1
|
Franklin HC, Makhlouf R, Ha AD, Bataglioli RA, Baker ZR, Murphy SA, Jirsa H, Heuler J, Southard T, Aylward FO, Hsu BB. A bacteriophage-conditional mouse model reveals the impact of phages within a conventionally colonized gut microbiota. Cell Host Microbe 2025; 33:745-758.e6. [PMID: 40300596 DOI: 10.1016/j.chom.2025.04.002] [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: 08/26/2024] [Revised: 02/14/2025] [Accepted: 04/03/2025] [Indexed: 05/01/2025]
Abstract
The significance of bacteriophages in the gut microbiota remains poorly understood due, in part, to an absence of an animal model that allows for comparative study of conditions with or without phages while retaining the microbial diversity attained by conventional colonization. We describe a mouse model that uses a broadly available chemical compound, acriflavine, to preferentially deplete virulent phages from the gut without significantly impacting gut bacteria. We then show that gut phage density can be reconstituted by oral gavage. Using this bacteriophage-conditional (BaCon) mouse model, we reveal that while phages have comparatively minimal impact during equilibrium conditions, they increase the potency of ampicillin against commensal gut bacteria. Collectively, our work presents an animal model that can be leveraged to conditionally study the role of phages in complex, physiologically relevant systems and further identifies virulent gut phages as potential sources of bacterial variability during major perturbations.
Collapse
Affiliation(s)
- Hollyn C Franklin
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Rita Makhlouf
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Anh D Ha
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Rogerio A Bataglioli
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zachary R Baker
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Sydney A Murphy
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Hannah Jirsa
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Joshua Heuler
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Teresa Southard
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Frank O Aylward
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Bryan B Hsu
- Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
2
|
Yang QE, Gao JT, Zhou SG, Walsh TR. Cutting-edge tools for unveiling the dynamics of plasmid-host interactions. Trends Microbiol 2025; 33:496-509. [PMID: 39843314 DOI: 10.1016/j.tim.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/24/2025]
Abstract
The plasmid-mediated transfer of antibiotic resistance genes (ARGs) in complex microbiomes presents a significant global health challenge. This review examines recent technological advancements that have enabled us to move beyond the limitations of culture-dependent detection of conjugation and have enhanced our ability to track and understand the movement of ARGs in real-world scenarios. We critically assess the applications of single-cell sequencing, fluorescence-based techniques and advanced high-throughput chromatin conformation capture (Hi-C) approaches in elucidating plasmid-host interactions at unprecedented resolution. We also evaluate emerging techniques such as CRISPR-based phage engineering and discuss their potential for developing targeted strategies to curb ARG dissemination. Emerging data derived from these technologies have challenged our previous paradigms on plasmid-host compatibility and an awareness of an emerging uncharted realm for ARGs.
Collapse
Affiliation(s)
- Qiu E Yang
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jiang Tao Gao
- Key BioAI Synthetica Lab for Natural Product Drug Discovery, National and Local United Engineering Laboratory of Natural Biotoxin, College of Bee and Biomedical Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Shun Gui Zhou
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Timothy R Walsh
- Ineos Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford OX1 3RE, UK.
| |
Collapse
|
3
|
Kerek Á, Román I, Szabó Á, Kovács D, Kardos G, Kovács L, Jerzsele Á. Antibiotic resistance genes in Escherichia coli - literature review. Crit Rev Microbiol 2025:1-35. [PMID: 40249005 DOI: 10.1080/1040841x.2025.2492156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Antimicrobial resistance threatens humans and animals worldwide and is recognized as one of the leading global public health issues. Escherichia coli (E. coli) has an unquestionable role in carrying and transmitting antibiotic resistance genes (ARGs), which in many cases are encoded on plasmids or phage, thus creating the potential for horizontal gene transfer. In this literature review, the authors summarize the major antibiotic resistance genes occurring in E. coli bacteria, through the major antibiotic classes. The aim was not only listing the resistance genes against the clinically relevant antibiotics, used in the treatment of E. coli infections, but also to cover the entire resistance gene carriage in E. coli, providing a more complete picture. We started with the long-standing antibiotic groups (beta-lactams, aminoglycosides, tetracyclines, sulfonamides and diaminopyrimidines), then moved toward the newer groups (phenicols, peptides, fluoroquinolones, nitrofurans and nitroimidazoles), and in every group we summarized the resistance genes grouped by the mechanism of their action (enzymatic inactivation, antibiotic efflux, reduced permeability, etc.). We observed that the frequency of antibiotic resistance mechanisms changes in the different groups.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - István Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Dóra Kovács
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Gábor Kardos
- One Health Institute, University of Debrecen, Debrecen, Hungary
- National Public Health Center, Budapest, Hungary
- Department of Gerontology, Faculty of Health Sciences, University of Debrecen, Nyíregyháza, Hungary
| | - László Kovács
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
- Department of Animal Hygiene, Herd Health and Mobile Clinic, University of Veterinary Medicine, Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| |
Collapse
|
4
|
Lloyd G, Stephens E, Di Maio A, Thomas C. Activation, incompatibility, and displacement of FIB replicons in E. coli. Nucleic Acids Res 2025; 53:gkaf275. [PMID: 40207625 PMCID: PMC11983097 DOI: 10.1093/nar/gkaf275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Multi-replicon sex-factor F is the archetype of the largest plasmid group in clinical Enterobacteriaceae. Such plasmids spread antimicrobial resistance (AMR) and virulence functions in commensal bacteria of humans and animals. Displacing (curing) these plasmids by blocking replication and neutralizing addiction is successful with the curing cassette on a high-copy-number vector but, with conjugative IncP-1 plasmid RK2 as vector for our "anti-F cassette", displacement of F'prolac is inefficient unless curing-plasmid copy-number is raised 1.5- to 2-fold. Here we report that it is the anti-FIB segment, originating from FIB-FII plasmid pO157, which needs potentiation. We show that the FIB replicon in F (F-FIB) is defective due to a sub-optimal rep ribosome-binding-site (rbs) but can be activated by FIB-Rep protein expressed from our anti-FIB segment joined to RK2. Deleting FIB-rep from the anti-F cassette removed the need for potentiation. A pO157-FIB single-replicon plasmid was displaced efficiently by the complete anti-F cassette without potentiation, but an F-FIB plasmid, mutated to have a pO157-like rep rbs, was not, indicating that sequence divergence between F and pO157 FIB replicons has weakened their negative cross-reactivity. Thus, raising vector copy-number slightly may be sufficient to increase displacement of plasmids similar but not identical to the sequences in the curing cassette.
Collapse
Affiliation(s)
- Georgina S Lloyd
- School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Elton R Stephens
- School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Alessandro Di Maio
- School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Christopher M Thomas
- School of Biosciences and Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
5
|
Tsoi R, Son HI, Hamrick GS, Tang K, Bethke JH, Lu J, Maddamsetti R, You L. A predatory gene drive for targeted control of self-transmissible plasmids. SCIENCE ADVANCES 2025; 11:eads4735. [PMID: 40173243 PMCID: PMC11963995 DOI: 10.1126/sciadv.ads4735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Suppressing plasmid transfer in microbial communities has profound implications due to the role of horizontal gene transfer (HGT) in spreading and maintaining diverse functional traits such as metabolic functions, virulence factors, and antibiotic resistance. However, existing tools for inhibiting HGT are limited in their modes of delivery, efficacy, and scalability. Here, we present a versatile denial-of-spread (DoS) strategy to target and eliminate specific conjugative plasmids. Our strategy exploits retrotransfer, whereby an engineered DoS plasmid is introduced into host cells containing a target plasmid. Acting as a predatory gene drive, DoS propagates itself at the expense of the target plasmid, through competition or active elimination. Once the target plasmid is eradicated, DoS is removed via induced plasmid suicide, resulting in a community containing neither plasmid. The strategy is tunable and scalable for various conjugative plasmids, different mechanisms of plasmid inheritance interruption, and diverse environmental contexts. DoS represents a new tool for precise control of gene persistence in microbial communities.
Collapse
Affiliation(s)
- Ryan Tsoi
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Hye-In Son
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Grayson S. Hamrick
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Katherine Tang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Jonathan H. Bethke
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708, USA
| | - Jia Lu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Rohan Maddamsetti
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708, USA
- Center for Quantitative Biodesign, Duke University, Durham, NC 27708, USA
| |
Collapse
|
6
|
Sadanov AK, Baimakhanova BB, Orasymbet SE, Ratnikova IA, Turlybaeva ZZ, Baimakhanova GB, Amitova AA, Omirbekova AA, Aitkaliyeva GS, Kossalbayev BD, Belkozhayev AM. Engineering Useful Microbial Species for Pharmaceutical Applications. Microorganisms 2025; 13:599. [PMID: 40142492 PMCID: PMC11944651 DOI: 10.3390/microorganisms13030599] [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: 02/11/2025] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Microbial engineering has made a significant breakthrough in pharmaceutical biotechnology, greatly expanding the production of biologically active compounds, therapeutic proteins, and novel drug candidates. Recent advancements in genetic engineering, synthetic biology, and adaptive evolution have contributed to the optimization of microbial strains for pharmaceutical applications, playing a crucial role in enhancing their productivity and stability. The CRISPR-Cas system is widely utilized as a precise genome modification tool, enabling the enhancement of metabolite biosynthesis and the activation of synthetic biological pathways. Additionally, synthetic biology approaches allow for the targeted design of microorganisms with improved metabolic efficiency and therapeutic potential, thereby accelerating the development of new pharmaceutical products. The integration of artificial intelligence (AI) and machine learning (ML) plays a vital role in further advancing microbial engineering by predicting metabolic network interactions, optimizing bioprocesses, and accelerating the drug discovery process. However, challenges such as the efficient optimization of metabolic pathways, ensuring sustainable industrial-scale production, and meeting international regulatory requirements remain critical barriers in the field. Furthermore, to mitigate potential risks, it is essential to develop stringent biocontainment strategies and implement appropriate regulatory oversight. This review comprehensively examines recent innovations in microbial engineering, analyzing key technological advancements, regulatory challenges, and future development perspectives.
Collapse
Affiliation(s)
- Amankeldi K. Sadanov
- LLP “Research and Production Center for Microbiology and Virology”, Almaty 050010, Kazakhstan; (A.K.S.); (B.B.B.); (S.E.O.); (I.A.R.); (Z.Z.T.)
| | - Baiken B. Baimakhanova
- LLP “Research and Production Center for Microbiology and Virology”, Almaty 050010, Kazakhstan; (A.K.S.); (B.B.B.); (S.E.O.); (I.A.R.); (Z.Z.T.)
| | - Saltanat E. Orasymbet
- LLP “Research and Production Center for Microbiology and Virology”, Almaty 050010, Kazakhstan; (A.K.S.); (B.B.B.); (S.E.O.); (I.A.R.); (Z.Z.T.)
| | - Irina A. Ratnikova
- LLP “Research and Production Center for Microbiology and Virology”, Almaty 050010, Kazakhstan; (A.K.S.); (B.B.B.); (S.E.O.); (I.A.R.); (Z.Z.T.)
| | - Zere Z. Turlybaeva
- LLP “Research and Production Center for Microbiology and Virology”, Almaty 050010, Kazakhstan; (A.K.S.); (B.B.B.); (S.E.O.); (I.A.R.); (Z.Z.T.)
| | - Gul B. Baimakhanova
- LLP “Research and Production Center for Microbiology and Virology”, Almaty 050010, Kazakhstan; (A.K.S.); (B.B.B.); (S.E.O.); (I.A.R.); (Z.Z.T.)
| | - Aigul A. Amitova
- Department of Chemical and Biochemical Engineering, Geology and Oil-Gas Business Institute Named After K. Turyssov, Satbayev University, Almaty 050043, Kazakhstan; (G.S.A.); (A.M.B.)
| | - Anel A. Omirbekova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
| | - Gulzat S. Aitkaliyeva
- Department of Chemical and Biochemical Engineering, Geology and Oil-Gas Business Institute Named After K. Turyssov, Satbayev University, Almaty 050043, Kazakhstan; (G.S.A.); (A.M.B.)
| | - Bekzhan D. Kossalbayev
- Department of Chemical and Biochemical Engineering, Geology and Oil-Gas Business Institute Named After K. Turyssov, Satbayev University, Almaty 050043, Kazakhstan; (G.S.A.); (A.M.B.)
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
- Ecology Research Institute, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan
| | - Ayaz M. Belkozhayev
- Department of Chemical and Biochemical Engineering, Geology and Oil-Gas Business Institute Named After K. Turyssov, Satbayev University, Almaty 050043, Kazakhstan; (G.S.A.); (A.M.B.)
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
| |
Collapse
|
7
|
Hu Y, Tang R, Jin S, Guan J, Meng X, Dan Z, Wang R, Ou HY, Lu J. Molecular characterization of ST15 carbapenem-resistant Klebsiella pneumoniae isolated in a single patient. J Glob Antimicrob Resist 2025; 40:72-80. [PMID: 39631626 DOI: 10.1016/j.jgar.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/03/2024] [Accepted: 11/10/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The carbapenem-resistant Klebsiella pneumoniae (CRKP) poses a serious threat to antibiotic applicability and public health. During treatment, K. pneumoniae (KP) frequently exhibits shifts in drug-resistant phenotypes, complicating clinical treatment as it transitions from sensitivity to resistance. In this study, we analysed the clinical and molecular characteristics of drug resistance changes in KP strains isolated from a single patient, and the potential mechanisms underlying these resistance changes. METHODS Antimicrobial susceptibility test and string test were conducted to evaluate the resistant and virulent characterization of the strains. Pulsed-field gel electrophoresis (PFGE) was used to investigate the homology relationship between the strains. The whole genome sequencing and phylogenetic analysis of 9 representative isolates was also performed. The transfer ability of the drug-resistant plasmid was studied by plasmid conjugation experiment. The transconjugants were verified by polymerase chain reaction amplification of specific genes, antimicrobial susceptibility test and PFGE. RESULTS Our results revealed that 9 KP strains, isolated from the same patient, exhibited 'resistance-sensitivity-resistance-sensitivity' alternately to carbapenems. The differences in DNA fingerprint bands among the nine KP isolates were ≤3, which can be classified as the same PFGE type. Phylogenetic analysis showed that these 9 strains constituted a distinct branch within the phylogenetic tree. All nine KP strains belonged to the ST15-KL19 clone. Six of the strains were classified as CRKP, all of which carried 11 drug resistance genes: oqxB, oqxA, fosA6, aac(3)-lld, blaSHV-28, blaKPC-2, blaOXA-1, mph(A), tet(A), catB3 and aac(6')-lb-cr, mediating drug resistance to quinolones, fosfomycin, aminoglycosides, β-lactam, carbapenems, macrolides and chloramphenicol, belonging to multi-drug resistant bacteria. The carbapenem-resistant plasmid p2-KP3762-1 was found to transfer within species, from CRKP to hypervirulent KPRJF293HA, carbapenem-sensitive KP KP3657 and Escherichia coli C600 at a frequency of (1.19 ± 1.58) ×10-6, (1.09 ± 1.38) ×10-7 and (10.9 ± 9.53) ×10-6 respectively, resulting in the dissemination of carbapenem resistance genes. CONCLUSIONS In this study, KP strains isolated from a single patient exhibited an alternating phenotype of resistant-sensitive-resistant-sensitive to carbapenems. The 9 KP isolates share a high degree of genetic similarity. The plasmid p2-KP3762-1, harbouring the carbapenem resistance gene blaKPC-2, may undergo inter-strain and inter-clone transfer via conjugation in the patient during treatment. Furthermore, our findings suggest that the pathogens in this patient are likely to have a common ancestral origin.
Collapse
Affiliation(s)
- Yongjin Hu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Tang
- Department of Clinical Laboratory, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanshan Jin
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahao Guan
- School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxiao Meng
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zengpeijie Dan
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruilan Wang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Yu Ou
- School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Lu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Critical Care Medicine, Shanghai United Family Hospital, Shanghai, China.
| |
Collapse
|
8
|
Sathish Kumar K, Visnuvinayagam S, Teena G, Elavarasan K, Bindu J, Balange AK, Sivaranjani R, Narasimhamurthy L. Biochemical, Antioxidant, and Antimicrobial Profiling of Essential Oils of Indian Origin for Culinary Applications. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2024; 2024:9326683. [PMID: 39759801 PMCID: PMC11698606 DOI: 10.1155/ijfo/9326683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 11/06/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025]
Abstract
This study investigated the biochemical composition and tested the antioxidant and antimicrobial properties of four Indian-origin essential oils (EOs)-ginger, garlic, clove, and eucalyptus-to evaluate their potential for culinary applications. Gas chromatography-mass spectrometry (GC-MS) analysis was used to identify the chemical constituents of EOs. Antioxidant assays such as 2,2-diphenyl-1-picrylhydrazyl (DPPH) and ferric reducing antioxidant power (FRAP) and antimicrobial assays such as Agar well diffusion, minimum inhibitory concentration (MIC), and minimum bactericidal concentration (MBC) were carried out. In GC-MS analysis, α-zingiberene (28.01%), eugenol (70.12%), 1,8-cineole (52.95%), and allyl polysulfides and terpenes were the most dominant compounds in ginger, clove, eucalyptus, and garlic EOs, respectively, which are responsible for their antioxidant and antimicrobial properties. Based on the antioxidant assays, clove EO exhibited the strongest antioxidant activities in both DPPH (70.84 ± 3.95%) and FRAP (142.29 ± 1.05 μ mol Fe (II) g-1) even at 5% level, suggesting its potential to inhibit lipid peroxidation, a process linked to oxidative stress in food. The antimicrobial study demonstrated the potential of EOs against foodborne pathogens, particularly against methicillin-resistant S. aureus (MRSA), which reveals their potential against multidrug-resistant bacteria. Among them, clove EO demonstrated the most potent antimicrobial activity against both Gram-negative and Gram-positive bacteria, with notable activity against MRSA with an inhibition zone of 41.33 ± 0.57 mm. This strong antimicrobial activity of clove EO was directly correlated with its total phenolic content (375.91 ± 14.21 mg phenols 100 g-1 at 5% level). The results indicated that clove EO stands out for its strong antioxidant and antimicrobial properties, particularly against multidrug-resistant pathogens like MRSA. These findings suggest clove EO could be a promising natural alternative to synthetic preservatives and antibiotics in culinary applications, helping to preserve food and combat resistant bacteria.
Collapse
Affiliation(s)
- K. Sathish Kumar
- Fish Processing Division, ICAR-Central Institute of Fisheries Technology, Cochin, Kerala, India
- Fisheries Resources, Harvest & Post-Harvest Management Division, ICAR-Central Institute of Fisheries Education, Mumbai, Maharashtra, India
| | - S. Visnuvinayagam
- Microbiology, Fermentation and Biotechnology Division, ICAR-Central Institute of Fisheries Technology, Cochin, Kerala, India
| | - G. Teena
- Microbiology, Fermentation and Biotechnology Division, ICAR-Central Institute of Fisheries Technology, Cochin, Kerala, India
| | - K. Elavarasan
- Fish Processing Division, ICAR-Central Institute of Fisheries Technology, Cochin, Kerala, India
| | - J. Bindu
- Fish Processing Division, ICAR-Central Institute of Fisheries Technology, Cochin, Kerala, India
| | - Amjad K. Balange
- Fisheries Resources, Harvest & Post-Harvest Management Division, ICAR-Central Institute of Fisheries Education, Mumbai, Maharashtra, India
| | - R. Sivaranjani
- Crop Production and Post-Harvest Technology, ICAR-Indian Institute of Spices Research, Kozhikode, Kerala, India
| | | |
Collapse
|
9
|
Todorov SD, Lima JMS, Bucheli JEV, Popov IV, Tiwari SK, Chikindas ML. Probiotics for Aquaculture: Hope, Truth, and Reality. Probiotics Antimicrob Proteins 2024; 16:2007-2020. [PMID: 38801620 DOI: 10.1007/s12602-024-10290-8] [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] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
The use of microorganisms as beneficial crops for human and animal health has been studied for decades, and these microorganisms have been in practical use for quite some time. Nowadays, in addition to well-known examples of beneficial properties of lactic acid bacteria, bifidobacteria, selected Bacillus spp., and yeasts, there are several other bacteria considered next-generation probiotics that have been proposed to improve host health. Aquaculture is a rapidly growing area that provides sustainable proteins for consumption by humans and other animals. Thus, there is a need to develop new technologies for the production practices associated with cleaner and environment-friendly approaches. It is a well-known fact that proper selection of the optimal probiotics for use in aquaculture is an essential step to ensure effectiveness and safety. In this critical review, we discuss the evaluation of host-specific probiotics in aquaculture, challenges in using probiotics in aquaculture, methods to improve the survival of probiotics under different environmental conditions, technological approach to improving storage, and delivery along with possible negative consequences of using probiotics in aquaculture. A critical analysis of the identified challenges for the use of beneficial microbes in aquaculture will help in sustainable aquafarming, leading to improved agricultural practices with a clear aim to increase protein production.
Collapse
Affiliation(s)
- Svetoslav Dimitrov Todorov
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, 05508-000, SP, Brazil.
- CISAS-Center for Research and Development in Agrifood Systems and Sustainability, Instituto Politécnico de Viana Do Castelo, 4900-347, Viana Do Castelo, Portugal.
| | - Joao Marcos Scafuro Lima
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, 05508-000, SP, Brazil
| | - Jorge Enrique Vazquez Bucheli
- Facultad de Medicina Veterinaria y Zootecnia, Departamento de Bioestadistica y Genetica, Universidad Nacional Autonoma de Mexico, Av. Universidad 3000, C.P. 04510, Mexico City, Mexico
| | - Igor Vitalievich Popov
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1, Rostov-On-Don 344002, Rostov, Russia
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Sirius 354340, Krasnodar Region, Russia
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak 124001, Haryana, India
| | - Michael Leonidas Chikindas
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1, Rostov-On-Don 344002, Rostov, Russia
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, the State University of New Jersey, RutgersNew Brunswick, NJ 08901, USA
- I. M. Sechenov First Moscow State Medical University, Moscow 119435, Russia
| |
Collapse
|
10
|
Park CH, Yang H, Kim S, Yun CS, Jang BC, Hong YJ, Park DS. Comparison of Plasmid Curing Efficiency across Five Lactic Acid Bacterial Species. J Microbiol Biotechnol 2024; 34:2385-2395. [PMID: 39403731 PMCID: PMC11637826 DOI: 10.4014/jmb.2406.06003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/12/2024] [Accepted: 08/30/2024] [Indexed: 11/29/2024]
Abstract
With the recent stringent criteria for antibiotic susceptibility in probiotics, the presence of antibiotic resistance genes and plasmids associated with their transfer has become a limiting factor in the approval of probiotics. The need to remove genes related to antibiotic resistance and virulence through plasmid curing for the authorization of probiotics is increasing. In this study, we investigated the curing efficiency of ethidium bromide, acridine orange, and novobiocin at different concentrations and durations in five strains of plasmid-bearing lactic acid bacteria and examined the curing characteristics in each strain. Limosibacillus reuteri and Lacticaseibacillus paracasei exhibited curing efficiencies ranging from 5% to 44% following treatment with ethidium bromide (10-50 μg/ml) for 24-72 h, while Lactobacillus gasseri showed the highest efficiency at 14% following treatment with 10 μg/ml novobiocin for 24 h. Lactiplantibacillus plantarum, which harbors two or more plasmids, demonstrated curing efficiencies ranging from 1% to 8% after an additional 72-h treatment of partially cured strains with 10 μg/ml novobiocin. Plasmid curing in strains with larger plasmids exhibited lower efficiencies and required longer durations. In strains harboring two or more plasmids, a relatively low curing efficiency with a single treatment and a high frequency of false positives, wherein recovery occurred after curing, were observed. Although certain strains exhibited altered susceptibilities to specific antibiotics after curing, these outcomes could not be attributed to the loss of antibiotic resistance genes. Furthermore, the genomic data from the cured strains revealed minimal changes throughout the genome that did not lead to gene mutations.
Collapse
Affiliation(s)
- Chan-Hyeok Park
- Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Haneol Yang
- Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea
| | - Seunghyun Kim
- Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Chan-Seok Yun
- Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea
| | - Byung-Chun Jang
- Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Yeong-Jin Hong
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Doo-Sang Park
- Korean Collection for Type Cultures (KCTC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56212, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
11
|
Gudeta DD, Zhao S, Aljahdali N, Foley SL. Coupling antitoxins and blue/white screening with parAB/resolvase mutation as a strategy for Salmonella spp. plasmid curing. Microbiol Spectr 2024; 12:e0122024. [PMID: 39315784 PMCID: PMC11537010 DOI: 10.1128/spectrum.01220-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Despite the dissemination of multidrug resistance plasmids, including those carrying virulence genes in Salmonella spp., efficient plasmid curing tools are lacking. Plasmid partitioning and multimer resolution systems are attractive targets for plasmid cure. However, plasmid curing strategies targeting these systems are often hindered by the host addiction system through a process known as post-segregation killing. Here, we developed vector tools that can mutate the above systems while replenishing short-lived antitoxins. Cloning was performed using Gibson assembly. parAB or resolvase (res) genes on Incompatibility Group (Inc)FIB, IncA/C, IncX4, and plasmids carried by Salmonella species were deleted by first knocking in the N-terminal ß-galactosidase encoding gene (bgaB), followed by in-frame insertion of its C-terminal region using pDG1 and pDG2 vectors, respectively. pDG1 was used as a backbone to develop a vector, designated as pDG-At, expressing 13 antitoxins driven by strong promoters. Plasmid curing was achieved by transforming pDG-At to parAB or res mutants followed by blue-white screening and PCR; however, parAB mutant isolation with this method was low and often non-reproducible. To elucidate whether the prior presence of pDG-At in cells improves viable mutant isolation, we re-constructed pDG-At, designated as pDG-Atπ, using a vector with the R6Kϒ origin of replication with its π-factor required for replication under araBAD promoter. Results showed that pDG-Atπ can replicate in the absence of arabinose but can be cured by growing cells in glucose-rich media. Next, we repeated IncFIB's parAB deletion using pDG1 but in cells carrying pDG-Atπ. Many white colonies were detected on X-Gal-supplemented media but none of them carried the target parA mutation; however, ~80% of the white colonies lost IncFIB plasmid, while the others retained the wild-type plasmid. Similar results were obtained for IncX4 plasmid curing but also found that this method was not reproducible as the white colonies obtained after allelic replacement did not always result in plasmid curing or mutant isolation. This is the first report describing a simple blue/white screening method for plasmid curing that can avoid laborious screening procedures. IMPORTANCE Plasmids play an important role in bacterial physiology, adaptation, evolution, virulence, and antibiotic resistance. An in-depth study of these roles partly depends on the generation of plasmid-free cells. This study shows that vector tools that target genes required for plasmid stability in the presence of an antitoxin-expressing helper plasmid are a viable approach to cure specific plasmids. Expression of bgaB from target plasmids can greatly facilitate visual detection of plasmid cured colonies avoiding time-consuming screening procedures. This approach can be refined for the development of a universal plasmid curing system that can be used to generate plasmid-free cells in other human bacterial pathogens including Gram positives and Gram negatives.
Collapse
Affiliation(s)
- Dereje D. Gudeta
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Shaohua Zhao
- Office of Applied Science, Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Nesreen Aljahdali
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Steven L. Foley
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| |
Collapse
|
12
|
Yen KK, Terlecky AJ, Hao M, Cienfuegos V, Rojtman A, Chen L, Kreiswirth BN. Curing of common plasmids in gram-negative bacteria using a Cas9-based conjugative vector. J Microbiol Methods 2024; 226:107047. [PMID: 39303991 DOI: 10.1016/j.mimet.2024.107047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
We report the creation of 17 Escherichia coli strains harboring the conjugative plasmid pLCasCureT with a CRISPR-Cas9 system to surgically "cure" the most common plasmids among Enterobacterales species. This approach can create isogenic pairs of strains to study host-plasmid interactions, correlate plasmid genotype and phenotype, and create plasmid-free cloning strains.
Collapse
Affiliation(s)
- Kelly K Yen
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, United States of America.
| | - Austin J Terlecky
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, United States of America
| | - Mingju Hao
- The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | | | - Albert Rojtman
- Jersey Shore University Medical Center, Neptune, NJ 07753, United States of America
| | - Liang Chen
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, United States of America
| | - Barry N Kreiswirth
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, United States of America
| |
Collapse
|
13
|
Alav I, Buckner MMC. Non-antibiotic compounds associated with humans and the environment can promote horizontal transfer of antimicrobial resistance genes. Crit Rev Microbiol 2024; 50:993-1010. [PMID: 37462915 PMCID: PMC11523920 DOI: 10.1080/1040841x.2023.2233603] [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: 03/08/2023] [Revised: 05/23/2023] [Accepted: 06/30/2023] [Indexed: 02/15/2024]
Abstract
Horizontal gene transfer plays a key role in the global dissemination of antimicrobial resistance (AMR). AMR genes are often carried on self-transmissible plasmids, which are shared amongst bacteria primarily by conjugation. Antibiotic use has been a well-established driver of the emergence and spread of AMR. However, the impact of commonly used non-antibiotic compounds and environmental pollutants on AMR spread has been largely overlooked. Recent studies found common prescription and over-the-counter drugs, artificial sweeteners, food preservatives, and environmental pollutants, can increase the conjugative transfer of AMR plasmids. The potential mechanisms by which these compounds promote plasmid transmission include increased membrane permeability, upregulation of plasmid transfer genes, formation of reactive oxygen species, and SOS response gene induction. Many questions remain around the impact of most non-antibiotic compounds on AMR plasmid conjugation in clinical isolates and the long-term impact on AMR dissemination. By elucidating the role of routinely used pharmaceuticals, food additives, and pollutants in the dissemination of AMR, action can be taken to mitigate their impact by closely monitoring use and disposal. This review will discuss recent progress on understanding the influence of non-antibiotic compounds on plasmid transmission, the mechanisms by which they promote transfer, and the level of risk they pose.
Collapse
Affiliation(s)
- Ilyas Alav
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Michelle M. C. Buckner
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
14
|
Fang M, Zhang R, Wang C, Liu Z, Fei M, Tang B, Yang H, Sun D. Engineering probiotic Escherichia coli Nissle 1917 to block transfer of multiple antibiotic resistance genes by exploiting a type I CRISPR-Cas system. Appl Environ Microbiol 2024; 90:e0081124. [PMID: 39254327 PMCID: PMC11497782 DOI: 10.1128/aem.00811-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Many multidrug-resistant (MDR) bacteria have evolved through the accumulation of antibiotic resistance genes (ARGs). Although the potential risk of probiotics as reservoirs of ARGs has been recognized, strategies for blocking the transfer of ARGs while using probiotics have rarely been explored. The probiotic Escherichia coli Nissle 1917 (EcN) has long been used for treating intestinal diseases. Here, we demonstrate frequent transfer of ARGs into EcN both in vitro and in vivo, raising concerns about its potential risk of accumulating antibiotic resistance. Given that no CRISPR-Cas system was found in natural EcN, we integrated the type I-E CRISPR-Cas3 system derived from E. coli BW25113 into EcN. The engineered EcN was able to efficiently cleave multiple ARGs [i.e., mcr-1, blaNDM-1, and tet(X)] encoding enzymes for degrading last-resort antibiotics. Through co-incubation of EcN expressing Cas3-Cascade and that expressing Cas9, we showed that the growth of the former strain outcompeted the latter strain, demonstrating a better clinical application prospect of EcN expressing the type I-E CRISPR-Cas3 system. In the intestine of a model animal (i.e., zebrafish), the engineered EcN exhibited immunity against the transfer of CRISPR-targeted ARGs. Our work equips EcN with immunity against the transfer of multiple ARGs by exploiting the exogenous type I-E CRISPR-Cas3 system, thereby reducing the risk of the spread of ARGs while using it as a probiotic chassis for generating living therapeutics. IMPORTANCE To reduce the development of antibiotic resistance, probiotics have been considered as a substitute for antibiotics. However, probiotics themselves are reservoirs of antibiotic resistance genes (ARGs). This study introduces a new strategy for limiting the spread of ARGs by engineering the typical probiotic strain Escherichia coli Nissle 1917 (EcN), which has been used for treating intestinal diseases and developed as living therapeutics. We also demonstrate that the type I CRISPR-Cas system imposes a lower growth burden than the type II CRISPR-Cas system, highlighting its promising clinical application potential. Our work not only provides a new strategy for restricting the transfer of ARGs while using probiotics but also enriches the genetic engineering toolbox of EcN, paving the way for the safe use and development of probiotics as living therapeutics.
Collapse
Affiliation(s)
- Mengdie Fang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ruiting Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Chenyu Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Zhizhi Liu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Mingyue Fei
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Biao Tang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hua Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Dongchang Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Khan RT, Sharma V, Khan SS, Rasool S. Prevention and potential remedies for antibiotic resistance: current research and future prospects. Front Microbiol 2024; 15:1455759. [PMID: 39421555 PMCID: PMC11484029 DOI: 10.3389/fmicb.2024.1455759] [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: 06/27/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The increasing threat of antibiotic resistance and shrinking treatment options for infections have pushed mankind into a difficult position. The looming threat of the return of the pre-antibiotic era has caused a sense of urgency to protect and conserve the potency of antibiotic therapy. One of the perverse effects of antibiotic resistance is the dissemination of its causative agents from non-clinically important strains to clinically important strains and vice versa. The popular saying "Prevention is better than cure" is appropriate for tackling antibiotic resistance. On the one hand, new and effective antibiotics are required; on the other hand, better measures for the use of antibiotics, along with increased awareness in the general public related to antibiotic use, are essential. Awareness, especially of appropriate antibiotic use, antibiotic resistance, its dissemination, and potential threats, can help greatly in controlling the use and abuse of antibiotics, and the containment of antibiotic resistance. Antibiotic drugs' effectiveness can be enhanced by producing novel antibiotic analogs or adding adjuvants to current antibiotics. Combinatorial therapy of antibiotics has proven successful in treating multidrug-resistant (MDR) bacterial infections. This review aims to highlight the current global situation of antibiotic resistance and discuss the methods used to monitor, prevent, inhibit, or reverse bacterial resistance mechanisms in the fight against antibiotic resistance.
Collapse
Affiliation(s)
| | | | | | - Shafaq Rasool
- Molecular Biology Lab, School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| |
Collapse
|
16
|
Li G, Han L, Xia LJ, Gao A, Li ZP, Zhou SY, Wan L, Deng Y, Zhou TH, Lu XY, Luo Y, Liang DS, Wu GT, Tang SQ, Lian XL, Ren H, Liao XP, Chen L, Sun J. Waterborne polyurethane nanoparticles incorporating linoleic acid as a potential strategy for controlling antibiotic resistance spread in the mammalian intestine. Mater Today Bio 2024; 28:101181. [PMID: 39221217 PMCID: PMC11364912 DOI: 10.1016/j.mtbio.2024.101181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/10/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Plasmid-mediated conjugative transfer of antibiotic resistance genes (ARGs) within the human and animal intestine represents a substantial global health concern. linoleic acid (LA) has shown promise in inhibiting conjugation in vitro, but its in vivo effectiveness in the mammalian intestinal tract is constrained by challenges in efficiently reaching the target site. Recent advancements have led to the development of waterborne polyurethane nanoparticles for improved drug delivery. In this study, we synthesized four waterborne polyurethane nanoparticles incorporating LA (WPU@LA) using primary raw materials, including N-methyldiethanolamine, 2,2'-(piperazine-1,4-diyl) diethanol, isophorone diisocyanate, castor oil, and acetic acid. These nanoparticles, identified as WPU0.89@LA, WPU0.99@LA, WPU1.09@LA, and WPU1.19@LA, underwent assessment for their pH-responsive release property and biocompatibility. Among these, WPU0.99@LA displayed superior pH-responsive release properties and biocompatibility towards Caco-2 and IPEC-J2 cells. In a mouse model, a dosage of 10 mg/kg/day WPU0.99@LA effectively reduced the conjugation of IncX4 plasmids carrying the mobile colistin resistance gene (mcr-1) by more than 45.1-fold. In vivo toxicity assessment demonstrated that 10 mg/kg/day WPU0.99@LA maintains desirable biosafety and effectively preserves gut microbiota homeostasis. In conclusion, our study provides crucial proof-of-concept support, demonstrating that WPU0.99@LA holds significant potential in controlling the spread of antibiotic resistance within the mammalian intestine.
Collapse
Affiliation(s)
- Gong Li
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Lu Han
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Li-Juan Xia
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Ang Gao
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Zhi-Peng Li
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Shi-Ying Zhou
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Lei Wan
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Yao Deng
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Tian-Hong Zhou
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Xin-Yi Lu
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Yang Luo
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Dun-Sheng Liang
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Gui-Ting Wu
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Sheng-Qiu Tang
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, PR China
| | - Xin-Lei Lian
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Hao Ren
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Xiao-Ping Liao
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| | - Liang Chen
- Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, 14214, USA
| | - Jian Sun
- Lingnan Guangdong Laboratory of Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, PR China
| |
Collapse
|
17
|
Cocker D, Birgand G, Zhu N, Rodriguez-Manzano J, Ahmad R, Jambo K, Levin AS, Holmes A. Healthcare as a driver, reservoir and amplifier of antimicrobial resistance: opportunities for interventions. Nat Rev Microbiol 2024; 22:636-649. [PMID: 39048837 DOI: 10.1038/s41579-024-01076-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
Antimicrobial resistance (AMR) is a global health challenge that threatens humans, animals and the environment. Evidence is emerging for a role of healthcare infrastructure, environments and patient pathways in promoting and maintaining AMR via direct and indirect mechanisms. Advances in vaccination and monoclonal antibody therapies together with integrated surveillance, rapid diagnostics, targeted antimicrobial therapy and infection control measures offer opportunities to address healthcare-associated AMR risks more effectively. Additionally, innovations in artificial intelligence, data linkage and intelligent systems can be used to better predict and reduce AMR and improve healthcare resilience. In this Review, we examine the mechanisms by which healthcare functions as a driver, reservoir and amplifier of AMR, contextualized within a One Health framework. We also explore the opportunities and innovative solutions that can be used to combat AMR throughout the patient journey. We provide a perspective on the current evidence for the effectiveness of interventions designed to mitigate healthcare-associated AMR and promote healthcare resilience within high-income and resource-limited settings, as well as the challenges associated with their implementation.
Collapse
Affiliation(s)
- Derek Cocker
- David Price Evans Infectious Diseases & Global Health Group, University of Liverpool, Liverpool, UK
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Gabriel Birgand
- Centre d'appui pour la Prévention des Infections Associées aux Soins, Nantes, France
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Cibles et medicaments des infections et de l'immunitée, IICiMed, Nantes Universite, Nantes, France
| | - Nina Zhu
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jesus Rodriguez-Manzano
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Raheelah Ahmad
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Department of Health Services Research & Management, City University of London, London, UK
- Dow University of Health Sciences, Karachi, Pakistan
| | - Kondwani Jambo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Anna S Levin
- Department of Infectious Disease, School of Medicine & Institute of Tropical Medicine, University of São Paulo, São Paulo, Brazil
| | - Alison Holmes
- David Price Evans Infectious Diseases & Global Health Group, University of Liverpool, Liverpool, UK.
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK.
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
18
|
Ma HR, Xu HZ, Kim K, Anderson DJ, You L. Private benefit of β-lactamase dictates selection dynamics of combination antibiotic treatment. Nat Commun 2024; 15:8337. [PMID: 39333122 PMCID: PMC11436977 DOI: 10.1038/s41467-024-52711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
β-lactam antibiotics have been prescribed for most bacterial infections since their discovery. However, resistance to β-lactams, mediated by β-lactamase (Bla) enzymes such as extended spectrum β-lactamases (ESBLs), has become widespread. Bla inhibitors can restore the efficacy of β-lactams against resistant bacteria, an approach which preserves existing antibiotics despite declining industry investment. However, the effects of combination treatment on selection for β-lactam resistance are not well understood. Bla production confers both private benefits for resistant cells and public benefits which faster-growing sensitive cells can also exploit. These benefits may be differentially impacted by Bla inhibitors, leading to non-intuitive selection dynamics. In this study, we demonstrate strain-to-strain variation in effective combination doses, with complex growth dynamics in mixed populations. Using modeling, we derive a criterion for the selection outcome of combination treatment, dependent on the burden and effective private benefit of Bla production. We then use engineered strains and natural isolates to show that strong private benefits of Bla are associated with increased selection for resistance. Finally, we demonstrate that this parameter can be coarsely estimated using high-throughput phenotyping of clonal populations. Our analysis shows that quantifying the phenotypic responses of bacteria to combination treatment can facilitate resistance-minimizing optimization of treatment.
Collapse
Affiliation(s)
- Helena R Ma
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Quantitative Biodesign, Duke University, Durham, NC, USA
| | - Helen Z Xu
- Department of Biology, Duke University, Durham, NC, USA
- Department of Computer Science, Duke University, Durham, NC, USA
| | - Kyeri Kim
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Quantitative Biodesign, Duke University, Durham, NC, USA
| | - Deverick J Anderson
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Center for Antimicrobial Stewardship and Infection Prevention, Duke University School of Medicine, Durham, NC, USA
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Center for Quantitative Biodesign, Duke University, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
19
|
Shariati A, Kashi M, Chegini Z, Hosseini SM. Antibiotics-free compounds for managing carbapenem-resistant bacteria; a narrative review. Front Pharmacol 2024; 15:1467086. [PMID: 39355778 PMCID: PMC11442292 DOI: 10.3389/fphar.2024.1467086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 10/03/2024] Open
Abstract
Carbapenem-resistant (CR) Gram-negative bacteria have become a significant public health problem in the last decade. In recent years, the prevalence of CR bacteria has increased. The resistance to carbapenems could result from different mechanisms such as loss of porin, penicillin-binding protein alteration, carbapenemase, efflux pump, and biofilm community. Additionally, genetic variations like insertion, deletion, mutation, and post-transcriptional modification of corresponding coding genes could decrease the susceptibility of bacteria to carbapenems. In this regard, scientists are looking for new approaches to inhibit CR bacteria. Using bacteriophages, natural products, nanoparticles, disulfiram, N-acetylcysteine, and antimicrobial peptides showed promising inhibitory effects against CR bacteria. Additionally, the mentioned compounds could destroy the biofilm community of CR bacteria. Using them in combination with conventional antibiotics increases the efficacy of antibiotics, decreases their dosage and toxicity, and resensitizes CR bacteria to antibiotics. Therefore, in the present review article, we have discussed different aspects of non-antibiotic approaches for managing and inhibiting the CR bacteria and various methods and procedures used as an alternative for carbapenems against these bacteria.
Collapse
Affiliation(s)
- Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran
| | - Milad Kashi
- Student research committee, Arak University of Medical Sciences, Arak, Iran
| | - Zahra Chegini
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Mostafa Hosseini
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
20
|
Mathuria A, Vora C, Ali N, Mani I. Advances in CRISPR-Cas systems for human bacterial disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 208:19-41. [PMID: 39266183 DOI: 10.1016/bs.pmbts.2024.07.013] [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: 09/14/2024]
Abstract
Prokaryotic adaptive immune systems called CRISPR-Cas systems have transformed genome editing by allowing for precise genetic alterations through targeted DNA cleavage. This system comprises CRISPR-associated genes and repeat-spacer arrays, which generate RNA molecules that guide the cleavage of invading genetic material. CRISPR-Cas is classified into Class 1 (multi-subunit effectors) and Class 2 (single multi-domain effectors). Its applications span combating antimicrobial resistance (AMR), targeting antibiotic resistance genes (ARGs), resensitizing bacteria to antibiotics, and preventing horizontal gene transfer (HGT). CRISPR-Cas3, for example, effectively degrades plasmids carrying resistance genes, providing a precise method to disarm bacteria. In the context of ESKAPE pathogens, CRISPR technology can resensitize bacteria to antibiotics by targeting specific resistance genes. Furthermore, in tuberculosis (TB) research, CRISPR-based tools enhance diagnostic accuracy and facilitate precise genetic modifications for studying Mycobacterium tuberculosis. CRISPR-based diagnostics, leveraging Cas endonucleases' collateral cleavage activity, offer highly sensitive pathogen detection. These advancements underscore CRISPR's transformative potential in addressing AMR and enhancing infectious disease management.
Collapse
Affiliation(s)
- Anshu Mathuria
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Chaitali Vora
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, India
| | - Namra Ali
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
21
|
Wang S, Ma S, Sun S, Wang Q, Ding Q, Jin L, Chen F, Yin G, Wu X, Wang R, Wang H. Global evolutionary dynamics of virulence genes in ST11-KL47 carbapenem-resistant Klebsiella pneumoniae. Int J Antimicrob Agents 2024; 64:107245. [PMID: 38906484 DOI: 10.1016/j.ijantimicag.2024.107245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
ST11-KL47 is a hypervirulent carbapenem-resistant Klebsiella pneumoniae (CRKP) that is highly prevalent in China and poses a major public health risk. To investigate the evolutionary dynamics of virulence genes in this subclone, we analysed 78 sequenced isolates obtained from a long-term study across 29 centres from 17 cities in China. Virulence genes were located in large hybrid pNDM-Mar-like plasmids (length: ∼266 kilobases) rather than in classical pK2044-like plasmids. These hybrid plasmids, derived from the fusion of pK2044 and pNDM-Mar plasmids mediated by insertion sequence (IS) elements (such as ISKpn28 and IS26), integrated virulence gene fragments into the chromosome. Analysis of 217 sequences containing the special IncFIB (pNDM-Mar) replicon using public databases indicated that these plasmids typically contained T4SS-related and multiple antimicrobial resistance genes, were present in 24 countries, and were found in humans, animals, and the environment. Notably, the chromosomal integration of virulence genes was observed in strains across five countries across two continents. In vivo and in vitro models showed that the large hybrid plasmid increased the host fitness cost while increasing virulence. Conversely, virulence genes transferred to chromosomes resulted in increased fitness and lower virulence. In conclusion, virulence genes in the plasmids of ST11-KL47 CRKP are evolving, driven by adaptive negative selection, to enable vertical chromosomal inheritance along with conferring a survival advantage and low pathogenicity.
Collapse
Affiliation(s)
- Shuyi Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Shuai Ma
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Shijun Sun
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Qi Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Qi Ding
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Longyang Jin
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Fengning Chen
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Guankun Yin
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xingyu Wu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ruobing Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
22
|
Alav I, Pordelkhaki P, Rodriguez-Navarro J, Neo O, Kessler C, Awodipe RJ, Cliffe P, Pulavan N, Marton HL, Gibbons S, Buckner MMC. Natural products from food sources can alter the spread of antimicrobial resistance plasmids in Enterobacterales. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001496. [PMID: 39190025 PMCID: PMC11541548 DOI: 10.1099/mic.0.001496] [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: 03/06/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024]
Abstract
Antimicrobial resistance (AMR) poses a significant threat to global public health. Notably, resistance to carbapenem and extended-spectrum β-lactam antibiotics in Gram-negative bacteria is a major impediment to treating infections. Genes responsible for antibiotic resistance are frequently carried on plasmids, which can transfer between bacteria. Therefore, exploring strategies to prevent this transfer and the prevalence of AMR plasmids is timely and pertinent. Here, we show that certain natural product extracts and associated pure compounds can reduce the conjugation of AMR plasmids into new bacterial hosts. Using our established high-throughput fluorescence-based flow cytometry assay, we found that the natural products were more active in reducing transmission of the IncK extended-spectrum β-lactamase-encoding plasmid pCT in Escherichia coli EC958c, compared to Klebsiella pneumoniae Ecl8 carrying the IncFII carbapenemase-encoding plasmid pKpQIL. The exception was the natural product rottlerin, also active in K. pneumoniae. In classical conjugation assays, rottlerin also reduced the conjugation frequency of the IncFII bla NDM-1 carrying plasmid pCPE16_3 from a clinical K. pneumoniae isolate. Our data indicate that the natural products tested here, in their current molecular structure, reduced conjugation by a small amount, which is unlikely to achieve a large-scale reduction in AMR in bacterial populations. However, certain natural products like rottlerin could provide a foundation for further research into compounds with effective anti-plasmid activity.
Collapse
Affiliation(s)
- Ilyas Alav
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Parisa Pordelkhaki
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Judith Rodriguez-Navarro
- Department of Microbiology, Hospital de la Santa Creu i Sant Pau, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), Sant Quintıí 89, E-08041 Barcelona, Spain
| | - Onalenna Neo
- School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Celia Kessler
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Poppy Cliffe
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Nivethanaa Pulavan
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Huba L. Marton
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Simon Gibbons
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz, Nizwa 616, Sultanate of Oman
| | - Michelle M. C. Buckner
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
23
|
Hameed S, Sharif S, Ovais M, Xiong H. Emerging trends and future challenges of advanced 2D nanomaterials for combating bacterial resistance. Bioact Mater 2024; 38:225-257. [PMID: 38745587 PMCID: PMC11090881 DOI: 10.1016/j.bioactmat.2024.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
The number of multi-drug-resistant bacteria has increased over the last few decades, which has caused a detrimental impact on public health worldwide. In resolving antibiotic resistance development among different bacterial communities, new antimicrobial agents and nanoparticle-based strategies need to be designed foreseeing the slow discovery of new functioning antibiotics. Advanced research studies have revealed the significant disinfection potential of two-dimensional nanomaterials (2D NMs) to be severed as effective antibacterial agents due to their unique physicochemical properties. This review covers the current research progress of 2D NMs-based antibacterial strategies based on an inclusive explanation of 2D NMs' impact as antibacterial agents, including a detailed introduction to each possible well-known antibacterial mechanism. The impact of the physicochemical properties of 2D NMs on their antibacterial activities has been deliberated while explaining the toxic effects of 2D NMs and discussing their biomedical significance, dysbiosis, and cellular nanotoxicity. Adding to the challenges, we also discussed the major issues regarding the current quality and availability of nanotoxicity data. However, smart advancements are required to fabricate biocompatible 2D antibacterial NMs and exploit their potential to combat bacterial resistance clinically.
Collapse
Affiliation(s)
- Saima Hameed
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, PR China
- School of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, PR China
| | - Sumaira Sharif
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Muhammad Ovais
- BGI Genomics, BGI Shenzhen, Shenzhen, 518083, Guangdong, PR China
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, PR China
| |
Collapse
|
24
|
Prieto A, Miró L, Margolles Y, Bernabeu M, Salguero D, Merino S, Tomas J, Corbera JA, Perez-Bosque A, Huttener M, Fernández LÁ, Juarez A. Targeting plasmid-encoded proteins that contain immunoglobulin-like domains to combat antimicrobial resistance. eLife 2024; 13:RP95328. [PMID: 39046772 PMCID: PMC11268884 DOI: 10.7554/elife.95328] [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] [Indexed: 07/25/2024] Open
Abstract
Antimicrobial resistance (AMR) poses a significant threat to human health. Although vaccines have been developed to combat AMR, it has proven challenging to associate specific vaccine antigens with AMR. Bacterial plasmids play a crucial role in the transmission of AMR. Our recent research has identified a group of bacterial plasmids (specifically, IncHI plasmids) that encode large molecular mass proteins containing bacterial immunoglobulin-like domains. These proteins are found on the external surface of the bacterial cells, such as in the flagella or conjugative pili. In this study, we show that these proteins are antigenic and can protect mice from infection caused by an AMR Salmonella strain harboring one of these plasmids. Furthermore, we successfully generated nanobodies targeting these proteins, that were shown to interfere with the conjugative transfer of IncHI plasmids. Considering that these proteins are also encoded in other groups of plasmids, such as IncA/C and IncP2, targeting them could be a valuable strategy in combating AMR infections caused by bacteria harboring different groups of AMR plasmids. Since the selected antigens are directly linked to AMR itself, the protective effect extends beyond specific microorganisms to include all those carrying the corresponding resistance plasmids.
Collapse
Affiliation(s)
- Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Luïsa Miró
- Department of Biochemistry and Physiology, Universitat de BarcelonaBarcelonaSpain
- Institut de Nutrició i Seguretat Alimentària, Universitat de BarcelonaBarcelonaSpain
| | - Yago Margolles
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC)MadridSpain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - David Salguero
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Susana Merino
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Joan Tomas
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Juan Alberto Corbera
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Facultad de Veterinaria, Universidad de Las Palmas de Gran Canaria (ULPGC), Campus Universitario de ArucasLas PalmasSpain
| | - Anna Perez-Bosque
- Department of Biochemistry and Physiology, Universitat de BarcelonaBarcelonaSpain
- Institut de Nutrició i Seguretat Alimentària, Universitat de BarcelonaBarcelonaSpain
| | - Mario Huttener
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC)MadridSpain
| | - Antonio Juarez
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
- Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and TechnologyBarcelonaSpain
| |
Collapse
|
25
|
Gruzdev N, Katz C, Yadid I. Curing of a field strain of Salmonella enterica serovar Infantis isolated from poultry from its highly stable pESI like plasmid. J Microbiol Methods 2024; 222:106959. [PMID: 38782300 DOI: 10.1016/j.mimet.2024.106959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/05/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Salmonella enterica serovar Infantis (S. infantis) is an important emerging pathogen, associated with poultry and poultry products and related to an increasing number of human infections in many countries. A concerning trend among S. infantis isolates is the presence of plasmid-mediated multidrug resistance. In many instances, the genes responsible for this resistance are carried on a megaplasmid known as the plasmid of emerging S. infantis (pESI) or pESI like plasmids. Plasmids can be remarkably stable due to the presence of multiple replicons and post-segregational killing systems (PSKs), which contribute to their maintenance within bacterial populations. To enhance our understanding of S. infantis and its multidrug resistance determinants toward the development of new vaccination strategies, we have devised a new method for targeted plasmid curing. This approach effectively overcomes plasmid addiction by leveraging the temporal overproduction of specific antitoxins coupled with the deletion of the partition region. By employing this strategy, we successfully generated a plasmid-free strain from a field isolate derived from S. infantis 119,944. This method provides valuable tools for studying S. infantis and its plasmid-borne multidrug resistance mechanisms and can be easily adopted for plasmid curing from other related bacteria.
Collapse
Affiliation(s)
- Nadya Gruzdev
- Migal-Galilee Research Institute, Kiryat-Shmona 1101602, Israel
| | - Chen Katz
- Migal-Galilee Research Institute, Kiryat-Shmona 1101602, Israel
| | - Itamar Yadid
- Migal-Galilee Research Institute, Kiryat-Shmona 1101602, Israel; Tel-Hai College, Upper Galilee 1220800, Israel.
| |
Collapse
|
26
|
Al-Fadhli AH, Jamal WY. Recent advances in gene-editing approaches for tackling antibiotic resistance threats: a review. Front Cell Infect Microbiol 2024; 14:1410115. [PMID: 38994001 PMCID: PMC11238145 DOI: 10.3389/fcimb.2024.1410115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Antibiotic resistance, a known global health challenge, involves the flow of bacteria and their genes among animals, humans, and their surrounding environment. It occurs when bacteria evolve and become less responsive to the drugs designated to kill them, making infections harder to treat. Despite several obstacles preventing the spread of genes and bacteria, pathogens regularly acquire novel resistance factors from other species, which reduces their ability to prevent and treat such bacterial infections. This issue requires coordinated efforts in healthcare, research, and public awareness to address its impact on human health worldwide. This review outlines how recent advances in gene editing technology, especially CRISPR/Cas9, unveil a breakthrough in combating antibiotic resistance. Our focus will remain on the relationship between CRISPR/cas9 and its impact on antibiotic resistance and its related infections. Moreover, the prospects of this new advanced research and the challenges of adopting these technologies against infections will be outlined by exploring its different derivatives and discussing their advantages and limitations over others, thereby providing a corresponding reference for the control and prevention of the spread of antibiotic resistance.
Collapse
Affiliation(s)
- Amani H Al-Fadhli
- Laboratory Sciences, Department of Medical, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
| | - Wafaa Yousef Jamal
- Department of Microbiology, College of Medicine, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
27
|
Wang P, Du X, Zhao Y, Wang W, Cai T, Tang K, Wang X. Combining CRISPR/Cas9 and natural excision for the precise and complete removal of mobile genetic elements in bacteria. Appl Environ Microbiol 2024; 90:e0009524. [PMID: 38497640 PMCID: PMC11022536 DOI: 10.1128/aem.00095-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
Horizontal gene transfer, facilitated by mobile genetic elements (MGEs), is an adaptive evolutionary process that contributes to the evolution of bacterial populations and infectious diseases. A variety of MGEs not only can integrate into the bacterial genome but also can survive or even replicate like plasmids in the cytoplasm, thus requiring precise and complete removal for studying their strategies in benefiting host cells. Existing methods for MGE removal, such as homologous recombination-based deletion and excisionase-based methods, have limitations in effectively eliminating certain MGEs. To overcome these limitations, we developed the Cas9-NE method, which combines the CRISPR/Cas9 system with the natural excision of MGEs. In this approach, a specialized single guide RNA (sgRNA) element is designed with a 20-nucleotide region that pairs with the MGE sequence. This sgRNA is expressed from a plasmid that also carries the Cas9 gene. By utilizing the Cas9-NE method, both the integrative and circular forms of MGEs can be precisely and completely eliminated through Cas9 cleavage, generating MGE-removed cells. We have successfully applied the Cas9-NE method to remove four representative MGEs, including plasmids, prophages, and genomic islands, from Vibrio strains. This new approach not only enables various investigations on MGEs but also has significant implications for the rapid generation of strains for commercial purposes.IMPORTANCEMobile genetic elements (MGEs) are of utmost importance for bacterial adaptation and pathogenicity, existing in various forms and multiple copies within bacterial cells. Integrated MGEs play dual roles in bacterial hosts, enhancing the fitness of the host by delivering cargo genes and potentially modifying the bacterial genome through the integration/excision process. This process can lead to alterations in promoters or coding sequences or even gene disruptions at integration sites, influencing the physiological functions of host bacteria. Here, we developed a new approach called Cas9-NE, allowing them to maintain the natural sequence changes associated with MGE excision. Cas9-NE allows the one-step removal of integrated and circular MGEs, addressing the challenge of eliminating various MGE forms efficiently. This approach simplifies MGE elimination in bacteria, expediting research on MGEs.
Collapse
Affiliation(s)
- Pengxia Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, China, Guangzhou
- Guangdong Provincial Observation and Research Station for Coastal Upwelling Ecosystem, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Shantou, China
- China, Southern Marine Science and Engineering Guangdong LaboratoryGuangzhou
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaofei Du
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, China, Guangzhou
- China, Southern Marine Science and Engineering Guangdong LaboratoryGuangzhou
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Zhao
- University of Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Weiquan Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, China, Guangzhou
- China, Southern Marine Science and Engineering Guangdong LaboratoryGuangzhou
- University of Chinese Academy of Sciences, Beijing, China
| | - Tongxuan Cai
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, China, Guangzhou
- College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Kaihao Tang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, China, Guangzhou
- China, Southern Marine Science and Engineering Guangdong LaboratoryGuangzhou
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoxue Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, China, Guangzhou
- China, Southern Marine Science and Engineering Guangdong LaboratoryGuangzhou
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
28
|
Alav I, Pordelkhaki P, de Resende PE, Partington H, Gibbons S, Lord RM, Buckner MMC. Cobalt complexes modulate plasmid conjugation in Escherichia coli and Klebsiella pneumoniae. Sci Rep 2024; 14:8103. [PMID: 38582880 PMCID: PMC10998897 DOI: 10.1038/s41598-024-58895-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/04/2024] [Indexed: 04/08/2024] Open
Abstract
Antimicrobial resistance genes (ARG), such as extended-spectrum β-lactamase (ESBL) and carbapenemase genes, are commonly carried on plasmids. Plasmids can transmit between bacteria, disseminate globally, and cause clinically important resistance. Therefore, targeting plasmids could reduce ARG prevalence, and restore the efficacy of existing antibiotics. Cobalt complexes possess diverse biological activities, including antimicrobial and anticancer properties. However, their effect on plasmid conjugation has not been explored yet. Here, we assessed the effect of four previously characterised bis(N-picolinamido)cobalt(II) complexes lacking antibacterial activity on plasmid conjugation in Escherichia coli and Klebsiella pneumoniae. Antimicrobial susceptibility testing of these cobalt complexes confirmed the lack of antibacterial activity in E. coli and K. pneumoniae. Liquid broth and solid agar conjugation assays were used to screen the activity of the complexes on four archetypical plasmids in E. coli J53. The cobalt complexes significantly reduced the conjugation of RP4, R6K, and R388 plasmids, but not pKM101, on solid agar in E. coli J53. Owing to their promising activity, the impact of cobalt complexes was tested on the conjugation of fluorescently tagged extended-spectrum β-lactamase encoding pCTgfp plasmid in E. coli and carbapenemase encoding pKpQILgfp plasmid in K. pneumoniae, using flow cytometry. The complexes significantly reduced the conjugation of pKpQILgfp in K. pneumoniae but had no impact on pCTgfp conjugation in E. coli. The cobalt complexes did not have plasmid-curing activity, suggesting that they target conjugation rather than plasmid stability. To our knowledge, this is the first study to report reduced conjugation of clinically relevant plasmids with cobalt complexes. These cobalt complexes are not cytotoxic towards mammalian cells and are not antibacterial, therefore they could be optimised and employed as inhibitors of plasmid conjugation.
Collapse
Affiliation(s)
- Ilyas Alav
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Parisa Pordelkhaki
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Pedro Ernesto de Resende
- School of Pharmacy, Faculty of Science, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Hannah Partington
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Simon Gibbons
- Natural & Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, P.O. Box 33, Nizwa, 616, Oman
| | - Rianne M Lord
- School of Chemistry, Faculty of Science, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Michelle M C Buckner
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
29
|
Liu X, Zhao H, Wong A. Accounting for the health risk of probiotics. Heliyon 2024; 10:e27908. [PMID: 38510031 PMCID: PMC10950733 DOI: 10.1016/j.heliyon.2024.e27908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Probiotics have long been associated with a myriad of health benefits, so much so that their adverse effects whether mild or severe, are often neglected or overshadowed by the enormous volume of articles describing their beneficial effects in the current literature. Recent evidence has demonstrated several health risks of probiotics that warrant serious reconsideration of their applications and further investigations. This review aims to highlight studies that report on how probiotics might cause opportunistic systemic and local infections, detrimental immunological effects, metabolic disturbance, allergic reactions, and facilitating the spread of antimicrobial resistance. To offer a recent account of the literature, articles within the last five years were prioritized. The narration of these evidence was based on the nature of the studies in the following order of preference: clinical studies or human samples, in vivo or animal models, in situ, in vitro and/or in silico. We hope that this review will inform consumers, food scientists, and medical practitioners, on the health risks, while also encouraging research that will focus on and clarify the adverse effects of probiotics.
Collapse
Affiliation(s)
- Xiangyi Liu
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang Province, 325060, China
- Department of Biology, Dorothy and George Hennings College of Science, Mathematics and Technology, Kean, University, 1000 Morris Ave, Union, NJ, 07083, USA
| | - Haiyi Zhao
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang Province, 325060, China
- Department of Biology, Dorothy and George Hennings College of Science, Mathematics and Technology, Kean, University, 1000 Morris Ave, Union, NJ, 07083, USA
| | - Aloysius Wong
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang Province, 325060, China
- Department of Biology, Dorothy and George Hennings College of Science, Mathematics and Technology, Kean, University, 1000 Morris Ave, Union, NJ, 07083, USA
- Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Ouhai, Wenzhou, Zhejiang Province, 325060, China
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Ouhai, Wenzhou, Zhejiang Province, 325060, China
| |
Collapse
|
30
|
Mei L, Song Y, Liu X, Li K, Guo X, Liu L, Liu Y, Kozlakidis Z, Cheong IH, Wang D, Wei Q. Characterization and Implications of IncP-2A Plasmid pMAS152 Harboring Multidrug Resistance Genes in Extensively Drug-Resistant Pseudomonas aeruginosa. Microorganisms 2024; 12:562. [PMID: 38543613 PMCID: PMC10973999 DOI: 10.3390/microorganisms12030562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 04/21/2024] Open
Abstract
Bacterial antimicrobial resistance (AMR) poses a significant global public health challenge. The escalation of AMR is primarily attributed to the horizontal gene transfer (HGT) of antibiotic resistance genes (ARGs), often facilitated by plasmids. This underscores the critical need for a comprehensive understanding of the resistance mechanisms and transmission dynamics of these plasmids. In this study, we utilized in vitro drug sensitivity testing, conjugation transfer assays, and whole-genome sequencing to investigate the resistance mechanism of an extensively drug-resistant (XDR) Pseudomonas aeruginosa clinical isolate, MAS152. We specifically focused on analyzing the drug-resistant plasmid pMAS152 it harbors and its potential for widespread dissemination. Bioinformatics analysis revealed that MAS152 carries a distinct IncpP-2A plasmid, pMAS152, characterized by a 44.8 kb multidrug resistance (MDR) region. This region houses a 16S rRNA methyltransferase (16S-RMTase) gene, rmtB, conferring high-level resistance to aminoglycoside antibiotics. Notably, this region also contains an extended-spectrum β-Lactamase (ESBL) gene, blaPER-1, and an efflux pump operon, tmexCD-oprJ, which mediate resistance to β-Lactams and quinolone antibiotics, respectively. Such a combination of ARGs, unprecedented in reported plasmids, could significantly undermine the effectiveness of first-line antibiotics in treating P. aeruginosa infections. Investigation into the genetic environment of the MDR region suggests that Tn2 and IS91 elements may be instrumental in the horizontal transfer of rmtB. Additionally, a complex Class I integron with an ISCR1 structure, along with TnAs1, seems to facilitate the horizontal transfer of blaPER-1. The conjugation transfer assay, coupled with the annotation of conjugation-related genes and phylogenetic analysis, indicates that the plasmid pMAS152 functions as a conjugative plasmid, with other genus Pseudomonas species as potential hosts. Our findings provide vital insights into the resistance mechanisms and transmission potential of the XDR P. aeruginosa isolate MAS152, underlining the urgent need for novel strategies to combat the spread of AMR. This study highlights the complex interplay of genetic elements contributing to antibiotic resistance and underscores the importance of continuous surveillance of emerging ARGs in clinical isolates.
Collapse
Affiliation(s)
- Li Mei
- National Pathogen Resource Center, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.M.); (L.L.); (Y.L.)
| | - Yang Song
- Division of Infectious Disease, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China;
| | - Xiao Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (K.L.)
| | - Kun Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (K.L.)
| | - Xu Guo
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing 100050, China;
| | - Li Liu
- National Pathogen Resource Center, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.M.); (L.L.); (Y.L.)
| | - Yang Liu
- National Pathogen Resource Center, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.M.); (L.L.); (Y.L.)
| | - Zisis Kozlakidis
- International Agency for Research on Cancer, World Health Organization, 69007 Lyon, France;
| | - Io Hong Cheong
- State Key Laboratory of Systems Medicine for Cancer, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China;
| | - Duochun Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (X.L.); (K.L.)
| | - Qiang Wei
- National Pathogen Resource Center, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.M.); (L.L.); (Y.L.)
| |
Collapse
|
31
|
Alishahi M, Aboelkheir M, Chowdhury R, Altier C, Shen H, Uyar T. Functionalization of cotton nonwoven with cyclodextrin/lawsone inclusion complex nanofibrous coating for antibacterial wound dressing. Int J Pharm 2024; 652:123815. [PMID: 38242260 DOI: 10.1016/j.ijpharm.2024.123815] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 01/21/2024]
Abstract
Functionalizing cotton to induce biological activity is a viable approach for developing wound dressing. This study explores the development of cotton-based wound dressing through coating with biologically active nanofibers. Bioactive compounds like lawsone offer dual benefits of wound healing and infection prevention, however, their limited solubility and viability hinder their applications. To address this, Hydroxypropyl-beta-cyclodextrin (HP-β-CD) and Hydroxypropyl-gamma-cyclodextrin (HP-γ-CD) were employed. Inclusion complexations of CD/lawsone were achieved at 2:1 and 4:1 M ratios, followed by the fabrication of CD/lawsone nanofibrous systems via electrospinning. Phase solubility studies indicated a twofold increase in lawsone water-solubility with HP-β-CD. Electrospinning yielded smooth and uniform nanofibers with an average diameter of ∼300-700 nm. The results showed that while specific crystalline peaks of lawsone are apparent in the samples with a 2:1 M ratio, they disappeared in 4:1, indicating complete complexation. The nanofibers exhibited ∼100 % loading efficiency of lawsone and its rapid release upon dissolution. Notably, antibacterial assays demonstrated the complete elimination of Escherichia coli and Staphylococcus aureus colonies. The CD/lawsone nanofibers also showed suitable antioxidant activity ranging from 50 % to 70 %. This integrated approach effectively enhances lawsone's solubility through CD complexation and offers promise for bilayer cotton-based wound dressings.
Collapse
Affiliation(s)
- Mohsen Alishahi
- Fiber Science Program, Department of Human Centered Design, College of Human Ecology, Cornell University, Ithaca, NY 14853, United States
| | - Mahmoud Aboelkheir
- Fiber Science Program, Department of Human Centered Design, College of Human Ecology, Cornell University, Ithaca, NY 14853, United States
| | - Rimi Chowdhury
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States
| | - Craig Altier
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States
| | | | - Tamer Uyar
- Fiber Science Program, Department of Human Centered Design, College of Human Ecology, Cornell University, Ithaca, NY 14853, United States.
| |
Collapse
|
32
|
Kozaeva E, Nielsen ZS, Nieto-Domínguez M, Nikel P. The pAblo·pCasso self-curing vector toolset for unconstrained cytidine and adenine base-editing in Gram-negative bacteria. Nucleic Acids Res 2024; 52:e19. [PMID: 38180826 PMCID: PMC10899774 DOI: 10.1093/nar/gkad1236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
A synthetic biology toolkit, exploiting clustered regularly interspaced short palindromic repeats (CRISPR) and modified CRISPR-associated protein (Cas) base-editors, was developed for genome engineering in Gram-negative bacteria. Both a cytidine base-editor (CBE) and an adenine base-editor (ABE) have been optimized for precise single-nucleotide modification of plasmid and genome targets. CBE comprises a cytidine deaminase conjugated to a Cas9 nickase from Streptococcus pyogenes (SpnCas9), resulting in C→T (or G→A) substitutions. Conversely, ABE consists of an adenine deaminase fused to SpnCas9 for A→G (or T→C) editing. Several nucleotide substitutions were achieved using these plasmid-borne base-editing systems and a novel protospacer adjacent motif (PAM)-relaxed SpnCas9 (SpRY) variant. Base-editing was validated in Pseudomonas putida and other Gram-negative bacteria by inserting premature STOP codons into target genes, thereby inactivating both fluorescent proteins and metabolic (antibiotic-resistance) functions. The functional knockouts obtained by engineering STOP codons via CBE were reverted to the wild-type genotype using ABE. Additionally, a series of induction-responsive vectors was developed to facilitate the curing of the base-editing platform in a single cultivation step, simplifying complex strain engineering programs without relying on homologous recombination and yielding plasmid-free, modified bacterial cells.
Collapse
Affiliation(s)
- Ekaterina Kozaeva
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Zacharias S Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Manuel Nieto-Domínguez
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Pablo I Nikel
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
33
|
Yan Y, Li X, Yu K, Wu Z, Sun Y, Cheng Z, Zhao B, Nie C, Xia Y. Systematic evaluation of the impact of standard storage conditions on plasmid conjugation behavior in wastewater samples. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 343:123283. [PMID: 38176637 DOI: 10.1016/j.envpol.2023.123283] [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: 07/28/2023] [Revised: 11/29/2023] [Accepted: 12/30/2023] [Indexed: 01/06/2024]
Abstract
Filter mating experiment is widely used to study the conjugation behavior of plasmids and associated antibiotic resistance in environmental settings, however, the influence and biases brought by sample storage conditions (temperature and duration) were not yet systematically elaborated. This study systematically investigated the influence of standard storage conditions (4 °C, -20 °C, -80 °C) on plasmid conjugation behavior in influent (Inf) and activated sludge (AS) samples from sewage treatment plants (STP). The findings revealed a significant reduction in conjugation efficiency under all the tested storage conditions except for 1-week storage at 4 °C. Notably, storing at -80 °C maintained conjugation activities in activated sludge more effectively compared to -20 °C. However, the preservation performance was less effective for influent samples, which consist mainly of anaerobe-dominant communities. Systematic loss of IncH-type plasmids was observed in influent samples stored at 4 °C and -20 °C. Correspondingly, the plasmid-carrying resistome genotypes detected in the influent samples showed a clear downward trend with the increase in storage duration when stored at 4 °C and -20 °C. A relatively uniform composition in terms of incompatibility type and resistome profile was observed across activated sludge samples, regardless of the varied storage conditions. This study highlights the critical impact of storage conditions on plasmid conjugation behavior and resistome composition, offering valuable insights for optimal sample handling in resistome research.
Collapse
Affiliation(s)
- Yuxi Yan
- School of Environment, Harbin Institute of Technology, Harbin 150001, China; School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiang Li
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kaiqiang Yu
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ziqi Wu
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuhong Sun
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhanwen Cheng
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Bixi Zhao
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Cailong Nie
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yu Xia
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
34
|
Parra B, Cockx B, Lutz VT, Brøndsted L, Smets BF, Dechesne A. Isolation and characterization of novel plasmid-dependent phages infecting bacteria carrying diverse conjugative plasmids. Microbiol Spectr 2024; 12:e0253723. [PMID: 38063386 PMCID: PMC10782986 DOI: 10.1128/spectrum.02537-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/12/2023] [Indexed: 12/23/2023] Open
Abstract
IMPORTANCE This work was undertaken because plasmid-dependent phages can reduce the prevalence of conjugative plasmids and can be leveraged to prevent the acquisition and dissemination of ARGs by bacteria. The two novel phages described in this study, Lu221 and Hi226, can infect Escherichia coli, Salmonella enterica, Kluyvera sp. and Enterobacter sp. carrying conjugative plasmids. This was verified with plasmids carrying resistance determinants and belonging to the most common plasmid families among Gram-negative pathogens. Therefore, the newly isolated phages could have the potential to help control the spread of ARGs and thus help combat the antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Boris Parra
- Department of Environmental Engineering and Resource Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
- Laboratorio de Investigación de Agentes Antibacterianos, Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Instituto de Ciencias Naturales, Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Concepción, Chile
| | - Bastiaan Cockx
- Department of Environmental Engineering and Resource Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Veronika T. Lutz
- Department of Veterinary and Animal Sciences, University of Copenhagen, København, Denmark
| | - Lone Brøndsted
- Department of Veterinary and Animal Sciences, University of Copenhagen, København, Denmark
| | - Barth F. Smets
- Department of Environmental Engineering and Resource Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Arnaud Dechesne
- Department of Environmental Engineering and Resource Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
35
|
Castañeda-Barba S, Top EM, Stalder T. Plasmids, a molecular cornerstone of antimicrobial resistance in the One Health era. Nat Rev Microbiol 2024; 22:18-32. [PMID: 37430173 DOI: 10.1038/s41579-023-00926-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 07/12/2023]
Abstract
Antimicrobial resistance (AMR) poses a substantial threat to human health. The widespread prevalence of AMR is, in part, due to the horizontal transfer of antibiotic resistance genes (ARGs), typically mediated by plasmids. Many of the plasmid-mediated resistance genes in pathogens originate from environmental, animal or human habitats. Despite evidence that plasmids mobilize ARGs between these habitats, we have a limited understanding of the ecological and evolutionary trajectories that facilitate the emergence of multidrug resistance (MDR) plasmids in clinical pathogens. One Health, a holistic framework, enables exploration of these knowledge gaps. In this Review, we provide an overview of how plasmids drive local and global AMR spread and link different habitats. We explore some of the emerging studies integrating an eco-evolutionary perspective, opening up a discussion about the factors that affect the ecology and evolution of plasmids in complex microbial communities. Specifically, we discuss how the emergence and persistence of MDR plasmids can be affected by varying selective conditions, spatial structure, environmental heterogeneity, temporal variation and coexistence with other members of the microbiome. These factors, along with others yet to be investigated, collectively determine the emergence and transfer of plasmid-mediated AMR within and between habitats at the local and global scale.
Collapse
Affiliation(s)
- Salvador Castañeda-Barba
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
- Bioinformatics and Computational Biology Graduate Program, University of Idaho, Moscow, ID, USA
- Institute for Interdisciplinary Data Sciences, University of Idaho, Moscow, ID, USA
| | - Eva M Top
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
- Bioinformatics and Computational Biology Graduate Program, University of Idaho, Moscow, ID, USA
- Institute for Interdisciplinary Data Sciences, University of Idaho, Moscow, ID, USA
- Institute for Modelling Collaboration and Innovation, University of Idaho, Moscow, ID, USA
| | - Thibault Stalder
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA.
- Institute for Interdisciplinary Data Sciences, University of Idaho, Moscow, ID, USA.
- Institute for Modelling Collaboration and Innovation, University of Idaho, Moscow, ID, USA.
| |
Collapse
|
36
|
Mao Y, Liu X, Zhang N, Wang Z, Han M. NCRD: A non-redundant comprehensive database for detecting antibiotic resistance genes. iScience 2023; 26:108141. [PMID: 37876810 PMCID: PMC10590964 DOI: 10.1016/j.isci.2023.108141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/13/2023] [Accepted: 10/02/2023] [Indexed: 10/26/2023] Open
Abstract
Antibiotic resistance genes (ARGs) are emerging pollutants present in various environments. Identifying ARGs has become a growing concern in recent years. Several databases, including the Antibiotic Resistance Genes Database (ARDB), Comprehensive Antibiotic Resistance Database (CARD), and Structured Antibiotic Resistance Genes (SARG), have been applied to detect ARGs. However, these databases have limitations, which hinder the comprehensive profiling of ARGs in environmental samples. To address these issues, we constructed a non-redundant antibiotic resistance genes database (NRD) by consolidating sequences from ARDB, CARD, and SARG. We identified the homologous proteins of NRD from Non-redundant Protein Database (NR) and the Protein DataBank Database (PDB) and clustered them to establish a non-redundant comprehensive antibiotic resistance genes database (NCRD) with similarities of 100% (NCRD100) and 95% (NCRD95). To demonstrate the advantages of NCRD, we compared it with other databases by using metagenome datasets. Results revealed its strong ability in detecting potential ARGs.
Collapse
Affiliation(s)
- Yujie Mao
- Key Laboratory for Environment and Disaster Monitoring and Evaluation of Hubei, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430077, China
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohui Liu
- College of Environmental Science and Engineering, Ocean University of China, Qingdao 266003, China
- Key Laboratory of Marine Environmental Science and Ecology, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Na Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Zhi Wang
- Key Laboratory for Environment and Disaster Monitoring and Evaluation of Hubei, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430077, China
| | - Maozhen Han
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
37
|
Hill B, Schafer B, Vargas N, Zamora D, Shrotri R, Perez S, Farmer G, Avon A, Pai A, Mori H, Zhong J. Functional analysis of Rickettsia monacensis strain humboldt folA dihydrofolate reductase gene via complementation assay. Ticks Tick Borne Dis 2023; 14:102217. [PMID: 37379700 DOI: 10.1016/j.ttbdis.2023.102217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023]
Abstract
Nutritive symbiosis between bacteria and ticks is observed across a range of ecological contexts; however, little characterization on the molecular components responsible for this symbiosis has been done. Previous studies in our lab demonstrated that Rickettsia monacensis str. Humboldt (strain Humboldt) can synthesize folate de novo via the folate biosynthesis pathway involving folA, folC, folE, folKP, and ptpS genes. In this study, expression of the strain Humboldt folA gene within a folA mutant Escherichia coli construct was used to functionally characterize the strain Humboldt folA folate gene in vivo. The strain Humboldt folA folate gene was subcloned into a TransBac vector and transformed into a folA mutant E. coli construct. The mutant containing strain Humboldt folA subclone and a pFE604 clone of the knocked-out folA gene was cured of pFE604. Curing of the folA mutant E. coli construct was successful using acridine orange and 43.5 °C incubation temperature. The plasmid curing assay showed curing efficiency of the folA mutant at 100%. Functional complementation was assessed by growth phenotype on minimal media with and without IPTG between strain Humboldt folA and E. coli folA. Large and homogenous wild-type colony growth was observed for both strain Humboldt and E. coli folA on minimal media with 0.1 mM IPTG, wild-type growth for strain Humboldt folA and pin-point growth for E. coli folA on 0.01 mM IPTG, and pin-point growth without IPTG for both strain Humboldt and E. coli folA. This study provides evidence substantiating the in vivo functionality of strain Humboldt folA in producing functional gene products for folate biosynthesis.
Collapse
Affiliation(s)
- Brandon Hill
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Ben Schafer
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Nolan Vargas
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Danny Zamora
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Rohan Shrotri
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Sarahi Perez
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Geoffrey Farmer
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Aren Avon
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Anirudh Pai
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America
| | - Hirotada Mori
- Laboratory of Systems Microbiology, Data Science Center, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0101, Japan
| | - Jianmin Zhong
- Department of Biological Sciences, Cal Poly Humboldt, 1 Harpst Street, Arcata, CA 95521, United States of America.
| |
Collapse
|
38
|
Chen BC, Chen YZ, Lin HY. An Introduced RNA-Only Approach for Plasmid Curing via the CRISPR-Cpf1 System in Saccharomyces cerevisiae. Biomolecules 2023; 13:1561. [PMID: 37892243 PMCID: PMC10604987 DOI: 10.3390/biom13101561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The CRISPR-Cas system has been widely used for genome editing due to its convenience, simplicity and flexibility. Using a plasmid-carrying Cas protein and crRNA or sgRNA expression cassettes is an efficient strategy in the CRISPR-Cas genome editing system. However, the plasmid remains in the cells after genome editing. Development of general plasmid-curing strategies is necessary. Based on our previous CRISPR-Cpf1 genome-editing system in Saccharomyces cerevisiae, the crRNA, designed for the replication origin of the CRISPR-Cpf1 plasmid, and the ssDNA, as a template for homologous recombination, were introduced for plasmid curing. The efficiency of the plasmid curing was 96 ± 4%. In addition, we further simplified the plasmid curing system by transforming only one crRNA into S. cerevisiae, and the curing efficiency was about 70%. In summary, we have developed a CRISPR-mediated plasmid-curing system. The RNA-only plasmid curing system is fast and easy. This plasmid curing strategy can be applied in broad hosts by designing crRNA specific for the replication origin of the plasmid. The plasmid curing system via CRISPR-Cas editing technology can be applied to produce traceless products without foreign genes and to perform iterative processes in multiple rounds of genome editing.
Collapse
Affiliation(s)
- Bo-Chou Chen
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu 300, Taiwan;
| | - Yu-Zhen Chen
- Department of Food Science and Technology, Hungkuang University, Taichung 433, Taiwan;
| | - Huan-Yu Lin
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu 300, Taiwan;
| |
Collapse
|
39
|
Bhaskaran R, Ramachandra KSS, Peter R, Gopakumar ST, Gopalan MK, Mozhikulangara RR. Antimicrobial resistance and antagonistic features of bivalve-associated Vibrio parahaemolyticus from the south-west coast of India. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:107681-107692. [PMID: 37740157 DOI: 10.1007/s11356-023-29924-5] [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: 05/01/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
Vibrio parahaemolyticus, a potent human and aquatic pathogen, is usually found in estuaries and oceans. Human illness is associated with consuming uncooked/partially cooked contaminated seafood. The study on bivalve-associated V. parahaemolyticus revealed that the post-monsoon season had the highest bacterial abundance (9 ± 1.5 log cfu) compared to the monsoon season (8.03 ± 0.56 log cfu). Antimicrobial resistance (AMR) profiling was performed on 114 V. parahaemolyticus isolates obtained from bivalves. The highest AMR was observed against ampicillin (78%). Chloramphenicol was found to be effective against all the isolates. Multiple antibiotic resistance index values of 0.2 or higher were detected in 18% of the isolates. Molecular analysis of antimicrobial resistant genes (ARGs) revealed the high prevalence (100%) of the TEM-1 gene in the aquatic environment. After plasmid profiling and curing, 41.6% and 100% of the resistant isolates were found to be sensitive to ampicillin and cephalosporins, respectively, indicating the prevalence of plasmid-associated ARGs in the aquatic environment. A study to evaluate the antagonistic properties of Bacillus subtilis, Pseudomonas aeruginosa, and Bacillus amyloliquefaciens against V. parahaemolyticus isolates identified the potential of these bacteria to resist the growth of V. parahaemolyticus.
Collapse
Affiliation(s)
- Remya Bhaskaran
- Marine Biotechnology, Fish Nutrition and Health Division (MBFNHD), ICAR-Central Marine Fisheries Research Institute, Post Box No. 1603, Kochi, Ernakulam North (P.O.), 682 018, India
- Department of Biosciences, Mangalore University, Mangalagangotri - 574 199, Karnataka State, India
| | - Krupesha Sharma Sulumane Ramachandra
- Marine Biotechnology, Fish Nutrition and Health Division (MBFNHD), ICAR-Central Marine Fisheries Research Institute, Post Box No. 1603, Kochi, Ernakulam North (P.O.), 682 018, India.
| | - Reynold Peter
- Marine Biotechnology, Fish Nutrition and Health Division (MBFNHD), ICAR-Central Marine Fisheries Research Institute, Post Box No. 1603, Kochi, Ernakulam North (P.O.), 682 018, India
| | - Sumithra Thangalazhy Gopakumar
- Marine Biotechnology, Fish Nutrition and Health Division (MBFNHD), ICAR-Central Marine Fisheries Research Institute, Post Box No. 1603, Kochi, Ernakulam North (P.O.), 682 018, India
| | - Mini Kalappurakkal Gopalan
- Fishery Resources Assessment, Economics and Extension Division (FRAEED), ICAR-Central Marine Fisheries Research Institute, Post Box No. 1603, Kochi, Ernakulam North (P.O.), 682 018, India
| | - Rithin Raj Mozhikulangara
- School of Industrial Fisheries, Cochin University of Science and Technology (CUSAT), Lakeside Campus, Kochi, 682 016, India
| |
Collapse
|
40
|
Rabaan AA, Al Fares MA, Almaghaslah M, Alpakistany T, Al Kaabi NA, Alshamrani SA, Alshehri AA, Almazni IA, Saif A, Hakami AR, Khamis F, Alfaresi M, Alsalem Z, Alsoliabi ZA, Al Amri KAS, Hassoueh AK, Mohapatra RK, Arteaga-Livias K, Alissa M. Application of CRISPR-Cas System to Mitigate Superbug Infections. Microorganisms 2023; 11:2404. [PMID: 37894063 PMCID: PMC10609045 DOI: 10.3390/microorganisms11102404] [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: 08/08/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Multidrug resistance in bacterial strains known as superbugs is estimated to cause fatal infections worldwide. Migration and urbanization have resulted in overcrowding and inadequate sanitation, contributing to a high risk of superbug infections within and between different communities. The CRISPR-Cas system, mainly type II, has been projected as a robust tool to precisely edit drug-resistant bacterial genomes to combat antibiotic-resistant bacterial strains effectively. To entirely opt for its potential, advanced development in the CRISPR-Cas system is needed to reduce toxicity and promote efficacy in gene-editing applications. This might involve base-editing techniques used to produce point mutations. These methods employ designed Cas9 variations, such as the adenine base editor (ABE) and the cytidine base editor (CBE), to directly edit single base pairs without causing DSBs. The CBE and ABE could change a target base pair into a different one (for example, G-C to A-T or C-G to A-T). In this review, we addressed the limitations of the CRISPR/Cas system and explored strategies for circumventing these limitations by applying diverse base-editing techniques. Furthermore, we also discussed recent research showcasing the ability of base editors to eliminate drug-resistant microbes.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Mona A. Al Fares
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | - Manar Almaghaslah
- Infectious Disease Division, Department of Internal Medicine, Dammam Medical Complex, Dammam 32245, Saudi Arabia
| | - Tariq Alpakistany
- Bacteriology Department, Public Health Laboratory, Taif 26521, Saudi Arabia
| | - Nawal A. Al Kaabi
- College of Medicine and Health Science, Khalifa University, Abu Dhabi 127788, United Arab Emirates
- Sheikh Khalifa Medical City, Abu Dhabi Health Services Company (SEHA), Abu Dhabi 51900, United Arab Emirates
| | - Saleh A. Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Ahmad A. Alshehri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Ibrahim Abdullah Almazni
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Ahmed Saif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62223, Saudi Arabia
| | - Abdulrahim R. Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62223, Saudi Arabia
| | - Faryal Khamis
- Infection Diseases Unit, Department of Internal Medicine, Royal Hospital, Muscat 1331, Oman
| | - Mubarak Alfaresi
- Department of Pathology and Laboratory Medicine, Zayed Military Hospital, Abu Dhabi 3740, United Arab Emirates
- Department of Pathology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Zainab Alsalem
- Department of Epidemic Diseases Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | | | | | - Amal K. Hassoueh
- Pharmacy Department, King Saud Medical City, Riyadh 7790, Saudi Arabia
| | - Ranjan K. Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar 758002, India
| | - Kovy Arteaga-Livias
- Escuela de Medicina-Filial Ica, Universidad Privada San Juan Bautista, Ica 11000, Peru
- Escuela de Medicina, Universidad Nacional Hermilio Valdizán, Huanuco 10000, Peru
| | - Mohammed Alissa
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
41
|
Wang XY, Song HW, Yi T, Shen YB, Dai CS, Sun CT, Liu DJ, Shen JZ, Wu CM, Wang Y. Dihydroartemisinin inhibits plasmid transfer in drug-resistant Escherichia coli via limiting energy supply. Zool Res 2023; 44:894-904. [PMID: 37551137 PMCID: PMC10559095 DOI: 10.24272/j.issn.2095-8137.2023.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023] Open
Abstract
Conjugative transfer of antibiotic resistance genes (ARGs) by plasmids is an important route for ARG dissemination. An increasing number of antibiotic and nonantibiotic compounds have been reported to aid the spread of ARGs, highlighting potential challenges for controlling this type of horizontal transfer. Development of conjugation inhibitors that block or delay the transfer of ARG-bearing plasmids is a promising strategy to control the propagation of antibiotic resistance. Although such inhibitors are rare, they typically exhibit relatively high toxicity and low efficacy in vivo and their mechanisms of action are inadequately understood. Here, we studied the effects of dihydroartemisinin (DHA), an artemisinin derivative used to treat malaria, on conjugation. DHA inhibited the conjugation of the IncI2 and IncX4 plasmids carrying the mobile colistin resistance gene ( mcr-1) by more than 160-fold in vitro in Escherichia coli, and more than two-fold (IncI2 plasmid) in vivo in a mouse model. It also suppressed the transfer of the IncX3 plasmid carrying the carbapenem resistance gene bla NDM-5 by more than two-fold in vitro. Detection of intracellular adenosine triphosphate (ATP) and proton motive force (PMF), in combination with transcriptomic and metabolomic analyses, revealed that DHA impaired the function of the electron transport chain (ETC) by inhibiting the tricarboxylic acid (TCA) cycle pathway, thereby disrupting PMF and limiting the availability of intracellular ATP for plasmid conjugative transfer. Furthermore, expression levels of genes related to conjugation and pilus generation were significantly down-regulated during DHA exposure, indicating that the transfer apparatus for conjugation may be inhibited. Our findings provide new insights into the control of antibiotic resistance and the potential use of DHA.
Collapse
Affiliation(s)
- Xue-Yang Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Huang-Wei Song
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Tian Yi
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Ying-Bo Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Chong-Shan Dai
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Cheng-Tao Sun
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - De-Jun Liu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Jian-Zhong Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Cong-Ming Wu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China
| | - Yang Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong 510642, China. E-mail:
| |
Collapse
|
42
|
Xiao Y, Zhang Y, Xie F, Olsen RH, Shi L, Li L. Inhibition of Plasmid Conjugation in Escherichia coli by Targeting rbsB Gene Using CRISPRi System. Int J Mol Sci 2023; 24:10585. [PMID: 37445761 DOI: 10.3390/ijms241310585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Bacterial conjugation constitutes a major horizontal gene transfer mechanism for the dissemination of antibiotic-resistant genes (ARGs) among human pathogens. The spread of ARGs can be halted or diminished by interfering with the conjugation process. In this study, we explored the possibility of using an rbsB gene as a single target to inhibit plasmid-mediated horizontal gene transfer in Escherichia coli by CRISPR interference (CRISPRi) system. Three single-guide RNAs (sgRNAs) were designed to target the rbsB gene. The transcriptional levels of the rbsB gene, the conjugation-related genes, and the conjugation efficiency in the CRISPRi strain were tested. We further explored the effect of the repressed expression of the rbsB gene on the quorum sensing (QS) system and biofilm formation. The results showed that the constructed CRISPRi system was effective in repressing the transcriptional level of the rbsB gene at a rate of 66.4%. The repressed expression of the rbsB gene resulted in the reduced conjugation rate of RP4 plasmid by 88.7%, which significantly inhibited the expression of the conjugation-related genes (trbBp, trfAp, traF and traJ) and increased the global regulator genes (korA, korB and trbA). The repressed rbsB gene expression reduced the depletion of autoinducer 2 signals (AI-2) by 12.8% and biofilm formation by a rate of 68.2%. The results of this study indicated the rbsB gene could be used as a universal target for the inhibition of conjugation. The constructed conjugative CRISPRi system has the potential to be used in ARG high-risk areas.
Collapse
Affiliation(s)
- Yawen Xiao
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Yan Zhang
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Fengjun Xie
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Rikke Heidemann Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Lei Shi
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Lili Li
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| |
Collapse
|
43
|
Sünderhauf D, Klümper U, Pursey E, Westra ER, Gaze WH, van Houte S. Removal of AMR plasmids using a mobile, broad host-range CRISPR-Cas9 delivery tool. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001334. [PMID: 37226834 PMCID: PMC10268836 DOI: 10.1099/mic.0.001334] [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: 12/30/2022] [Accepted: 04/24/2023] [Indexed: 05/26/2023]
Abstract
Antimicrobial resistance (AMR) genes are widely disseminated on plasmids. Therefore, interventions aimed at blocking plasmid uptake and transfer may curb the spread of AMR. Previous studies have used CRISPR-Cas-based technology to remove plasmids encoding AMR genes from target bacteria, using either phage- or plasmid-based delivery vehicles that typically have narrow host ranges. To make this technology feasible for removal of AMR plasmids from multiple members of complex microbial communities, an efficient, broad host-range delivery vehicle is needed. We engineered the broad host-range IncP1-plasmid pKJK5 to encode cas9 programmed to target an AMR gene. We demonstrate that the resulting plasmid pKJK5::csg has the ability to block the uptake of AMR plasmids and to remove resident plasmids from Escherichia coli. Furthermore, due to its broad host range, pKJK5::csg successfully blocked AMR plasmid uptake in a range of environmental, pig- and human-associated coliform isolates, as well as in isolates of two species of Pseudomonas. This study firmly establishes pKJK5::csg as a promising broad host-range CRISPR-Cas9 delivery tool for AMR plasmid removal, which has the potential to be applied in complex microbial communities to remove AMR genes from a broad range of bacterial species.
Collapse
Affiliation(s)
- David Sünderhauf
- Centre for Ecology and Conservation, University of Exeter, Environment and Sustainability Institute, Penryn, TR10 9FE, UK
| | - Uli Klümper
- Institute of Hydrobiology, Technische Universität Dresden, 01217 Dresden, Germany
| | - Elizabeth Pursey
- Centre for Ecology and Conservation, University of Exeter, Environment and Sustainability Institute, Penryn, TR10 9FE, UK
| | - Edze R. Westra
- Centre for Ecology and Conservation, University of Exeter, Environment and Sustainability Institute, Penryn, TR10 9FE, UK
| | - William H. Gaze
- European Centre for Environment and Human Health, University of Exeter Medical School, Environment and Sustainability Institute, Penryn, TR10 9FE, UK
| | - Stineke van Houte
- Centre for Ecology and Conservation, University of Exeter, Environment and Sustainability Institute, Penryn, TR10 9FE, UK
| |
Collapse
|
44
|
Hilal MG, Han B, Yu Q, Feng T, Su W, Li X, Li H. Insight into the dynamics of drinking water resistome in China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 322:121185. [PMID: 36736566 DOI: 10.1016/j.envpol.2023.121185] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
Antibiotic resistance (AR) is a serious environmental hazard of the current age. Antibiotic resistance genes (ARGs) are the fundamental entities that spread AR in the environment. ARGs are likely to be transferred from the non-pathogenic to pathogenic microbes that might ultimately be responsible for the AR in humans and other organisms. Drinking water (DW) is the primary interaction route between ARGs and humans. Being the highest producer and consumer of antibiotics China poses a potential threat to developing superbugs and ARGs dissemination. Herein, we comprehensively seek to review the ARGs from dominant DW sources in China. Furthermore, the origin and influencing factors of the ARGs to the DW in China have been evaluated. Commonly used methods, both classical and modern, are being compiled. In addition, the risk posed and mitigation strategies of DW ARGs in China have been outlined. Overall, we believe this review would contribute to the assessment of ARGs in DW of China and their dissemination to humans and other animals and ultimately help the policymakers and scientists in the field to counteract this problem on an emergency basis.
Collapse
Affiliation(s)
- Mian Gul Hilal
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China; MOE, Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China
| | - Binghua Han
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Qiaoling Yu
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Tianshu Feng
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Wanghong Su
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Xiangkai Li
- MOE, Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China
| | - Huan Li
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
45
|
Exploiting a conjugative endogenous CRISPR-Cas3 system to tackle multidrug-resistant Klebsiella pneumoniae. EBioMedicine 2023; 88:104445. [PMID: 36696817 PMCID: PMC9879765 DOI: 10.1016/j.ebiom.2023.104445] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/25/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Mobile plasmids play a key role in spurring the global dissemination of multidrug-resistant (MDR) K. pneumoniae, while plasmid curing has been recognized as a promising strategy to combat antimicrobial resistance. Here we exploited a K. pneumoniae native CRISPR system to cure the high-risk IncFII plasmids. METHODS We examined matched protospacers in 725 completely sequenced IncFII plasmids from K. pneumoniae genomes. Then, we re-engineered a native CRISPR-Cas3 system and deliver the CRISPR-Cas3 system via conjugation. Plasmid killing efficiency and G. mellonella infection model were applied to evaluate the CRISPR-Cas3 immunity in vitro and in vivo. FINDINGS Genomic analysis revealed that most IncFII plasmids could be targeted by the native CRISPR-Cas3 system with multiple matched protospacers, and the targeting regions were highly conserved across different IncFII plasmids. This conjugative endogenous CRISPR-Cas3 system demonstrated high plasmid curing efficiency in vitro (8-log decrease) and in vivo (∼100% curing) in a Galleria mellonella infection model, as well as provided immunization against the invasion of IncFII plasmids once the system entering a susceptible bacterial host. INTERPRETATION Overall, our work demonstrated the applicability of using native CRISPR-mediated plasmid curing to re-sensitize drug-resistant K. pneumoniae to multiple antibiotics. This work provided strong support for the idea of utilizing native CRISPR-Cas systems to tackle AMR in K. pneumoniae. FUNDING This work was supported by research grants National Natural Science Foundation of China [grant numbers 81871692, 82172315, 82102439, and 82202564], the Shanghai Science and Technology Commission [grant number 19JC1413002], and Shanghai Sailing Program [grant number 22YF1437500].
Collapse
|
46
|
Lahiri D, Nag M, Dey A, Sarkar T, Pati S, Nirmal NP, Ray RR, Upadhye VJ, Pandit S, Moovendhan M, Kavisri M. Marine bioactive compounds as antibiofilm agent: a metabolomic approach. Arch Microbiol 2023; 205:54. [PMID: 36602609 DOI: 10.1007/s00203-022-03391-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/17/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023]
Abstract
The ocean is a treasure trove of both living and nonliving creatures, harboring incredibly diverse group of organisms. A plethora of marine sourced bioactive compounds are discovered over the past few decades, many of which are found to show antibiofilm activity. These are of immense clinical significance since the formation of microbial biofilm is associated with the development of high antibiotic resistance. Biofilms are also responsible to bring about problems associated with industries. In fact, the toilets and wash-basins also show degradation due to development of biofilm on their surfaces. Antimicrobial resistance exhibited by the biofilm can be a potent threat not only for the health care unit along with industries and daily utilities. Various recent studies have shown that the marine members of various kingdom are capable of producing antibiofilm compounds. Many such compounds are with unique structural features and metabolomics approaches are essential to study such large sets of metabolites. Associating holobiome metabolomics with analysis of their chemical attribute may bring new insights on their antibiofilm effect and their applicability as a substitute for conventional antibiotics. The application of computer-aided drug design/discovery (CADD) techniques including neural network approaches and structured-based virtual screening, ligand-based virtual screening in combination with experimental validation techniques may help in the identification of these molecules and evaluation of their drug like properties.
Collapse
Affiliation(s)
- Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata, 700160, West Bengal, India
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata, 700160, West Bengal, India
| | - Ankita Dey
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, West Bengal, India
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda, 732102, West Bengal, India
| | - Siddhartha Pati
- Nat Nov Bioscience Private Limited, Balasore, 756001, Odisha, India
| | - Nilesh P Nirmal
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, 73170, Nakhon Pathom, Thailand.
| | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, West Bengal, India.
| | - Vijay Jagdish Upadhye
- Center of Research for Development (CR4D), Parul Institute of Applied Sciences (PIAS), Parul University, Vadodara, Gujarat, India
| | - Soumya Pandit
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, 201306, India
| | - M Moovendhan
- Centre for Ocean Research (DST-FIST Sponsored Centre) MoES-Earth Science & Technology Cell, Col. Dr. Jeppiaar Research Park, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - M Kavisri
- Department of Civil Engineering, School of Building and Environment, Sathyabama Institute of Science and Technology, Chennai, 600119, India
| |
Collapse
|
47
|
Al-Madboly LA. A Novel Triple Combination To Combat Serious Infections with Carbapenem-Resistant Acinetobacter baumannii in a Mouse Pneumonia Model. Microbiol Spectr 2022; 10:e0271021. [PMID: 35975993 PMCID: PMC9603289 DOI: 10.1128/spectrum.02710-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/17/2022] [Indexed: 01/04/2023] Open
Abstract
The ongoing crisis of antimicrobial resistance demands novel combinations between antimicrobials and nonantimicrobials to manage infections caused by highly resistant pathogens. This study aimed to evaluate the effect of combining sodium ascorbate and/or apo-transferrin with imipenem, forming double and triple combinations, against 20 multiple-carbapenemase-producing Acinetobacter baumannii strains using the checkerboard test, time-kill assay, and disc diffusion test. The results of the checkerboard assay revealed that all double combinations showed indifference, while only triple combination recorded a synergistic effect (fractional inhibitory concentration index [FICI] < 0.8) in 95% the test isolates. Moreover, the MIC of imipenem (MICimp) was strongly reduced (up to 128-fold reduction) after treatment with the triple combination against highly resistant isolates and reached the susceptible range. The time-kill assay revealed that the triple combination led to a 4-log10 reduction in the CFU at 8 h compared with the initial bacterial count, and no viable count was recorded at 10 h. The mouse pneumonia model showed restoration of lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following administration of the triple rescue therapy. Additionally, normal lungs with normal patent alveoli were detected 72 h following treatment. Accordingly, sodium ascorbate and apo-transferrin are promising adjunct biological agents with the potential to restore the effectiveness of critically essential antibiotics like imipenem, commonly used for the treatment of A. baumannii infections. IMPORTANCE Combination therapy provides a perspective to threat multidrug-resistant (MDR) strains. The present study sheds light on a novel and effective triple combination against carbapenem-resistant A. baumannii. Our in vitro results showed that combining imipenem with apo-transferrin and sodium ascorbate yielded synergism in 95% of test isolates, and this was associated with a marked reduction in imipenem MIC, shifting it below the breakpoint. Furthermore, a bactericidal effect was recorded, with no viable count detected at 10 h. An in vivo murine model of pneumonia was induced to mimic human disease. The triple combination therapy restored lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following the initiation of therapy. Therefore, our findings suggest novel insights about a promising new combination therapy against highly resistant carbapenemase-producing A. baumannii to restore the effectiveness of imipenem.
Collapse
Affiliation(s)
- Lamiaa A. Al-Madboly
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
48
|
de Nies L, Busi SB, Kunath BJ, May P, Wilmes P. Mobilome-driven segregation of the resistome in biological wastewater treatment. eLife 2022; 11:81196. [PMID: 36111782 PMCID: PMC9643006 DOI: 10.7554/elife.81196] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2022] Open
Abstract
Biological wastewater treatment plants (BWWTP) are considered to be hotspots for the evolution and subsequent spread of antimicrobial resistance (AMR). Mobile genetic elements (MGEs) promote the mobilization and dissemination of antimicrobial resistance genes (ARGs) and are thereby critical mediators of AMR within the BWWTP microbial community. At present, it is unclear whether specific AMR categories are differentially disseminated via bacteriophages (phages) or plasmids. To understand the segregation of AMR in relation to MGEs, we analyzed meta-omic (metagenomic, metatranscriptomic and metaproteomic) data systematically collected over 1.5 years from a BWWTP. Our results showed a core group of 15 AMR categories which were found across all timepoints. Some of these AMR categories were disseminated exclusively (bacitracin) or primarily (aminoglycoside, MLS and sulfonamide) via plasmids or phages (fosfomycin and peptide), whereas others were disseminated equally by both. Combined and timepoint-specific analyses of gene, transcript and protein abundances further demonstrated that aminoglycoside, bacitracin and sulfonamide resistance genes were expressed more by plasmids, in contrast to fosfomycin and peptide AMR expression by phages, thereby validating our genomic findings. In the analyzed communities, the dominant taxon Candidatus Microthrix parvicella was a major contributor to several AMR categories whereby its plasmids primarily mediated aminoglycoside resistance. Importantly, we also found AMR associated with ESKAPEE pathogens within the BWWTP, and here MGEs also contributed differentially to the dissemination of the corresponding ARGs. Collectively our findings pave the way toward understanding the segmentation of AMR within MGEs, thereby shedding new light on resistome populations and their mediators, essential elements that are of immediate relevance to human health.
Collapse
Affiliation(s)
- Laura de Nies
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg
| | | | | | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg
| |
Collapse
|
49
|
Uddin Mahamud AGMS, Nahar S, Ashrafudoulla M, Park SH, Ha SD. Insights into antibiofilm mechanisms of phytochemicals: Prospects in the food industry. Crit Rev Food Sci Nutr 2022; 64:1736-1763. [PMID: 36066482 DOI: 10.1080/10408398.2022.2119201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The recalcitrance of microbial aggregation or biofilm in the food industry underpins the emerging antimicrobial resistance among foodborne pathogens, exacerbating the phenomena of food spoilage, processing and safety management failure, and the prevalence of foodborne illnesses. The challenges of growing tolerance to current chemical and disinfectant-based antibiofilm strategies have driven the urgency in finding a less vulnerable to bacterial resistance, effective alternative antibiofilm agent. To address these issues, various novel strategies are suggested in current days to combat bacterial biofilm. Among the innovative approaches, phytochemicals have already demonstrated their excellent performance in preventing biofilm formation and bactericidal actions against resident bacteria within biofilms. However, the diverse group of phytochemicals and their different modes of action become a barrier to applying them against specific pathogenic biofilm-formers. This phenomenon mandates the need to elucidate the multi-mechanistic actions of phytochemicals to design an effective novel antibiofilm strategy. Therefore, this review critically illustrates the structure - activity relationship, functional sites of actions, and target molecules of diverse phytochemicals regarding multiple major antibiofilm mechanisms and reversal mechanisms of antimicrobial resistance. The implementation of the in-depth knowledge will hopefully aid future studies for developing phytochemical-based next-generation antimicrobials.
Collapse
Affiliation(s)
- A G M Sofi Uddin Mahamud
- School of Food Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Shamsun Nahar
- School of Food Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Md Ashrafudoulla
- School of Food Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Si Hong Park
- Department of Food Science and Technology, Oregon State University, Corvallis, OR, USA
| | - Sang-Do Ha
- School of Food Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
50
|
Shan X, Li XS, Schwarz S, Chen Y, Xu C, Du XD. Plasmid-Assisted Horizontal Transfer of a Large lsa(E)-Carrying Genomic Island in Enterococcus faecalis. Microbiol Spectr 2022; 10:e0015422. [PMID: 35863017 PMCID: PMC9430800 DOI: 10.1128/spectrum.00154-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022] Open
Abstract
The horizontal transfer of genomic islands is essential for the adaptation and evolution of Enterococcus faecalis. In this study, three porcine E. faecalis strains, each harboring a large lsa(E)-carrying genomic island, were identified. When using the E. faecalis OG1RF as the recipient, the horizontal transfer of the lsa(E)-carrying genomic island occurred only from E. faecalis E512, which also harbored a pheromone-responsive conjugative plasmid, but not from the other two E. faecalis strains, E533 and E509, which lacked such a plasmid. Subsequently, through plasmid curing of E. faecalis E512 and plasmid introduction into E. faecalis E533, the pheromone-responsive conjugative plasmid was identified to be indispensable for the horizontal transfer of the lsa(E)-carrying genomic island and a subsequent homologous recombination between the chromosomal DNA of the donor and the recipient. In addition, the presence of a chromosomally-located conjugative transposon, Tn916, in E. faecalis E509 could not mediate the horizontal transfer of the lsa(E)-carrying genomic island, although Tn916 itself could transfer by conjugation. Thus, these data highlight the role of the pheromone-responsive conjugative plasmid in the transfer of the lsa(E)-carrying genomic island in E. faecalis, thereby establishing the dual role of pheromone-responsive conjugative plasmids in contributing to the dissemination of both plasmid-borne resistance genes and chromosomally-located genomic islands. IMPORTANCE In this study, it was shown that a pheromone-responsive conjugative plasmid played an indispensable role in the horizontal transfer of a lsa(E)-carrying genomic island. This finding indicates a dual role of the pheromone-responsive conjugative plasmid in disseminating both plasmid-borne resistance genes and chromosomally-located genomic islands. The role of the pheromone-responsive conjugative plasmid in disseminating chromosomal genomic islands is suggested to be essential in the genomic evolution of E. faecalis, which has become one of the leading nosocomial pathogens worldwide.
Collapse
Affiliation(s)
- Xinxin Shan
- College of Veterinary Medicine, Zhengzhou, People’s Republic of China
| | - Xin-Sheng Li
- College of Veterinary Medicine, Zhengzhou, People’s Republic of China
| | - Stefan Schwarz
- Institute of Microbiology and Epizootics, Centre for Infection Medicine, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre of Resistance Research (TZR), Freie Universität Berlin, Berlin, Germany
| | - Yuxia Chen
- College of Veterinary Medicine, Zhengzhou, People’s Republic of China
| | - Chunyan Xu
- College of Veterinary Medicine, Zhengzhou, People’s Republic of China
| | - Xiang-Dang Du
- College of Veterinary Medicine, Zhengzhou, People’s Republic of China
| |
Collapse
|