Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.669
Filtrar
1.
Pol J Microbiol ; 73(3): 349-362, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39268957

RESUMEN

The aim of this work was to provide a theoretical and scientific basis for the treatment, prevention, and control of clinical drug-resistant bacterial infections by studying the molecular epidemiology and horizontal transfer mechanism of optrA-carrying linezolid-resistant Enterococcus faecalis strains (LREfs) that were clinically isolated in a tertiary hospital in Kunming, China. Non-repetitive LREfs retained in a tertiary A hospital in Kunming, China. The strains were identified by Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). The transferability and horizontal transfer mechanism of optrA gene were analyzed using polymerase chain reaction (PCR), whole-genome sequencing (WGS), and conjugation experiments. A total of 39 LREfs strains were collected, and all of them were multi-drug resistant. There were 30 LREfs strains (76.9%) carrying the optrA gene, The cfr, poxtA genes and mutations in the 23S rRNA gene were not detected. The conjugation experiments showed that only three of 10 randomly selected optrA-carrying LREfs were successfully conjugated with JH2-2. Further analysis of one successfully conjugated strain revealed that the optrA gene, located in the donor bacterium, formed the IS1216E-erm(A)-optrA-fexA-IS1216E transferable fragment under the mediation of the mobile genetic element (MGE) IS1216E, which was then transferred to the recipient bacterium via horizontal plasmid transfer. Carrying the optrA gene is the primary resistance mechanism of LREfs strains. The optrA gene could carry the erm(A) and fexA genes to co-transfer among E. faecalis. MGEs such as insertion sequence IS1216E play an important role in the horizontal transfer of the optrA gene.


Asunto(s)
Antibacterianos , Enterococcus faecalis , Transferencia de Gen Horizontal , Infecciones por Bacterias Grampositivas , Linezolid , Enterococcus faecalis/genética , Enterococcus faecalis/efectos de los fármacos , Linezolid/farmacología , Antibacterianos/farmacología , Humanos , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/epidemiología , China/epidemiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Epidemiología Molecular , Farmacorresistencia Bacteriana/genética , Pruebas de Sensibilidad Microbiana , Farmacorresistencia Bacteriana Múltiple/genética , Secuenciación Completa del Genoma , Conjugación Genética
2.
Microb Biotechnol ; 17(9): e70015, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39315602

RESUMEN

Wastewater treatment plants are one of the major pathways for microplastics to enter the environment. In general, microplastics are contaminants of global concern that pose risks to ecosystems and human health. Here, we present a proof-of-concept for reduction of microplastic pollution emitted from wastewater treatment plants: delivery of recombinant DNA to bacteria in wastewater to enable degradation of polyethylene terephthalate (PET). Using a broad-host-range conjugative plasmid, we enabled various bacterial species from a municipal wastewater sample to express FAST-PETase, which was released into the extracellular environment. We found that FAST-PETase purified from some transconjugant isolates could degrade about 40% of a 0.25 mm thick commercial PET film within 4 days at 50°C. We then demonstrated partial degradation of a post-consumer PET product over 5-7 days by exposure to conditioned media from isolates. These results have broad implications for addressing the global plastic pollution problem by enabling environmental bacteria to degrade PET.


Asunto(s)
Bacterias , Biodegradación Ambiental , Tereftalatos Polietilenos , Aguas Residuales , Tereftalatos Polietilenos/metabolismo , Tereftalatos Polietilenos/química , Aguas Residuales/microbiología , Aguas Residuales/química , Bacterias/metabolismo , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/clasificación , Conjugación Genética , Plásmidos/genética , Contaminantes Químicos del Agua/metabolismo
3.
World J Microbiol Biotechnol ; 40(10): 319, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39261358

RESUMEN

The present work reports the development and validation of a chromosomal expression system in Streptococcus pneumoniae which permits gene expression under the control of Lactococcus lactis lantibiotic nisin. The system is based on the integrative and conjugative element (ICE) Tn5253 of S. pneumoniae capable of site-specific chromosomal integration and conjugal transfer to a variety of bacterial species. We constructed an insertion vector that integrates in Tn5251, an ICE contained in Tn5253, which carries the tetracycline resistance tet(M) gene. The vector contains the nisRK regulatory system operon, the L. lactis nisin inducible promoter PnisA upstream of a multiple cloning site for target DNA insertion, and is flanked by two DNA regions of Tn5251 which drive homologous recombination in ICE Tn5253. For system evaluation, the emm6.1::ha1 fusion gene was cloned and integrated into the chromosome of the Tn5253-carrying pneumococcal strain FR24 by transformation. This gene encodes a fusion protein containing the signal peptide, the 122 N-terminal and the 140 C-terminal aa of the Streptococcus pyogenes M6 surface protein joined to the HA1 subunit of the influenza virus A hemagglutinin. Quantitative RT-PCR analysis carried out on total RNA purified from nisin treated and untreated cultures showed an increase in emm6.1::ha1 transcript copy number with growing nisin concentration. The expression of M6-HA1 protein was detected by Western blot and quantified by Dot blot, while Flow cytometry analysis confirmed the presence on the pneumococcal surface. Recombinant ICE Tn5253::[nisRK]-[emm6.1::ha1] containing the nisin-inducible expression system was successfully transferred by conjugation in different streptococcal species including Streptococcus gordonii, S. pyogenes, Streptococcus agalactiae and Enterococcus faecalis. As for S. pneumoniae, the emm6.1::ha1 transcript copy number and the amount of M6-HA1 protein produced correlated with the nisin concentration used for induction in all investigated bacterial hosts. We demonstrated that this host-vector expression system is stably integrated as a single copy within the bacterial chromosome, is transferable to both transformable and non transformable bacterial species, and allows fine tuning of protein expression modulated by nisin concentration. These characteristics make our system suitable for a wide range of applications including complementation assays, physiological studies, host-pathogen interaction studies.


Asunto(s)
Cromosomas Bacterianos , Elementos Transponibles de ADN , Nisina , Streptococcus pneumoniae , Nisina/farmacología , Nisina/genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/efectos de los fármacos , Cromosomas Bacterianos/genética , Elementos Transponibles de ADN/genética , Regulación Bacteriana de la Expresión Génica , Enterococcus/genética , Enterococcus/efectos de los fármacos , Vectores Genéticos/genética , Conjugación Genética , Streptococcus/genética , Streptococcus/efectos de los fármacos , Streptococcus/metabolismo , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
4.
Curr Microbiol ; 81(10): 344, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39235595

RESUMEN

The problem of bacterial resistance caused by antibiotic abuse is seriously detrimental to global human health and ecosystem security. The two-dimensional nanomaterial (2D) such as black phosphorus (BP) is recently expected to become a new bacterial inhibitor and has been widely used in the antibacterial field due to its specific physicochemical properties. Nevertheless, the effects of 2D-BP on the propagation of antibiotic resistance genes (ARGs) in environments and the relevant mechanisms are not clear. Herein, we observed that the sub-inhibitory concentrations of 2D-BP dramatically increased the conjugative transfer of ARGs mediated by the RP4 plasmid up to 2.6-fold at the 125 mg/L exposure level compared with the untreated bacterial cells. Nevertheless, 2D-BP with the inhibitory concentration caused a dramatic decrease in the conjugative frequency. The phenotypic changes revealed that the increase of the conjugative transfer caused by 2D-BP exposure were attributed to the excessive reactive oxygen species and oxidative stress, and increased bacterial cell membrane permeability. The genotypic evidence demonstrated that 2D-BP affecting the horizontal gene transfer of ARGs was probably through the upregulation of mating pair formation genes (trbBp and traF) and DNA transfer and replication genes (trfAp and traJ), as well as the downregulation of global regulatory gene expression (korA, korB, and trbA). In summary, the changes in the functional and regulatory genes in the conjugative transfer contributed to the stimulation of conjugative transfer. This research aims to broaden our comprehension of how nanomaterials influence the dissemination of ARGs by elucidating their effects and mechanisms.


Asunto(s)
Antibacterianos , Farmacorresistencia Bacteriana , Transferencia de Gen Horizontal , Fósforo , Plásmidos , Plásmidos/genética , Fósforo/metabolismo , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Conjugación Genética , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Nanoestructuras , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Bacterias/genética , Bacterias/efectos de los fármacos
5.
J Hazard Mater ; 478: 135436, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39141944

RESUMEN

Plasmid-mediated conjugative transfer has emerged as a major driver accounting for the dissemination of antibiotic resistance genes (ARGs). In addition to the use of antimicrobial agents, there is growing evidence that non-antibiotic factors also play an important role. Pesticides are widely used to protect crops against vectors of diseases, and are indispensable agents in agricultural production, whereas the impact of pesticide pollution on the transmission of antimicrobial resistance remains poorly understood. Here we reveal that the pesticides at environmentally relevant concentrations, especially cyromazine (Cyr) and kresoxim-methyl (Kre), greatly facilitate the conjugative transfer of antibiotic-resistance plasmids carrying clinically important ARGs. Mechanistic studies indicate that Cyr and Kre treatments trigger reactive oxygen species (ROS) production and SOS response, increase membrane permeability, upregulate bacterial proton motive force (PMF) and promote ATP supply. Further non-targeted metabolomics and biochemical analysis demonstrate that the addition of Cyr and Kre accelerates tricarboxylic acid (TCA) cycle and electron transport chain (ETC), thereby activating bacterial energy metabolism. In the constructed soil model, we prove that two pesticides contribute to the dissemination of resistance plasmids in the soil microbiota. 16S rRNA sequencing analyses indicate that pesticides alter transconjugant microbial communities, and enable more opportunistic pathogens, such as Pseudomonas and Enterobacter, to acquire the multidrug resistance plasmids. Collectively, our work indicates the potential risk in accelerating the spread of antimicrobial resistance owing to pesticide pollution, highlighting the importance of continuous surveillance of pesticide residues in complex environmental settings.


Asunto(s)
Plaguicidas , Plásmidos , Plásmidos/genética , Plaguicidas/toxicidad , Microbiología del Suelo , Farmacorresistencia Bacteriana Múltiple/genética , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Conjugación Genética/efectos de los fármacos , Bacterias/genética , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Antibacterianos/toxicidad , Antibacterianos/farmacología , Microbiota/efectos de los fármacos
6.
Front Cell Infect Microbiol ; 14: 1445850, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39108982

RESUMEN

Plasmid-mediated conjugation is a common mechanism for most bacteria to transfer antibiotic resistance genes (ARGs). The conjugative transfer of ARGs is emerging as a major threat to human beings. Although several transfer-related factors are known to regulate this process, small RNAs (sRNAs)-based regulatory roles remain to be clarified. Here, the Hfq-binding sRNA GadY in donor strain Escherichia coli (E. coli) SM10λπ was identified as a new regulator for bacterial conjugation. Two conjugation models established in our previous studies were used, which SM10λπ carrying a chromosomally integrated IncP-1α plasmid RP4 and a mobilizable plasmid pUCP24T served as donor cells, and P. aeruginosa PAO1 or E. coli EC600 as the recipients. GadY was found to promote SM10λπ-PAO1 conjugation by base-pairing with its target mRNA SdiA, an orphan LuxR-type receptor that responds to exogenous N-acylated homoserine lactones (AHLs). However, SM10λπ-EC600 conjugation was not affected due to EC600 lacking AHLs synthase. It indicates that the effects of GadY on conjugation depended on AHLs-SdiA signalling. Further study found GadY bound SdiA to negatively regulate the global RP4 repressors KorA and KorB. When under ciprofloxacin or levofloxacin treatment, GadY expression in donor strain was enhanced, and it positively regulated quinolone-induced SM10λπ-PAO1 conjugation. Thus, our study provides a novel role for sRNA GadY in regulating plasmid-mediated conjugation, which helps us better understand bacterial conjugation to counter antibiotic resistance.


Asunto(s)
Conjugación Genética , Proteínas de Escherichia coli , Escherichia coli , Plásmidos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Plásmidos/genética , Regulación Bacteriana de la Expresión Génica , Transactivadores/genética , Transactivadores/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/metabolismo , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Antibacterianos/farmacología , Proteína de Factor 1 del Huésped/genética , Proteína de Factor 1 del Huésped/metabolismo
7.
Environ Pollut ; 360: 124676, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39103039

RESUMEN

The emergence and spread of antibiotic resistance in the environment pose a serious threat to global public health. It is acknowledged that non-antibiotic stresses, including disinfectants, pharmaceuticals and organic pollutants, play a crucial role in horizontal transmission of antibiotic resistance genes (ARGs). Despite the widespread presence of non-steroidal anti-inflammatory drugs (NSAIDs), notably in surface water, their contributions to the transfer of ARGs have not been systematically explored. Furthermore, previous studies have primarily concentrated on model strains to investigate whether contaminants promote the conjugative transfer of ARGs, leaving the mechanisms of ARG transmission among antibiotic resistant bacteria in natural aqueous environments under the selective pressures of non-antibiotic contaminants remains unclear. In this study, the Escherichia coli (E. coli) K12 carrying RP4 plasmid was used as the donor strain, indigenous strain Aeromonas veronii containing rifampicin resistance genes in Taihu Lake, and E. coli HB101 were used as receptor strains to establish inter-genus and intra-genus conjugative transfer systems, examining the conjugative transfer frequency under the stress of ketoprofen. The results indicated that ketoprofen accelerated the environmental spread of ARGs through several mechanisms. Ketoprofen promoted cell-to-cell contact by increasing cell surface hydrophobicity and reducing cell surface charge, thereby mitigating cell-to-cell repulsion. Furthermore, ketoprofen induced increased levels of reactive oxygen species (ROS) production, activated the DNA damage-induced response (SOS), and enhanced cell membrane permeability, facilitating ARG transmission in intra-genus and inter-genus systems. The upregulation of outer membrane proteins, oxidative stress, SOS response, mating pair formation (Mpf) system, and DNA transfer and replication (Dtr) system related genes, as well as the inhibition of global regulatory genes, all contributed to higher transfer efficiency under ketoprofen treatment. These findings served as an early warning for a comprehensive assessment of the roles of NSAIDs in the spread of antibiotic resistance in natural aqueous environments.


Asunto(s)
Transferencia de Gen Horizontal , Cetoprofeno , Cetoprofeno/farmacología , Conjugación Genética , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Microbiana/genética , Contaminantes Químicos del Agua/toxicidad , Antiinflamatorios no Esteroideos/farmacología
8.
J Hazard Mater ; 479: 135639, 2024 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-39191006

RESUMEN

Chlormequat (CCC) is widely used in agricultural production to increase the crop yield. However, the effects of CCC on transfer of ARGs in agricultural system are still unclear. In this study, using E.coli DH5α (carrying RP4 plasmid with AmpR, TetR, KanR) as the donor bacterium, E.coli HB101, endophytic Pseudomonas sp. Ph6 or rhizosphere Pseudomonas putida KT2440 as the recipient strain, three conjugative systems were designed to investigate the effects of CCC on ARG transfer. Meanwhile, hydroponics experiments were designed to study the ARG spread in the rice-nutrient solution system after CCC application. The results showed that CCC significantly promoted the RP4 conjugation by expanding cell membrane permeability and improving the relative transcription levels of trfAp, trbBp, traA and traL genes in RP4. Furthermore, the conjugation frequency between E. coli and Pseudomonas was much higher than that between E. coli cells. Compared with spraying foliage with 2500 mg·L-1 of CCC, soaking seeds with 250 mg·L-1 of CCC was more beneficial to the colonization of ARB in rice, and also increased the abundance of ARGs in rice cultivation system. These results remind that the use of CCC in agricultural production might promote the ARG transmission in agro-ecosystems; however, foliage spraying with 2500 mg·L-1 of CCC could control its spread.


Asunto(s)
Conjugación Genética , Escherichia coli , Transferencia de Gen Horizontal , Oryza , Plásmidos , Pseudomonas , Plásmidos/genética , Oryza/microbiología , Oryza/crecimiento & desarrollo , Oryza/efectos de los fármacos , Oryza/genética , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Pseudomonas/genética , Pseudomonas/efectos de los fármacos , Pseudomonas/metabolismo , Pseudomonas putida/genética , Pseudomonas putida/efectos de los fármacos , Pseudomonas putida/metabolismo , Genes Bacterianos/efectos de los fármacos , Antibacterianos/farmacología , Agricultura , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Microbiana/genética
9.
J Hazard Mater ; 477: 135403, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39096644

RESUMEN

Drug resistance poses a high risk to human health. Extensive use of non-antibiotic drugs contributes to antibiotic resistance genes (ARGs) transfer. However, how they affect the spread of broad-host plasmids in complex biological systems remains unknown. This study investigated the effect of metoprolol on the transfer frequency and host range of ARGs in both intrageneric and intergeneric pure culture systems, as well as in anammox microbiome. The results showed that environmental concentrations of metoprolol significantly promoted the intrageneric and intergeneric conjugative transfer. Initially, metoprolol induced excessive oxidative stress, resulting in high cell membrane permeability and bacterial SOS response. Meanwhile, more pili formation increased the adhesion and contact between bacteria, and the abundance of conjugation-related genes also increased significantly. Activation of the electron transport chain provided more ATP for this energy-consuming process. The underlying mechanism was further verified in the complex anammox conjugative system. Metoprolol induced the enrichment of ARGs and mobile genetic elements. The enhanced bacterial interaction and energy generation facilitated the high conjugative transfer frequency of ARGs. In addition, plasmid-borne ARGs tended to transfer to opportunistic pathogens. This work raises public concerns about the health and ecological risks of non-antibiotic drugs.


Asunto(s)
Conjugación Genética , Metoprolol , Plásmidos , Plásmidos/genética , Conjugación Genética/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Antagonistas Adrenérgicos beta/farmacología , Transferencia de Gen Horizontal , Bacterias/genética , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Antibacterianos/farmacología , Genes MDR/genética , Microbiota/efectos de los fármacos
10.
Microbiology (Reading) ; 170(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39190025

RESUMEN

Antimicrobial resistance (AMR) poses a significant threat to global public health. Notably, resistance to carbapenem and extended-spectrum ß-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 ß-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.


Asunto(s)
Antibacterianos , Productos Biológicos , Escherichia coli , Klebsiella pneumoniae , Plásmidos , beta-Lactamasas , Plásmidos/genética , Antibacterianos/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Productos Biológicos/farmacología , Farmacorresistencia Bacteriana/genética , Conjugación Genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Enterobacteriaceae/efectos de los fármacos , Enterobacteriaceae/genética , Pruebas de Sensibilidad Microbiana , Microbiología de Alimentos , Transferencia de Gen Horizontal
11.
Methods Mol Biol ; 2851: 23-37, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39210169

RESUMEN

Lactobacillus bulgaricus is an industrial strain that has been used in the dairy products since ancient times. Because of the difficulty of chromosomal gene manipulation, there have been few reports of gene deletion, insertion, or replacement. We have developed a system that enables chromosomal gene manipulation of L. bulgaricus using a conjugal transfer vector and easily vector construction in E. coli. As an example, we have deleted a regulatory gene for the extracellular polysaccharide synthesis of L. bulgaricus to elucidate the function of the gene in question. Methods for constructing vectors for chromosomal integration, conjugation experiment, and obtaining deletion strains by double recombination were presented in detail. This conjugative shuttle vector, pGMß1, has been deposited at Addgene ( https://www.addgene.org )and can be used by anyone for academic purposes.


Asunto(s)
Cromosomas Bacterianos , Vectores Genéticos , Lactobacillus delbrueckii , Lactobacillus delbrueckii/genética , Vectores Genéticos/genética , Cromosomas Bacterianos/genética , Conjugación Genética , Escherichia coli/genética , Plásmidos/genética , Eliminación de Gen , Ingeniería Genética/métodos
12.
Microb Ecol ; 87(1): 109, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39198281

RESUMEN

Mating assays are common laboratory experiments for measuring the conjugation frequency, i.e. efficiency at which a plasmid transfers from a population of donor cells to a population of recipient cells. Selective plating remains a widely used quantification method to enumerate transconjugants at the end of such assays. However, conjugation frequencies may be inaccurately estimated because plasmid transfer can occur on transconjugant-selective plates rather than only during the intended mating duration. We investigated the influence of cell density on this phenomenon. We conducted mating experiments with IncPα plasmid RP4 harbored in Escherichia coli at a fixed cell density and mating conditions, inoculated a serial dilution of the mating mixture on transconjugant-selective plates or in transconjugant-selective broth, and compared the results to a model of cell-to-cell distance distribution. Our findings suggest that irrespective of the mating mode (liquid vs solid), the enumeration of transconjugants becomes significantly biased if the plated cell density exceeds 28 Colony Forming Unit (CFU)/mm2 (or 1.68•105 CFU/standard 9 cm Petri dish). This threshold is determined with a 95% confidence interval of ± 4 CFU/mm2 (± 2.46•104 CFU/standard 9 cm Petri dish). Liquid mating assays were more sensitive to this bias because the conjugation frequency of RP4 is several orders of magnitude lower in suspension compared to surface mating. Therefore, if selective plating is used, we recommend to plate at this density threshold and that negative controls are performed where donors and recipients are briefly mixed before plating at the same dilutions as for the actual mating assay. As an alternative, a liquid enumeration method can be utilized to increase the signal-to-noise ratio and allow for more accurate enumeration of transconjugants.


Asunto(s)
Conjugación Genética , Escherichia coli , Plásmidos , Escherichia coli/genética , Plásmidos/genética , Transferencia de Gen Horizontal
13.
BMC Genomics ; 25(1): 734, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39080539

RESUMEN

Dairy industries apply selected lactococcal strains and mixed cultures to produce diverse fermented products with distinctive flavor and texture properties. Innovation of the starter culture functionality in cheese applications embraces natural biodiversity of the Lactococcus species to identify novel strains with alternative flavor or texture forming capacities and/or increased processing robustness and phage resistance. Mobile genetic elements (MGE), like integrative conjugative elements (ICEs) play an important role in shaping the biodiversity of bacteria. Besides the genes involved in the conjugation of ICEs from donor to recipient strains, these elements also harbor cargo genes that encode a wide range of functions. The definition of such cargo genes can only be achieved by accurate identification of the ICE boundaries (delimiting). Here, we delimited 25 ICEs in lactococcal genome sequences with low contig numbers using insertion-sites flanking single-copy core-genome genes as markers for each of the distinct ICE-integrases we identified previously within the conserved ICE-core genes. For ICEs in strains for which genome information with large numbers of contigs is available, we exemplify that CRISPR-Cas9 driven ICE-curing, followed by resequencing, allows accurate delimitation and cargo definition of ICEs. Finally, we compare and contrast the cargo gene repertoire of the 26 delimited lactococcal ICEs, identifying high plasticity among the cargo of lactococccal ICEs and a range of encoded functions that is of apparent industrial interest, including restriction modification, abortive infection, and stress adaptation genes.


Asunto(s)
Genoma Bacteriano , Lactococcus/genética , Secuencias Repetitivas Esparcidas/genética , Sistemas CRISPR-Cas , Conjugación Genética
14.
Int J Antimicrob Agents ; 64(3): 107290, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39084573

RESUMEN

OBJECTIVES: Carbapenem-resistant Klebsiella pneumoniae (CRKP), a superbug that can be difficult or impossible to treat, has become a worldwide problem. This study presents the first report of a CRKP strain carrying a plasmid co-harbouring blaNDM-1, blaKPC-2, and tet(A) and the subsequent analysis of its genomic features. METHODS: Isolation and identification of bacteria, antimicrobial susceptibility test, whole genome sequencing, and conjugation experiments assay were conducted in clinical epidemiological investigations and plasmid genetic characterisation analysis. RESULTS: A total of 116 strains of bacteria were isolated from patients with bloodstream infections (BSI) between 2018 and 2023. A total of 89.66% of the isolates were carbapenem-resistant Enterobacteriaceae (CRE), with the majority (75/116) being CRKP. Among these, a novel plasmid co-harbouring blaNDM-1, blaKPC-2, and tet(A) simultaneously was found in CRKP46, and the three genes mediated conjugation by IS26, ISAba125, and IS26, respectively. This plasmid conferred carbapenem resistance to E. coli J53 after conjugative transfer, which was 2 times greater than that of CRKP46. CONCLUSION: The present study identified the occurrence of a rare plasmid co-harbouring blaNDM-1, blaKPC-2, and tet(A), and the spread of these genes was mediated by the corresponding mobile elements. The increased carbapenem resistance created by this novel plasmid challenges public health security and poses a potential threat to human health; therefore, it deserves attention.


Asunto(s)
Antibacterianos , Proteínas Bacterianas , Farmacorresistencia Bacteriana Múltiple , Infecciones por Klebsiella , Klebsiella pneumoniae , beta-Lactamasas , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antibacterianos/farmacología , Antiportadores , Bacteriemia/microbiología , Proteínas Bacterianas/genética , beta-Lactamasas/genética , Enterobacteriaceae Resistentes a los Carbapenémicos/genética , Enterobacteriaceae Resistentes a los Carbapenémicos/aislamiento & purificación , Enterobacteriaceae Resistentes a los Carbapenémicos/efectos de los fármacos , Conjugación Genética , Farmacorresistencia Bacteriana Múltiple/genética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Plásmidos/genética , Secuenciación Completa del Genoma
15.
J Glob Antimicrob Resist ; 38: 227-230, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39004340

RESUMEN

OBJECTIVES: To investigate the tet(X) gene, a determinant of tigecycline resistance, in the emerging pathogen Elizabethkingia meningoseptica and its association with an integrative and conjugative element (ICE). METHODS: All E. meningoseptica genomes from the National Center for Biotechnology Information (n = 87) were retrieved and annotated for resistome searches using the CARD database. A phylogenic analysis was performed based on the E. meningoseptica core genome. The ICE was identified through comparative genomics with other ICEs occurring in Elizabethkingia spp. RESULTS: Phylogenetic analysis revealed E. meningoseptica genomes from six countries distributed across different lineages, some of which persisted for years. The common resistome of these genomes included blaBlaB, blaCME, blaGOB, ranA/B, aadS, and catB (genes associated with resistance to ß-lactams, aminoglycosides, and chloramphenicol). Some genomes also presented additional resistance genes (dfrA, ereD, blaVEB, aadS, and tet(X)). Interestingly, tet(X) and aadS were located in an ICE of 49 769 bp (ICEEmSQ101), which was fully obtained from the E. meningoseptica SQ101 genome. We also showed evidence that the other 27 genomes harboured this ICE. The distribution of ICEEmSQ101, carrying tet(X), was restricted to a single Chinese lineage. CONCLUSIONS: The tet(X) gene is not prevalent in the species E. meningoseptica, as previously stated for the genus Elizabethkingia, since it is present only in a single Chinese lineage. We identified that several E. meningoseptica genomes harboured an ICE that mobilized the Elizabethkingia tet(X) gene and exhibited characteristics similar to the ICEs of other Flavobacteria, which would favour their transmission in this bacterial family.


Asunto(s)
Antibacterianos , Infecciones por Flavobacteriaceae , Flavobacteriaceae , Genoma Bacteriano , Filogenia , Antibacterianos/farmacología , Infecciones por Flavobacteriaceae/microbiología , Flavobacteriaceae/genética , Flavobacteriaceae/efectos de los fármacos , Flavobacteriaceae/clasificación , Humanos , Pruebas de Sensibilidad Microbiana , Tigeciclina/farmacología , Conjugación Genética , Prevalencia , Farmacorresistencia Bacteriana/genética
16.
Microb Pathog ; 193: 106788, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38986823

RESUMEN

The use of antimicrobials in poultry leaves residues in the litter, favoring the emergence of antimicrobial-resistant pathogens and making it a source of contamination. An in vitro 4 × 4 factorial trial was performed to investigate the influence of four treatments, consisting of antimicrobial sub-concentrations, on the transference of IncB/O-plasmid through conjugation in four groups. Each group was composed of one plasmid donor bacterium (Escherichia coli H2332) and a recipient bacterium (Escherichia coli J62 or Salmonella enterica serovars, Enteritidis, Typhimurium, or Heidelberg). Our results showed a little decrease in the conjugation frequency in almost all treatments between the two bacterial species, which varied according to each strain. The MIC test revealed an increase of up to 4096-fold in resistance to beta-lactams in Salmonella serovars after plasmid acquisition. This finding suggests that some genetic apparatus may be involved in increased antimicrobial resistance in Salmonella serovars after the acquisition of primary resistance determinants.


Asunto(s)
Antibacterianos , Conjugación Genética , Escherichia coli , Pruebas de Sensibilidad Microbiana , Plásmidos , Salmonella enterica , beta-Lactamas , Salmonella enterica/efectos de los fármacos , Salmonella enterica/genética , Plásmidos/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , beta-Lactamas/farmacología , Antibacterianos/farmacología
17.
BMC Microbiol ; 24(1): 240, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961341

RESUMEN

OBJECTIVE: We explored whether the Clustered regularly interspaced short palindromic repeat (CRISPR)-Cas and restriction-modification (R-M) systems are compatible and act together to resist plasmid attacks. METHODS: 932 global whole-genome sequences from GenBank, and 459 K. pneumoniae isolates from six provinces of China, were collected to investigate the co-distribution of CRISPR-Cas, R-M systems, and blaKPC plasmid. Conjugation and transformation assays were applied to explore the anti-plasmid function of CRISPR and R-M systems. RESULTS: We found a significant inverse correlation between the presence of CRISPR and R-M systems and blaKPC plasmids in K. pneumoniae, especially when both systems cohabited in one host. The multiple matched recognition sequences of both systems in blaKPC-IncF plasmids (97%) revealed that they were good targets for both systems. Furthermore, the results of conjugation assay demonstrated that CRISPR-Cas and R-M systems in K. pneumoniae could effectively hinder blaKPC plasmid invasion. Notably, CRISPR-Cas and R-M worked together to confer a 4-log reduction in the acquisition of blaKPC plasmid in conjugative events, exhibiting robust synergistic anti-plasmid immunity. CONCLUSIONS: Our results indicate the synergistic role of CRISPR and R-M in regulating horizontal gene transfer in K. pneumoniae and rationalize the development of antimicrobial strategies that capitalize on the immunocompromised status of KPC-KP.


Asunto(s)
Sistemas CRISPR-Cas , Conjugación Genética , Klebsiella pneumoniae , Plásmidos , Klebsiella pneumoniae/genética , Plásmidos/genética , beta-Lactamasas/genética , Enzimas de Restricción-Modificación del ADN/genética , China , Infecciones por Klebsiella/microbiología , Transferencia de Gen Horizontal , Humanos , Genoma Bacteriano/genética
18.
Nat Commun ; 15(1): 5853, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997267

RESUMEN

Transcriptional heterogeneity in isogenic bacterial populations can play various roles in bacterial evolution, but its detection remains technically challenging. Here, we use microbial split-pool ligation transcriptomics to study the relationship between bacterial subpopulation formation and plasmid-host interactions at the single-cell level. We find that single-cell transcript abundances are influenced by bacterial growth state and plasmid carriage. Moreover, plasmid carriage constrains the formation of bacterial subpopulations. Plasmid genes, including those with core functions such as replication and maintenance, exhibit transcriptional heterogeneity associated with cell activity. Notably, we identify a cell subpopulation that does not transcribe conjugal plasmid transfer genes, which may help reduce plasmid burden on a subset of cells. Our study advances the understanding of plasmid-mediated subpopulation dynamics and provides insights into the plasmid-bacteria interplay.


Asunto(s)
Plásmidos , Análisis de la Célula Individual , Plásmidos/genética , Análisis de la Célula Individual/métodos , Escherichia coli/genética , Análisis de Secuencia de ARN/métodos , Conjugación Genética , Bacterias/genética , Regulación Bacteriana de la Expresión Génica , Heterogeneidad Genética
19.
Ann Clin Microbiol Antimicrob ; 23(1): 62, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38978096

RESUMEN

BACKGROUND: This study analyzed the genetic traits and fitness costs of vancomycin-resistant Enterococcus faecium (VREfm) blood isolates carrying Tn1546-type transposons harboring the vanA operon. METHODS: All E. faecium blood isolates were collected from eight general hospitals in South Korea during one-year study period. Antimicrobial susceptibility testing and vanA and vanB PCR were performed. Growth rates of E. faecium isolates were determined. The vanA-positive isolates were subjected to whole genome sequencing and conjugation experiments. RESULTS: Among 308 E. faecium isolates, 132 (42.9%) were positive for vanA. All Tn1546-type transposons harboring the vanA operon located on the plasmids, but on the chromosome in seven isolates. The plasmids harboring the vanA operon were grouped into four types; two types of circular, nonconjugative plasmids (Type A, n = 50; Type B, n = 46), and two types of putative linear, conjugative plasmids (Type C, n = 16; Type D, n = 5). Growth rates of vanA-positive E. faecium isolates were significantly lower than those of vanA-negative isolates (P < 0.001), and reduction in growth rate under vancomycin pressure was significantly larger in isolates harboring putative linear plasmids than in those harboring circular plasmids (P = 0.020). CONCLUSIONS: The possession of vanA operon was costly to bacterial hosts in antimicrobial-free environment, which provide evidence for the importance of reducing vancomycin pressure for prevention of VREfm dissemination. Fitness burden to bacterial hosts was varied by type and size of the vanA operon-harboring plasmid.


Asunto(s)
Antibacterianos , Proteínas Bacterianas , Ligasas de Carbono-Oxígeno , Elementos Transponibles de ADN , Enterococcus faecium , Pruebas de Sensibilidad Microbiana , Operón , Plásmidos , Plásmidos/genética , Enterococcus faecium/genética , Humanos , Proteínas Bacterianas/genética , República de Corea , Ligasas de Carbono-Oxígeno/genética , Antibacterianos/farmacología , Secuenciación Completa del Genoma , Infecciones por Bacterias Grampositivas/microbiología , Enterococos Resistentes a la Vancomicina/genética , Resistencia a la Vancomicina/genética , Aptitud Genética , Vancomicina/farmacología , Conjugación Genética
20.
Structure ; 32(9): 1335-1347.e5, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39002540

RESUMEN

Bacterial conjugation is a process by which DNA is transferred unidirectionally from a donor cell to a recipient cell. It is the main means by which antibiotic resistance genes spread among bacterial populations. It is crucially dependent upon the elaboration of an extracellular appendage, termed "pilus," by a large double-membrane-spanning secretion system termed conjugative "type IV secretion system." Here we present the structure of the conjugative pilus encoded by the R388 plasmid. We demonstrate that, as opposed to all conjugative pili produced so far for cryoelectron microscopy (cryo-EM) structure determination, the conjugative pilus encoded by the R388 plasmid is greatly stimulated by the presence of recipient cells. Comparison of its cryo-EM structure with existing conjugative pilus structures highlights a number of important differences between the R388 pilus structure and that of its homologs, the most prominent being the highly distinctive conformation of its bound lipid.


Asunto(s)
Microscopía por Crioelectrón , Proteínas Fimbrias , Fimbrias Bacterianas , Modelos Moleculares , Plásmidos , Proteínas Fimbrias/química , Proteínas Fimbrias/metabolismo , Proteínas Fimbrias/genética , Plásmidos/metabolismo , Plásmidos/química , Fimbrias Bacterianas/metabolismo , Fimbrias Bacterianas/química , Fimbrias Bacterianas/genética , Fosfolípidos/metabolismo , Fosfolípidos/química , Conjugación Genética , Escherichia coli/metabolismo , Escherichia coli/genética , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA