multidrug transporters. Nat Prod Rep 2016; 33:1255-1267. [DOI: 10.1039/c6np00006a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The review article surveys recent structural and mechanistic advances in the field of multi-drug and natural product transporters.
Collapse
|
39
|
Marslin G, Revina AM, Khandelwal VKM, Balakumar K, Sheeba CJ, Franklin G. PEGylated ofloxacin nanoparticles render strong antibacterial activity against many clinically important human pathogens. Colloids Surf B Biointerfaces 2015; 132:62-70. [DOI: 10.1016/j.colsurfb.2015.04.050] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 10/23/2022]
|
40
|
Nguyen AQD, Schneider J, Wendisch VF. Elimination of polyamine N-acetylation and regulatory engineering improved putrescine production by Corynebacterium glutamicum. J Biotechnol 2014; 201:75-85. [PMID: 25449016 DOI: 10.1016/j.jbiotec.2014.10.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 10/27/2014] [Accepted: 10/29/2014] [Indexed: 11/25/2022]
Abstract
Corynebacterium glutamicum has been engineered for production of the polyamide monomer putrescine or 1,4-diaminobutane. Here, N-acetylputrescine was shown to be a significant by-product of putrescine production by recombinant putrescine producing C. glutamicum strains. A systematic gene deletion approach of 18 (putative) N-acetyltransferase genes revealed that the cg1722 gene product was responsible for putrescine acetylation. The encoded enzyme was purified and characterized as polyamine N-acetyltransferase. The enzyme accepted acetyl-CoA and propionyl-CoA as donors for acetylation of putrescine and other diamines as acceptors, but showed highest catalytic efficiency with the triamine spermidine and the tetraamine spermine and, hence, was named SnaA. Upon deletion of snaA in the putrescine producing strain PUT21, no acteylputrescine accumulated, but about 41% more putrescine as compared to the parent strain. Moreover, a transcriptome approach identified increased expression of the cgmAR operon encoding a putative permease and a transcriptional TetR-family repressor upon induction of putrescine production in C. glutamicum PUT21. CgmR is known to bind to cgmO upstream of cgmAR and gel mobility shift experiments with purified CgmR revealed that putrescine and other diamines perturbed CgmR-cgmO complex formation, but not migration of free cgmO DNA. Deletion of the repressor gene cgmR resulted in expression changes of a number of genes and increased putrescine production of C. glutamicum PUT21 by 19% as compared to the parent strain. Overexpression of the putative transport gene cgmA increased putrescine production by 24% as compared to the control strain. However, cgmA overexpression in PUT21ΔsnaA did not further improve putrescine production, hence, the beneficial effects of both targets were not synergistic at the highest described yield of 0.21 g g(-1).
Collapse
Affiliation(s)
- Anh Q D Nguyen
- Chair of Genetics of Prokaryotes, Faculty of Biology & CeBiTec, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Jens Schneider
- Chair of Genetics of Prokaryotes, Faculty of Biology & CeBiTec, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Volker F Wendisch
- Chair of Genetics of Prokaryotes, Faculty of Biology & CeBiTec, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany.
| |
Collapse
|
41
|
Srisucharitpanit K, Yao M, Promdonkoy B, Chimnaronk S, Tanaka I, Boonserm P. Crystal structure of BinB: A receptor binding component of the binary toxin from Lysinibacillus sphaericus. Proteins 2014; 82:2703-12. [DOI: 10.1002/prot.24636] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/12/2014] [Accepted: 06/18/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Kanokporn Srisucharitpanit
- Institute of Molecular Biosciences, Mahidol University; Salaya, Phuttamonthon Nakhon Pathom 73170 Thailand
- Faculty of Allied Health Science; Burapha University, Saensook; Muang District Chon Buri 20131 Thailand
| | - Min Yao
- Faculty of Advanced Life Sciences; Hokkaido University; Sapporo 060-0810 Japan
| | - Boonhiang Promdonkoy
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency; 113 Pahonyothin Road, Khlong Nueng Khlong Luang Pathum Thani 12120 Thailand
| | - Sarin Chimnaronk
- Institute of Molecular Biosciences, Mahidol University; Salaya, Phuttamonthon Nakhon Pathom 73170 Thailand
| | - Isao Tanaka
- Faculty of Advanced Life Sciences; Hokkaido University; Sapporo 060-0810 Japan
| | - Panadda Boonserm
- Institute of Molecular Biosciences, Mahidol University; Salaya, Phuttamonthon Nakhon Pathom 73170 Thailand
| |
Collapse
|
42
|
Sun J, Deng Z, Yan A. Bacterial multidrug efflux pumps: mechanisms, physiology and pharmacological exploitations. Biochem Biophys Res Commun 2014; 453:254-67. [PMID: 24878531 DOI: 10.1016/j.bbrc.2014.05.090] [Citation(s) in RCA: 468] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 05/20/2014] [Indexed: 01/11/2023]
Abstract
Multidrug resistance (MDR) refers to the capability of bacterial pathogens to withstand lethal doses of structurally diverse drugs which are capable of eradicating non-resistant strains. MDR has been identified as a major threat to the public health of human being by the World Health Organization (WHO). Among the four general mechanisms that cause antibiotic resistance including target alteration, drug inactivation, decreased permeability and increased efflux, drug extrusion by the multidrug efflux pumps serves as an important mechanism of MDR. Efflux pumps not only can expel a broad range of antibiotics owing to their poly-substrate specificity, but also drive the acquisition of additional resistance mechanisms by lowering intracellular antibiotic concentration and promoting mutation accumulation. Over-expression of multidrug efflux pumps have been increasingly found to be associated with clinically relevant drug resistance. On the other hand, accumulating evidence has suggested that efflux pumps also have physiological functions in bacteria and their expression is subject tight regulation in response to various of environmental and physiological signals. A comprehensive understanding of the mechanisms of drug extrusion, and regulation and physiological functions of efflux pumps is essential for the development of anti-resistance interventions. In this review, we summarize the development of these research areas in the recent decades and present the pharmacological exploitation of efflux pump inhibitors as a promising anti-drug resistance intervention.
Collapse
Affiliation(s)
- Jingjing Sun
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Ziqing Deng
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Aixin Yan
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region.
| |
Collapse
|
43
|
Modulation of Bacterial Multidrug Resistance Efflux Pumps of the Major Facilitator Superfamily. INTERNATIONAL JOURNAL OF BACTERIOLOGY 2013; 2013. [PMID: 25750934 PMCID: PMC4347946 DOI: 10.1155/2013/204141] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial infections pose a serious public health concern, especially when an infectious disease has a multidrug resistant causative agent. Such multidrug resistant bacteria can compromise the clinical utility of major chemotherapeutic antimicrobial agents. Drug and multidrug resistant bacteria harbor several distinct molecular mechanisms for resistance. Bacterial antimicrobial agent efflux pumps represent a major mechanism of clinical resistance. The major facilitator superfamily (MFS) is one of the largest groups of solute transporters to date and includes a significant number of bacterial drug and multidrug efflux pumps. We review recent work on the modulation of multidrug efflux pumps, paying special attention to those transporters belonging primarily to the MFS.
Collapse
|
44
|
Hashimoto K, Ogawa W, Nishioka T, Tsuchiya T, Kuroda T. Functionally cloned pdrM from Streptococcus pneumoniae encodes a Na(+) coupled multidrug efflux pump. PLoS One 2013; 8:e59525. [PMID: 23555691 PMCID: PMC3608713 DOI: 10.1371/journal.pone.0059525] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 02/15/2013] [Indexed: 11/29/2022] Open
Abstract
Multidrug efflux pumps play an important role as a self-defense system in bacteria. Bacterial multidrug efflux pumps are classified into five families based on structure and coupling energy: resistance−nodulation−cell division (RND), small multidrug resistance (SMR), major facilitator (MF), ATP binding cassette (ABC), and multidrug and toxic compounds extrusion (MATE). We cloned a gene encoding a MATE-type multidrug efflux pump from Streptococcus pneumoniae R6, and designated it pdrM. PdrM showed sequence similarity with NorM from Vibrio parahaemolyticus, YdhE from Escherichia coli, and other bacterial MATE-type multidrug efflux pumps. Heterologous expression of PdrM let to elevated resistance to several antibacterial agents, norfloxacin, acriflavine, and 4′,6-diamidino-2-phenylindole (DAPI) in E. coli KAM32 cells. PdrM effluxes acriflavine and DAPI in a Na+- or Li+-dependent manner. Moreover, Na+ efflux via PdrM was observed when acriflavine was added to Na+-loaded cells expressing pdrM. Therefore, we conclude that PdrM is a Na+/drug antiporter in S. pneumoniae. In addition to pdrM, we found another two genes, spr1756 and spr1877,that met the criteria of MATE-type by searching the S. pneumoniae genome database. However, cloned spr1756 and spr1877 did not elevate the MIC of any of the investigated drugs. mRNA expression of spr1756, spr1877, and pdrM was detected in S. pneumoniae R6 under laboratory growth conditions. Therefore, spr1756 and spr1877 are supposed to play physiological roles in this growth condition, but they may be unrelated to drug resistance.
Collapse
Affiliation(s)
- Kohei Hashimoto
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Tsushima, Okayama, Japan
| | - Wakano Ogawa
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Tsushima, Okayama, Japan
- * E-mail:
| | - Toshihiro Nishioka
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Tsushima, Okayama, Japan
| | - Tomofusa Tsuchiya
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Tsushima, Okayama, Japan
| | - Teruo Kuroda
- Department of Molecular Microbiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Tsushima, Okayama, Japan
| |
Collapse
|
45
|
Ibarra JA, Pérez-Rueda E, Carroll RK, Shaw LN. Global analysis of transcriptional regulators in Staphylococcus aureus. BMC Genomics 2013; 14:126. [PMID: 23442205 PMCID: PMC3616918 DOI: 10.1186/1471-2164-14-126] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 02/12/2013] [Indexed: 02/01/2023] Open
Abstract
Background Staphylococcus aureus is a widely distributed human pathogen capable of infecting almost every ecological niche of the host. As a result, it is responsible for causing many different diseases. S. aureus has a vast array of virulence determinants whose expression is modulated by an intricate regulatory network, where transcriptional factors (TFs) are the primary elements. In this work, using diverse sequence analysis, we evaluated the repertoire of TFs and sigma factors in the community-associated methicillin resistant S. aureus (CA-MRSA) strain USA300-FPR3757. Results A total of 135 TFs and sigma factors were identified and classified into 36 regulatory families. From these around 43% have been experimentally characterized to date, which demonstrates the significant work still at hand to unravel the regulatory network in place for this important pathogen. A comparison of the TF repertoire of S. aureus against 1209 sequenced bacterial genomes was carried out allowing us to identify a core set of orthologous TFs for the Staphylococacceae, and also allowing us to assign potential functions to previously uncharacterized TFs. Finally, the USA300 TFs were compared to those in eleven other S. aureus strains including: Newman, COL, JH1, JH9, MW2, Mu3, Mu50, N315, RF122, MRSA252 and MSSA476. We identify conserved TFs among these strains and suggest possible regulatory interactions. Conclusions The analysis presented herein highlights the complexity of regulatory networks in S. aureus strains, identifies key conserved TFs among the Staphylococacceae, and offers unique insights into several as yet uncharacterized TFs.
Collapse
Affiliation(s)
- Jose Antonio Ibarra
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, ISA 2015, Tampa, FL 33620-5150, USA.
| | | | | | | |
Collapse
|
46
|
Biocide tolerance in bacteria. Int J Food Microbiol 2013; 162:13-25. [PMID: 23340387 DOI: 10.1016/j.ijfoodmicro.2012.12.028] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 10/24/2012] [Accepted: 12/15/2012] [Indexed: 02/07/2023]
Abstract
Biocides have been employed for centuries, so today a wide range of compounds showing different levels of antimicrobial activity have become available. At the present time, understanding the mechanisms of action of biocides has also become an important issue with the emergence of bacterial tolerance to biocides and the suggestion that biocide and antibiotic resistance in bacteria might be linked. While most of the mechanisms providing antibiotic resistance are agent specific, providing resistance to a single antimicrobial or class of antimicrobial, there are currently numerous examples of efflux systems that accommodate and, thus, provide tolerance to a broad range of structurally unrelated antimicrobials, both antibiotics and biocides. If biocide tolerance becomes increasingly common and it is linked to antibiotic resistance, not only resistant (even multi-resistant) bacteria could be passed along the food chain, but also there are resistance determinants that can spread and lead to the emergence of new resistant microorganisms, which can only be detected and monitored when the building blocks of resistance traits are understood on the molecular level. This review summarizes the main advances reached in understanding the mechanism of action of biocides, the mechanisms of bacterial resistance to both biocides and antibiotics, and the incidence of biocide tolerance in bacteria of concern to human health and the food industry.
Collapse
|
47
|
|
48
|
Abstract
The dramatic changes in the environmental conditions that organisms encountered during evolution and adaptation to life in specific niches, have influenced intracellular and extracellular metal ion contents and, as a consequence, the cellular ability to sense and utilize different metal ions. This metal-driven differentiation is reflected in the specific panels of metal-responsive transcriptional regulators found in different organisms, which finely tune the intracellular metal ion content and all metal-dependent processes. In order to understand the processes underlying this complex metal homeostasis network, the study of the molecular processes that determine the protein-metal ion recognition, as well as how this event is transduced into a transcriptional output, is necessary. This chapter describes how metal ion binding to specific proteins influences protein interaction with DNA and how this event can influence the fate of genetic expression, leading to specific transcriptional outputs. The features of representative metal-responsive transcriptional regulators, as well as the molecular basis of metal-protein and protein-DNA interactions, are discussed on the basis of the structural information available. An overview of the recent advances in the understanding of how these proteins choose specific metal ions among the intracellular metal ion pool, as well as how they allosterically respond to their effector binding, is given.
Collapse
|
49
|
Zharov IA, Gelfand MS, Kazakov AE. Regulation of multidrug resistance genes by transcription factors of the BltR subfamily. Mol Biol 2011. [DOI: 10.1134/s002689331103023x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Torres C, Galián C, Freiberg C, Fantino JR, Jault JM. The YheI/YheH heterodimer from Bacillus subtilis is a multidrug ABC transporter. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:615-22. [DOI: 10.1016/j.bbamem.2008.12.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 12/12/2008] [Accepted: 12/22/2008] [Indexed: 12/12/2022]
|