Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 671
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Bacteriol ; 206(4): e0003124, 2024 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-38534115

RESUMO

A hallmark of Proteus mirabilis infection of the urinary tract is the formation of stones. The ability to induce urinary stone formation requires urease, a nickel metalloenzyme that hydrolyzes urea. This reaction produces ammonia as a byproduct, which can serve as a nitrogen source and weak base that raises the local pH. The resulting alkalinity induces the precipitation of ions to form stones. Transcriptional regulator UreR activates expression of urease genes in a urea-dependent manner. Thus, urease genes are highly expressed in the urinary tract where urea is abundant. Production of mature urease also requires the import of nickel into the cytoplasm and its incorporation into the urease apoenzyme. Urease accessory proteins primarily acquire nickel from one of two nickel transporters and facilitate incorporation of nickel to form mature urease. In this study, we performed a comprehensive RNA-seq to define the P. mirabilis urea-induced transcriptome as well as the UreR regulon. We identified UreR as the first defined regulator of nickel transport in P. mirabilis. We also offer evidence for the direct regulation of the Ynt nickel transporter by UreR. Using bioinformatics, we identified UreR-regulated urease loci in 15 Morganellaceae family species across three genera. Additionally, we located two mobilized UreR-regulated urease loci that also encode the ynt transporter, implying that UreR regulation of nickel transport is a conserved regulatory relationship. Our study demonstrates that UreR specifically regulates genes required to produce mature urease, an essential virulence factor for P. mirabilis uropathogenesis. IMPORTANCE: Catheter-associated urinary tract infections (CAUTIs) account for over 40% of acute nosocomial infections in the USA and generate $340 million in healthcare costs annually. A major causative agent of CAUTIs is Proteus mirabilis, an understudied Gram-negative pathogen noted for its ability to form urinary stones via the activity of urease. Urease mutants cannot induce stones and are attenuated in a murine UTI model, indicating this enzyme is essential to P. mirabilis pathogenesis. Transcriptional regulation of urease genes by UreR is well established; here, we expand the UreR regulon to include regulation of nickel import, a function required to produce mature urease. Furthermore, we reflect on the role of urea catalysis in P. mirabilis metabolism and provide evidence for its importance.


Assuntos
Infecções por Proteus , Infecções Urinárias , Animais , Camundongos , Proteus mirabilis/genética , Urease/metabolismo , Níquel/metabolismo , Proteínas de Bactérias/genética , Escherichia coli/genética , Ureia/metabolismo
2.
BMC Microbiol ; 24(1): 216, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38890647

RESUMO

OBJECTIVE: This study aims to conduct an in-depth genomic analysis of a carbapenem-resistant Proteus mirabilis strain to uncover the distribution and mechanisms of its resistance genes. METHODS: The research primarily utilized whole-genome sequencing to analyze the genome of the Proteus mirabilis strain. Additionally, antibiotic susceptibility tests were conducted to evaluate the strain's sensitivity to various antibiotics, and related case information was collected to analyze the clinical distribution characteristics of the resistant strain. RESULTS: Study on bacterial strain WF3430 from a tetanus and pneumonia patient reveals resistance to multiple antibiotics due to extensive use. Whole-genome sequencing exposes a 4,045,480 bp chromosome carrying 29 antibiotic resistance genes. Two multidrug-resistant (MDR) gene regions, resembling Tn6577 and Tn6589, were identified (MDR Region 1: 64.83 Kb, MDR Region 2: 85.64 Kbp). These regions, consist of integrative and conjugative elements (ICE) structures, highlight the intricate multidrug resistance in clinical settings. CONCLUSION: This study found that a CR-PMI strain exhibits a unique mechanism for acquiring antimicrobial resistance genes, such as blaNDM-1, located on the chromosome instead of plasmids. According to the results, there is increasing complexity in the mechanisms of horizontal transmission of resistance, necessitating a comprehensive understanding and implementation of targeted control measures in both hospital and community settings.


Assuntos
Antibacterianos , Proteínas de Bactérias , Farmacorresistência Bacteriana Múltipla , Testes de Sensibilidade Microbiana , Infecções por Proteus , Proteus mirabilis , Sequenciamento Completo do Genoma , beta-Lactamases , Proteus mirabilis/genética , Proteus mirabilis/efeitos dos fármacos , Proteus mirabilis/enzimologia , Proteus mirabilis/isolamento & purificação , beta-Lactamases/genética , Humanos , Farmacorresistência Bacteriana Múltipla/genética , Antibacterianos/farmacologia , Infecções por Proteus/microbiologia , Proteínas de Bactérias/genética , Cromossomos Bacterianos/genética , Genoma Bacteriano/genética , Carbapenêmicos/farmacologia
3.
Protein Expr Purif ; 221: 106518, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38821452

RESUMO

Chiral amino acids and their deamination products, α-keto acids, have important applications in food, medicine, and fine chemicals. In this study, two l-amino acid deaminase genes from Proteus mirabilis, PM473 of type Ⅰ and PM471 of type Ⅱ were cloned and expressed in Escherichia coli respectively, expected to achieve the chiral separation of amino acids. Extensive substrate preference testing showed that both deaminases had catalytic effects on the d-amino acid component of the D, l-amino acids, and PM473 has a wider catalytic range for amino acids. When D, L-Cys was used as the substrate, all L-Cys components and 75.1 % of D-Cys were converted to mercapto pyruvate, and the remaining D-Cys was a single chiral enantiomer. Molecular docking analysis showed that the interaction between the substrate and the key residues affected the stereoselectivity of enzymes. The compatibility of hydrophobicity between the binding pocket and substrate may be the basic factor that affects the substrate selectivity. This work provides an alternative method for the production of α-keto acids and the resolution of chiral amino acids.


Assuntos
Escherichia coli , Cetoácidos , Simulação de Acoplamento Molecular , Proteus mirabilis , Proteus mirabilis/enzimologia , Proteus mirabilis/genética , Cetoácidos/metabolismo , Cetoácidos/química , Escherichia coli/genética , Escherichia coli/metabolismo , Estereoisomerismo , Especificidade por Substrato , Aminoácidos/genética , Aminoácidos/química , Aminoácidos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/biossíntese , Clonagem Molecular
4.
Vet Res ; 55(1): 50, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38594755

RESUMO

Proteus mirabilis is a commensal bacterium dwelling in the gastrointestinal (GI) tract of humans and animals. Although New Delhi metallo-ß-lactamase 1 (NDM-1) producing P. mirabilis is emerging as a threat, its epidemiology in our society remains largely unknown. LHPm1, the first P. mirabilis isolate harboring NDM-1, was detected from a companion dog that resides with a human owner. The whole-genome study revealed 20 different antimicrobial resistance (AMR) genes against various classes of antimicrobial agents, which corresponded to the MIC results. Genomic regions, including MDR genes, were identified with multiple variations and visualized in a comparative manner. In the whole-genome epidemiological analysis, multiple phylogroups were identified, revealing the genetic relationship of LHPm1 with other P. mirabilis strains carrying various AMR genes. These genetic findings offer comprehensive insights into NDM-1-producing P. mirabilis, underscoring the need for urgent control measures and surveillance programs using a "one health approach".


Assuntos
Doenças do Cão , Infecções por Proteus , Cães , Humanos , Animais , Antibacterianos/farmacologia , Proteus mirabilis/genética , Animais de Estimação/genética , Infecções por Proteus/veterinária , Infecções por Proteus/microbiologia , Farmacorresistência Bacteriana Múltipla/genética , Genômica , República da Coreia , Testes de Sensibilidade Microbiana/veterinária , Plasmídeos , Doenças do Cão/genética
5.
Mol Biol Rep ; 51(1): 446, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38532199

RESUMO

BACKGROUND: Bacterial pathogens are the causative agents of some of the most serious disease problems in cultured fish causing mortalities and severe economic losses. This study was conducted to determine the occurrence and characterization of Proteus mirabilis from infected farmed African catfish in Ogun State, Nigeria. METHODOLOGY: The bacteria were isolated from diseased farmed African catfish (Clarias gariepinus, n=128) with clinical signs of skin haemorrhages, ulceration, and ascites purposively sampled from farms within three senatorial districts namely Ogun East (OE; n=76), Ogun Central (OC; n=30) and Ogun West (OW; n=22) in Ogun State. The isolates were identified based on morphological characteristics, biochemical tests, and 16S rRNA gene characterisation. The 16S rRNA gene sequences were analysed using BLAST, submitted to the NCBI database, and an accession number was generated. RESULTS: The occurrence of Proteus mirabilis in infected Clarias gariepinus was 13.16%, 25%, and 31.25% in OE, OC, and OW, respectively. A significantly higher incidence was recorded in OW compared to other areas. All the Proteus mirabilis isolates were motile, gram-negative, short rod, non-lactose fermenter bacteria that showed positive catalase reactions, negative oxidase, and positive for methyl-red. The Proteus mirabilis isolates (OP 594726.1) were closely related to isolates from Pakistan, Italy, and India CONCLUSIONS: We conclude that Proteus mirabilis colonises farmed Clarias gariepinus in Ogun State, Nigeria and the identified strain showed an evolutionary relationship with known pathogenic NCBI reference strains from other countries.


Assuntos
Peixes-Gato , Proteus mirabilis , Animais , Proteus mirabilis/genética , Peixes-Gato/genética , Nigéria , RNA Ribossômico 16S/genética , Bactérias/genética
6.
J Appl Microbiol ; 135(7)2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38991984

RESUMO

AIMS: We aimed to identify mechanisms underlying the tolerance of Proteus mirabilis-a common cause of catheter associated urinary tract infection-to the clinically used biocides chlorhexidine (CHD) and octenidine (OCT). METHODS AND RESULTS: We adapted three clinical isolates to grow at concentrations of 512 µg ml-1 CHD and 128 µg ml-1 OCT. Genetic characterization and complementation studies revealed mutations inactivating the smvR repressor and increasing smvA efflux expression were associated with adaptation to both biocides. Mutations in mipA (encoding the MltA interacting protein) were less prevalent than smvR mutations and only identified in CHD adapted populations. Mutations in the rppA response regulator were exclusive to one adapted isolate and were linked with reduced polymyxin B susceptibility and a predicted gain of function after biocide adaptation. Biocide adaptation had no impact on crystalline biofilm formation. CONCLUSIONS: SmvR inactivation is a key mechanism in both CHD and OCT tolerance. MipA inactivation alone confers moderate protection against CHD, and rppA showed no direct role in either CHD or OCT susceptibility.


Assuntos
Clorexidina , Iminas , Proteus mirabilis , Piridinas , Proteus mirabilis/efeitos dos fármacos , Proteus mirabilis/genética , Proteus mirabilis/fisiologia , Clorexidina/farmacologia , Iminas/farmacologia , Piridinas/farmacologia , Testes de Sensibilidade Microbiana , Humanos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Infecções por Proteus/microbiologia , Mutação , Farmacorresistência Bacteriana/genética , Anti-Infecciosos Locais/farmacologia , Desinfetantes/farmacologia , Infecções Relacionadas a Cateter/microbiologia , Infecções Urinárias/microbiologia
7.
Ann Clin Microbiol Antimicrob ; 23(1): 46, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38790053

RESUMO

BACKGROUND: Proteus mirabilis is an opportunistic pathogen that has been held responsible for numerous nosocomial and community-acquired infections which are difficult to be controlled because of its diverse antimicrobial resistance mechanisms. METHODS: Antimicrobial susceptibility patterns of P. mirabilis isolates collected from different clinical sources in Mansoura University Hospitals, Egypt was determined. Moreover, the underlying resistance mechanisms and genetic relatedness between isolates were investigated. RESULTS: Antimicrobial susceptibility testing indicated elevated levels of resistance to different classes of antimicrobials among the tested P. mirabilis clinical isolates (n = 66). ERIC-PCR showed great diversity among the tested isolates. Six isolates (9.1%) were XDR while all the remaining isolates were MDR. ESBLs and AmpCs were detected in 57.6% and 21.2% of the isolates, respectively, where blaTEM, blaSHV, blaCTX-M, blaCIT-M and blaAmpC were detected. Carbapenemases and MBLs were detected in 10.6 and 9.1% of the isolates, respectively, where blaOXA-48 and blaNDM-1 genes were detected. Quinolone resistant isolates (75.8%) harbored acc(6')-Ib-cr, qnrD, qnrA, and qnrS genes. Resistance to aminoglycosides, trimethoprim-sulfamethoxazole and chloramphenicol exceeded 80%. Fosfomycin was the most active drug against the tested isolates as only 22.7% were resistant. Class I or II integrons were detected in 86.4% of the isolates. Among class I integron positive isolates, four different gene cassette arrays (dfrA17- aadA5, aadB-aadA2, aadA2-lnuF, and dfrA14-arr-3-blaOXA-10-aadA15) and two gene cassettes (dfrA7 and aadA1) were detected. While class II integron positive isolates carried four different gene cassette arrays (dfrA1-sat1-aadA1, estXVr-sat2-aadA1, lnuF- dfrA1-aadA1, and dfrA1-sat2). CONCLUSION: P. Mirabilis ability to acquire resistance determinants via integrons may be held responsible for the elevated rates of antimicrobial resistance and emergence of XDR or even PDR strains limiting the available therapeutic options for management of infections caused by those strains.


Assuntos
Antibacterianos , Farmacorresistência Bacteriana Múltipla , Testes de Sensibilidade Microbiana , Infecções por Proteus , Proteus mirabilis , Egito/epidemiologia , Humanos , Proteus mirabilis/genética , Proteus mirabilis/efeitos dos fármacos , Proteus mirabilis/isolamento & purificação , Farmacorresistência Bacteriana Múltipla/genética , Infecções por Proteus/microbiologia , Infecções por Proteus/epidemiologia , Antibacterianos/farmacologia , Prevalência , beta-Lactamases/genética , Integrons/genética , Proteínas de Bactérias/genética , Infecção Hospitalar/microbiologia , Infecção Hospitalar/epidemiologia , Masculino
8.
Ann Clin Microbiol Antimicrob ; 23(1): 48, 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38802894

RESUMO

BACKGROUND: Proteus mirabilis is a significant nosocomial pathogen that is frequently associated with a wide range of infections, necessitating heightened attention to mitigate potential health risks. Hence, this study was performed to investigate the impact of sub-minimum inhibitory concentrations (MICs) of ciprofloxacin (CIP) on Proteus mirabilis clinical isolates. METHODS: The sub-MICs of CIP were selected using the growth curve approach. The untreated and treated isolates with sub-MICs of CIP were assessed for their biofilm development, motilities on agar, and other virulence factors. The cell morphology of untreated and treated isolates with sub-MIC of CIP was explored using electron microscope. Moreover, the expression levels of the virulence genes in isolates were measured using quantitative real-time PCR. RESULTS: Data revealed that sub-MICs of CIP significantly (p < 0.05), in a concentration-dependent manner, inhibited biofilm formation and other virulence factors in the selected isolates. Electron microscope analysis showed cell enlargement and various abnormalities in the cell wall and membrane integrity. CONCLUSION: Sub-MICs of CIP exhibited inhibition of virulence and alterations in morphological integrity against P. mirabilis isolates.


Assuntos
Antibacterianos , Biofilmes , Ciprofloxacina , Testes de Sensibilidade Microbiana , Infecções por Proteus , Proteus mirabilis , Fatores de Virulência , Proteus mirabilis/efeitos dos fármacos , Proteus mirabilis/genética , Ciprofloxacina/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Humanos , Antibacterianos/farmacologia , Infecções por Proteus/microbiologia , Fatores de Virulência/genética , Virulência/efeitos dos fármacos
9.
J Infect Chemother ; 30(9): 881-886, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38442770

RESUMO

INTRODUCTION: Carbon dioxide-dependent Proteus mirabilis has been isolated from clinical specimens. It is not clear whether mutations in carbonic anhydrase are responsible for the carbon dioxide dependence of P. mirabilis. The pathogenicity of carbon dioxide-dependent P. mirabilis also remains unclear. The purpose of this study was to determine the cause carbon dioxide dependence of P. mirabilis and its pathogenicity. METHODS: The DNA sequence of can encoding carbonic anhydrase of a carbon dioxide-dependent P. mirabilis small colony variant (SCV) isolate was analyzed. To confirm that impaired carbonic anhydrase activity is responsible for the formation of the carbon dioxide-dependent SCV phenotype of P. mirabilis, we performed complementation experiments using plasmids with intact can. Additionally, mouse infection experiments were performed to confirm the change in virulence due to the mutation of carbonic anhydrase. RESULTS: We found that the can gene of the carbon dioxide-dependent P. mirabilis SCV isolate showed had a frameshift mutation with a deletion of 1 bp (c. 173delC). The can of P. mirabilis encodes carbonic anhydrase was also found to function in Escherichia coli. The cause of the carbon dioxide-dependent SCV phenotype of P. mirabilis was an abnormality in carbonic anhydrase. Nevertheless, no changes were observed in virulence due to the mutation of carbonic anhydrase in mouse infection experiments. CONCLUSIONS: The can gene is essential for the growth of P. mirabilis in ambient air. The mechanisms underlying this fitness advantage in terms of infection warrant further investigation.


Assuntos
Dióxido de Carbono , Anidrases Carbônicas , Infecções por Proteus , Proteus mirabilis , Proteus mirabilis/genética , Proteus mirabilis/patogenicidade , Proteus mirabilis/isolamento & purificação , Dióxido de Carbono/metabolismo , Animais , Camundongos , Infecções por Proteus/microbiologia , Anidrases Carbônicas/genética , Anidrases Carbônicas/metabolismo , Humanos , Virulência/genética , Feminino , Mutação da Fase de Leitura
10.
Acta Vet Hung ; 72(1): 11-20, 2024 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-38578700

RESUMO

Proteus mirabilis is a common enteric bacterium in livestock and humans. The increase and spread of the antimicrobial resistant P. mirabilis is considered alarming worldwide. Transmission mainly occurs through consumption of contaminated poultry products. We investigated antimicrobial resistance (AMR) and virulence markers in broiler chicken-originated P. mirabilis isolates from 380 fecal samples. Phenotypic AMR test was performed against seventeen different antimicrobials. Genotypic AMR test was performed to detect sixteen different AMR genes. The samples were also tested for the presence of eight different virulence genes and biofilm formation. P. mirabilis was isolated in 11% of the samples, with significantly high multidrug-resistant (MDR) prevalence (63%). All isolates were resistant to tetracycline (100%). The combined disc method indicated that all isolates were of extended-spectrum beta-lactamase (ESBL) producers, which was compatible with the high blaTEM prevalence (95%). This was associated with blaTEM being responsible for more than 80% of ampicillin resistance in enteric pathogens. The absence of phenotypically carbapenem-resistant isolates was compatible with the very low prevalences of blaOXA (2%) and blaNDM (0%). All isolates were positive for pmfA, atfA, hpmA, and zapA (100%) virulence genes, while biofilm formation rate (85%) indicated high adherence abilities of the isolates.


Assuntos
Antibacterianos , Proteus mirabilis , Humanos , Animais , Antibacterianos/farmacologia , Virulência , Proteus mirabilis/genética , Galinhas , beta-Lactamases/genética , Farmacorresistência Bacteriana , Testes de Sensibilidade Microbiana/veterinária
11.
BMC Microbiol ; 23(1): 215, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37553593

RESUMO

Proteus mirabilis, a naturally resistant zoonotic bacterium belonging to the Enterobacteriaceae family, has exhibited an alarming increase in drug resistance. Consequently, there is an urgent need to explore alternative antimicrobial agents. Bacteriophages, viruses that selectively target bacteria, are abundant in the natural environment and have demonstrated potential as a promising alternative to antibiotics. In this study, we successfully isolated four strains of Proteus mirabilis phages from sewage obtained from a chicken farm in Sichuan, China. Subsequently, we characterized one of the most potent lytic phages, Q29, by examining its biological and genomic features. Comparative genomic analysis revealed the functional genes and phylogenetic evolution of Q29 phages. Our findings revealed that Proteus mirabilis bacteriophage Q29 possesses an icosahedral symmetrical head with a diameter of 95 nm and a tail length of 240 nm. Moreover, phage Q29 exhibited stability within a temperature range of 37 ℃ to 55 ℃ and under pH conditions ranging from 4 to 9. The optimal multiplicity of infection (MOI) for this phage was determined to be 0.001. Furthermore, the one-step growth curve results indicated an incubation period of approximately 15 min, an outbreak period of approximately 35 min, and an average cleavage quantity of approximately 60 plaque-forming units (PFU) per cell. The genome of phage Q29 was found to have a total length of 58,664 base pairs and encoded 335 open reading frames (ORFs) without carrying any antibiotic resistance genes. Additionally, genetic evolutionary analysis classified phage Q29 within the family Caudalidae and the genus Myotail. This study provides valuable research material for further development of Proteus mirabilis bacteriophage biologics as promising alternatives to antibiotics, particularly in light of the growing challenge of antibiotic resistance posed by this bacterium.


Assuntos
Bacteriófagos , Proteus mirabilis/genética , Filogenia , Genômica , Antibacterianos/farmacologia , Genoma Viral
12.
Biotechnol Bioeng ; 120(1): 239-249, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36123299

RESUMO

Biofilms are often polymicrobial in nature, which can impact their behavior and overall structure, often resulting in an increase in biomass and enhanced antimicrobial resistance. Using plate counts and locked nucleic acid/2'-O-methyl-RNA fluorescence in situ hybridization (LNA/2'OMe-FISH), we studied the interactions of four species commonly associated with catheter-associated urinary tract infections (CAUTI): Enterococcus faecalis, Escherichia coli, Candida albicans, and Proteus mirabilis. Eleven combinations of biofilms were grown on silicone coupons placed in 24-well plates for 24 h, 37°C, in artificial urine medium (AUM). Results showed that P. mirabilis was the dominant species and was able to inhibit both E. coli and C. albicans growth. In the absence of P. mirabilis, an antagonistic relationship between E. coli and C. albicans was observed, with the former being dominant. E. faecalis growth was not affected in any combination, showing a more mutualistic relationship with the other species. Imaging results correlated with the plate count data and provided visual verification of species undetected using the viable plate count. Moreover, the three bacterial species showed overall good repeatability SD (Sr ) values (0.1-0.54) in all combinations tested, whereas C. albicans had higher repeatability Sr values (0.36-1.18). The study showed the complexity of early-stage interactions in polymicrobial biofilms. These interactions could serve as a starting point when considering targets for preventing or treating CAUTI biofilms containing these species.


Assuntos
Cateteres Urinários , Infecções Urinárias , Cateteres Urinários/microbiologia , Escherichia coli/genética , Hibridização in Situ Fluorescente , Proteus mirabilis/genética , Biofilmes , Infecções Urinárias/prevenção & controle , Candida albicans
13.
Eur J Clin Microbiol Infect Dis ; 42(10): 1275-1280, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37688673

RESUMO

To investigate the acquisition and relatedness of New Delhi Metallo-beta-lactamase among multiple separate species from one patient. Five isolates from three species (Pseudomonas aeruginosa; Pa, Acinetobacter baumannii; Ab and Proteus mirabilis; Pm) suspected of harbouring a carbapenemase were investigated by phenotype (antimicrobial susceptibilities) and whole genome sequencing. Epidemiological data was collected on this patient. Three different carbapenemase genes were detected; blaVIM-1 (Pa; ST773), blaOXA-23 (Ab, ST499) and blaNDM-1 identified in all isolates. NDM regions were found chromosomally integrated in all isolates. Data showed no evidence of NDM-1 transfer within this patient suggesting the enzyme was acquired in three separate events.


Assuntos
Acinetobacter baumannii , Humanos , Acinetobacter baumannii/genética , Pacientes , Fenótipo , Proteus mirabilis/genética
14.
Mol Biol Rep ; 50(7): 5817-5826, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37219671

RESUMO

BACKGROUND: Proteus mirabilis is a Gram-negative bacteria most noted for its involvement with catheter-associated urinary tract infections. It is also known for its multicellular migration over solid surfaces, referred to as 'swarming motility'. Here we analyzed the genomic sequences of two P. mirabilis isolates, designated K38 and K39, which exhibit varied swarming ability. METHODS AND RESULTS: The isolates genomes were sequenced using Illumina NextSeq sequencer, resulting in about 3.94 Mbp, with a GC content of 38.6%, genomes. Genomes were subjected for in silico comparative investigation. We revealed that, despite a difference in swarming motility, the isolates showed high genomic relatedness (up to 100% ANI similarity), suggesting that one of the isolates probably originated from the other. CONCLUSIONS: The genomic sequences will allow us to investigate the mechanism driving this intriguing phenotypic heterogeneity between closely related P. mirabilis isolates. Phenotypic heterogeneity is an adaptive strategy of bacterial cells to several environmental pressures. It is also an important factor related to their pathogenesis. Therefore, the availability of these genomic sequences will facilitate studies that focus on the host-pathogen interactions during catheter-associated urinary tract infections.


Assuntos
Infecções por Proteus , Infecções Urinárias , Humanos , Proteus mirabilis/genética , Infecções Urinárias/genética , Infecções Urinárias/microbiologia , Células Clonais , Infecções por Proteus/microbiologia
15.
PLoS Genet ; 16(8): e1008965, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32760058

RESUMO

The mobilizable resistance island Salmonella genomic island 1 (SGI1) is specifically mobilized by IncA and IncC conjugative plasmids. SGI1, its variants and IncC plasmids propagate multidrug resistance in pathogenic enterobacteria such as Salmonella enterica serovars and Proteus mirabilis. SGI1 modifies and uses the conjugation apparatus encoded by the helper IncC plasmid, thus enhancing its own propagation. Remarkably, although SGI1 needs a coresident IncC plasmid to excise from the chromosome and transfer to a new host, these elements have been reported to be incompatible. Here, the stability of SGI1 and its helper IncC plasmid, each expressing a different fluorescent reporter protein, was monitored using fluorescence-activated cell sorting (FACS). Without selective pressure, 95% of the cells segregated into two subpopulations containing either SGI1 or the helper plasmid. Furthermore, FACS analysis revealed a high level of SGI1-specific fluorescence in IncC+ cells, suggesting that SGI1 undergoes active replication in the presence of the helper plasmid. SGI1 replication was confirmed by quantitative PCR assays, and extraction and restriction of its plasmid form. Deletion of genes involved in SGI1 excision from the chromosome allowed a stable coexistence of SGI1 with its helper plasmid without selective pressure. In addition, deletion of S003 (rep) or of a downstream putative iteron-based origin of replication, while allowing SGI1 excision, abolished its replication, alleviated the incompatibility with the helper plasmid and enabled its cotransfer to a new host. Like SGI1 excision functions, rep expression was found to be controlled by AcaCD, the master activator of IncC plasmid transfer. Transient SGI1 replication seems to be a key feature of the life cycle of this family of genomic islands. Sequence database analysis revealed that SGI1 variants encode either a replication initiator protein with a RepA_C domain, or an alternative replication protein with N-terminal replicase and primase C terminal 1 domains.


Assuntos
Proteínas de Bactérias/genética , Conjugação Genética/genética , Ilhas Genômicas/genética , Fosfoproteínas/genética , Plasmídeos/genética , Antibacterianos/farmacologia , Cromossomos/efeitos dos fármacos , Cromossomos/genética , DNA Helicases/genética , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/genética , Plasmídeos/efeitos dos fármacos , Proteus mirabilis/genética , Salmonella enterica/genética , Transativadores/genética
16.
Genomics ; 114(1): 38-44, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34839020

RESUMO

Proteus phage vB_PvuS_Pm34 (Pm34) isolated from the sewage, is a novel virus specific to Proteus vulgaris. Pm34 belonged to the family Siphovirodae with an icosahedron capsid head and a non-contractile tail. Its genome was 39,558 bp in length with a G + C content of 41.4%. Similarity analysis showed that Pm34 shared low identities of 27.6%-38.4% with any other Proteus phages, but had the 96% high identity with Proteus mirabilis AOUC-001. In the genome of Pm34, 70 open reading frames was deduced and 32 had putative functions including integrase and host lysis proteins. No tRNAs, antibiotic resistance and virulence genes were detected. Pm 34 presented a broad pH (4-8) and good temperature tolerance (<40 °C). This is the first report of the bacteriophage specific to P. vulgaris, which can enrich the knowledge of bacteriophages of Prouteus bacteria and provide the possibility for the alternative treatment of P. vulgaris infection.


Assuntos
Bacteriófagos , Siphoviridae , Bacteriófagos/genética , Genoma Viral , Genômica , Fases de Leitura Aberta , Proteus mirabilis/genética , Proteus vulgaris/genética , Siphoviridae/genética
17.
J Bacteriol ; 204(1): e0034721, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34662238

RESUMO

Cells can use self recognition to achieve cooperative behaviors. Self-recognition genes are thought to principally evolve in tandem with partner self-recognition alleles. However, other constraints on protein evolution could exist. Here, we have identified an interaction outside self-recognition loci that could constrain the sequence variation of a self-recognition protein. We show that during collective swarm expansion in Proteus mirabilis, self-recognition signaling co-opts SdaC, a serine transporter. Serine uptake is crucial for bacterial survival and colonization. Single-residue variants of SdaC reveal that self recognition requires an open conformation of the protein; serine transport is dispensable. A distant ortholog from Escherichia coli is sufficient for self recognition; however, a paralogous serine transporter, YhaO, is not. Thus, SdaC couples self recognition and serine transport, likely through a shared molecular interface. Self-recognition proteins may follow the framework of a complex interaction network rather than an isolated two-protein system. Understanding the molecular and ecological constraints on self-recognition proteins lays the groundwork for insights into the evolution of self recognition and emergent collective behaviors. IMPORTANCE Bacteria can receive secret messages from kin during migration. For Proteus mirabilis, these messages are necessary for virulence in multispecies infections. We show that a serine transporter, conserved among gammaproteobacteria, enables self-recognition. Molecular co-option of nutrient uptake could limit the sequence variation of these message proteins. SdaC is the primary transporter for l-serine, a vital metabolite for colonization during disease. Unlike many self-recognition receptors, SdaC is sufficiently conserved between species to achieve recognition. The predicted open conformation is shared by transport and recognition. SdaC reveals the interdependence of communication and nutrient acquisition. As the broader interactions of self-recognition proteins are studied, features shared among microbial self-recognition systems, such as those of Dictyostelium spp. and Neurospora spp., could emerge.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Proteus mirabilis/metabolismo , Proteínas de Bactérias/genética , Transporte Biológico , Locomoção , Proteínas de Membrana/genética , Proteus mirabilis/genética
18.
Antimicrob Agents Chemother ; 66(2): e0198321, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34930033

RESUMO

In this multicentric study performed in 12 French hospitals, we reported that 26.9% (14/52) of the amoxicillin-clavulanate-resistant Proteus mirabilis isolates produced the OXA-23 carbapenemase. We found that an inhibition zone diameter of <11 mm around the amoxicillin-clavulanate disc was an accurate screening cutoff to detect these OXA-23 producers. We confirmed by whole-genome sequencing that these OXA-23-producers all belonged to the same lineage that has been demonstrated to disseminate OXA-23 or OXA-58 in P. mirabilis.


Assuntos
Proteus mirabilis , beta-Lactamases , Combinação Amoxicilina e Clavulanato de Potássio/farmacologia , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Testes de Sensibilidade Microbiana , Prevalência , Proteus mirabilis/genética , beta-Lactamases/genética
19.
J Antimicrob Chemother ; 77(11): 3009-3015, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-35971566

RESUMO

OBJECTIVES: To detect a potential hidden dissemination of the blaOXA-48 gene among Proteus mirabilis isolates obtained from a single centre. METHODS: P. mirabilis from diverse clinical samples presenting an ESBL phenotype or obtained from blood cultured from 2017 to 2019 were evaluated. Bacterial identification was performed using MALDI-TOF MS. MICs were determined using International Organization for Standardization (ISO) standard microdilution and interpreted following EUCAST guidelines. WGS was performed using both short- and long-read technologies and assemblies were done using Unicycler. Resistomes were assessed using the ResFinder database. SNPs were detected using the PATRIC bioinformatics platform. Cloning experiments were performed using the pCRII-TOPO cloning kit. RESULTS: Thirty-one out of 108 (28.7%) isolates were positive for blaOXA-48 and blaCTX-M-15. Twenty-nine out of 31 of the isolates were susceptible to temocillin, piperacillin/tazobactam, ertapenem and meropenem, whereas only 2/31 showed a resistance phenotype against these antibiotics. Both blaOXA-48 and blaCTX-M-15 genes were detected within the same chromosomally integrated new transposon in all isolates. The resistant isolates displayed a single mutation located in the putative promoter upstream of blaOXA-48. Cloning experiments confirmed that the mutation was responsible for the resistance phenotype. CONCLUSIONS: The presence of a chromosomal copy of blaOXA-48 did not confer resistance to carbapenems, but a single mutation in the promoter could lead to an increase in resistance. This study shows a hidden circulation of OXA-48-positive, but carbapenem- and piperacillin/tazobactam-susceptible, P. mirabilis isolates that can become resistant to ß-lactams after a single mutation.


Assuntos
Carbapenêmicos , Proteus mirabilis , Carbapenêmicos/farmacologia , Proteus mirabilis/genética , beta-Lactamases/genética , Testes de Sensibilidade Microbiana , Antibacterianos/farmacologia , Combinação Piperacilina e Tazobactam
20.
PLoS Pathog ; 16(8): e1008707, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32780778

RESUMO

Proteus mirabilis, a Gram-negative uropathogen, is a major causative agent in catheter-associated urinary tract infections (CAUTI). Mannose-resistant Proteus-like fimbriae (MR/P) are crucially important for P. mirabilis infectivity and are required for biofilm formation and auto-aggregation, as well as for bladder and kidney colonization. Here, the X-ray crystal structure of the MR/P tip adhesin, MrpH, is reported. The structure has a fold not previously described and contains a transition metal center with Zn2+ coordinated by three conserved histidine residues and a ligand. Using biofilm assays, chelation, metal complementation, and site-directed mutagenesis of the three histidines, we show that an intact metal binding site occupied by zinc is essential for MR/P fimbria-mediated biofilm formation, and furthermore, that P. mirabilis biofilm formation is reversible in a zinc-dependent manner. Zinc is also required for MR/P-dependent agglutination of erythrocytes, and mutation of the metal binding site renders P. mirabilis unfit in a mouse model of UTI. The studies presented here provide important clues as to the mechanism of MR/P-mediated biofilm formation and serve as a starting point for identifying the physiological MR/P fimbrial receptor.


Assuntos
Adesinas Bacterianas/metabolismo , Biofilmes , Proteínas de Fímbrias/metabolismo , Proteus mirabilis/metabolismo , Infecções Urinárias/microbiologia , Zinco/metabolismo , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Sequência de Aminoácidos , Proteínas de Fímbrias/química , Proteínas de Fímbrias/genética , Humanos , Infecções por Proteus/metabolismo , Infecções por Proteus/microbiologia , Proteus mirabilis/química , Proteus mirabilis/genética , Alinhamento de Sequência , Infecções Urinárias/metabolismo , Zinco/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa