Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Arch Microbiol ; 206(4): 139, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38436732

RESUMEN

Salmonella exhibits a strong inducible acid tolerance response (ATR) under weak acid conditions, and can also induce high-risk strains that are highly toxic, acid resistant, and osmotic pressure resistant to aquatic products. However, the induction mechanism is not yet clear. Therefore, this study aims to simulate the slightly acidic, low-temperature, and high-protein environment during squid processing and storage. Through λRed gene knockout, exploring the effects of low-acid induction, long-term low-temperature storage, and two-component regulation on Salmonella ATR. In this study, we found the two-component system, PhoP/PhoQ and PmrA/PmrB in Salmonella regulates the amino acid metabolism system and improves bacterial acid tolerance by controlling arginine and lysine. Compared with the two indicators of total biogenic amine and diamine content, biogenic amine index and quality index were more suitable for evaluating the quality of aquatic products. The result showed that low-temperature treatment could inhibit Salmonella-induced ATR, which further explained the ATR mechanism from the amino acid metabolism.


Asunto(s)
Arginina , Diaminas , Animales , Decapodiformes , Salmonella/genética , Aminas Biogénicas
2.
Mol Biol Rep ; 51(1): 357, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38400950

RESUMEN

INTRODUCTION: Acinetobacter baumanii (AB) is a bacterium of concern in the hospital setup due to its ability to thrive in unfavorable conditions and the rapid emergence of antibiotic resistance. Carbapenem resistance in this organism is disheartening, further clouded by the emergence of colistin resistance. AIM: The present prospective study aims to note the epidemiology, molecular profile, and clinical outcome of patients with colistin resistance AB infections in a multispecialty tertiary care setup in Odisha, Eastern India. METHODS: All AB strains received from March 2021 to February 2022, identified by Vitek2 (Biomerieux) and confirmed by oxa-51 genes, were included. Carbapenem and colistin resistance were identified as per CLSI guidelines. Known mutations for blaOXA-23-like, blaIMP, blaVIM, blaKP, lpxA, lpxC, pmrA, pmrB, and plasmid mediated mcr (mcr1-5) were screened by conventional PCR techniques. The clinical outcome was noted retrospectively from case sheets. Data was entered in MS Excel and tabulated using SPSS software. RESULTS: In the study period, 350 AB were obtained, of which 317(90.5%) were carbapenem resistant (CRAB). Among the CRAB isolates, 19 (5.9%) were colistin resistant (ABCoR). The most valuable antibiotics in the study were tigecycline (65.4% in ABCoI; 31.6% in ABCoR) and minocycline (44.3% in CI; 36.8% in CR). There was a significant difference in mortality among ABCoI and ABCoR infections. bla OXA was the predominant carbapenem resistance genotype, while pmrA was the predominant colistin resistant genotype. There were no plasmid mediated mcr genes detected in the present study.


Asunto(s)
Acinetobacter , Colistina , Humanos , Colistina/farmacología , Carbapenémicos/farmacología , Estudios Prospectivos , Estudios Retrospectivos , Centros de Atención Terciaria , beta-Lactamasas/genética , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología
3.
Microb Pathog ; 154: 104852, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33762201

RESUMEN

Cellular Perforin-2 (MPEG1) is a pore-forming MACPF family protein that plays a critical role in the defense against bacterial pathogens. Macrophages, neutrophils, and several other cell types that are part of the front line of innate defenses constitutively express high levels of Perforin-2; whereas, most other cell types must be induced to express Perforin-2 by interferons (α, ß and γ) and/or PAMPs such as LPS. In this study, we demonstrate that many bacterial pathogens can limit the expression of Perforin-2 in cells normally inducible for Perforin-2 expression, while ordinarily commensal or non-pathogenic bacteria triggered high levels of Perforin-2 expression in these same cell types. The mechanisms by which pathogens suppress Perforin-2 expression was explored further using Salmonella enterica serovar Typhimurium and cultured MEFs as well as intestinal epithelial cell lines. These studies identified multiple factors required to minimize the expression of Perforin-2 in cell types inducible for Perforin-2 expression. These included the PmrAB and PhoPQ two-component systems, select LPS modification enzymes and the Type III secretion effector protein AvrA.


Asunto(s)
Lipopolisacáridos , Salmonella typhimurium , Proteínas Bacterianas/genética , Células Epiteliales , Fibroblastos , Perforina/genética , Serogrupo
4.
Food Microbiol ; 98: 103785, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33875213

RESUMEN

Cronobacter sakazakii is an emerging opportunistic foodborne pathogen causing rare but severe infections in neonates. Furthermore, the formation of biofilm allows C. sakazakii to persist in different environments. We have demonstrated that the mutator phenotype ascribed to deficiency of the pmrA gene results in more biomass in the first 24 h but less during the post maturation stage (7-14 d) compared with BAA 894. The present study aimed to investigate the regulatory mechanism modulating biofilm formation due to pmrA mutation. The transcriptomic analyses of BAA 894 and s-3 were performed by RNA-sequencing on planktonic and biofilm cells collected at different time points. According to the results, when comparing biofilm to planktonic cells, expression of genes encoding outer membrane proteins, lysozyme, etc. were up-regulated, with LysR family transcriptional regulators, periplasmic proteins, etc. down-regulated. During biofilm formation, cellulose synthase operon genes, flagella-related genes, etc. played essential roles in different stages. Remarkably, pmrA varies the expression of a number of genes related to motility, biofilm formation, and antimicrobial resistance, including srfB, virK, mviM encoding virulence factor, flgF, fliN, etc. encoding flagellar assembly, and marA, ramA, etc. encoding AraC family transcriptional regulators in C. sakazakii. This study provides valuable insights into transcriptional regulation of C. sakazakii pmrA mutant during biofilm formation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas , Cronobacter sakazakii/genética , Plancton/genética , Transcriptoma , Proteínas Bacterianas/genética , Cronobacter sakazakii/crecimiento & desarrollo , Cronobacter sakazakii/fisiología , Regulación Bacteriana de la Expresión Génica , Plancton/crecimiento & desarrollo , Plancton/fisiología , Transcripción Genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
5.
J Bacteriol ; 202(21)2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32839173

RESUMEN

The facultative intracellular bacterial pathogen Francisella tularensis is the causative agent of tularemia in humans and animals. Gram-negative bacteria utilize two-component regulatory systems (TCS) to sense and respond to their changing environment. No classical, tandemly arranged sensor kinase and response regulator TCS genes exist in the human virulent Francisella tularensis subsp. tularensis, but orphaned members are present. PmrA is an orphan response regulator responsible for intramacrophage growth and virulence; however, the regulation of PmrA activity is not understood. We and others have shown that PmrA represses the expression of priM, described to encode an antivirulence determinant. By screening a mutant library for increased priM promoter activity, we identified the sensor kinase homolog QseC as an upstream regulator of priM expression, and this regulation is in part dependent upon the aspartate phosphorylation site of PmrA (D51). Several examined environmental signals, including epinephrine, which is reported to activate QseC in other bacteria, do not affect priM expression in a manner dependent on PmrA. Intramacrophage survival assays also question the finding that PriM is an antivirulence factor. Thus, these data suggest that the PmrA-regulated gene priM is modulated by the QseC-PmrA (QseB) TCS in FrancisellaIMPORTANCE The disease tularemia is caused by the highly infectious Gram-negative pathogen Francisella tularensis This bacterium encodes few regulatory factors (e.g., two-component systems [TCS]). PmrA, required for intramacrophage survival and virulence in the mouse model, is encoded by an orphan TCS response regulator gene. It is unclear how PmrA is responsive to environmental signals to regulate loci, including the PmrA-repressed gene priM We identify an orphan sensor kinase (QseC) that is required for priM repression and further explore both environmental signals that might regulate the QseC-PmrA TCS and the function of PriM.


Asunto(s)
Proteínas Bacterianas/metabolismo , Francisella/enzimología , Histidina Quinasa/metabolismo , Proteínas de la Membrana/metabolismo , Factores de Virulencia/metabolismo , Animales , Línea Celular , Francisella/patogenicidad , Regulación Bacteriana de la Expresión Génica , Macrófagos/microbiología , Ratones , Virulencia
6.
Proteins ; 88(1): 69-81, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31293000

RESUMEN

In class II transcription activation, the transcription factor normally binds to the promoter near the -35 position and contacts the domain 4 of σ factors (σ4 ) to activate transcription. However, σ4 of σ70 appears to be poorly folded on its own. Here, by fusing σ4 with the RNA polymerase ß-flap-tip-helix, we constructed two σ4 chimera proteins, one from σ70σ470c and another from σSσ4Sc of Klebsiella pneumoniae. The two chimera proteins well folded into a monomeric form with strong binding affinities for -35 element DNA. Determining the crystal structure of σ4Sc in complex with -35 element DNA revealed that σ4Sc adopts a similar structure as σ4 in the Escherichia coli RNA polymerase σS holoenzyme and recognizes -35 element DNA specifically by several conserved residues from the helix-turn-helix motif. By using nuclear magnetic resonance (NMR), σ470c was demonstrated to recognize -35 element DNA similar to σ4Sc . Carr-Purcell-Meiboom-Gill relaxation dispersion analyses showed that the N-terminal helix and the ß-flap-tip-helix of σ470c have a concurrent transient α-helical structure and DNA binding reduced the slow dynamics on σ470c . Finally, only σ470c was shown to interact with the response regulator PmrA and its promoter DNA. The chimera proteins are capable of -35 element DNA recognition and can be used for study with transcription factors or other factors that interact with domain 4 of σ factors.


Asunto(s)
Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación Bacteriana de la Expresión Génica , Klebsiella pneumoniae/metabolismo , Factor sigma/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Cristalografía por Rayos X , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/genética , Klebsiella pneumoniae/química , Klebsiella pneumoniae/genética , Modelos Moleculares , Regiones Promotoras Genéticas , Unión Proteica , Mapas de Interacción de Proteínas , Factor sigma/química , Factor sigma/genética , Activación Transcripcional
7.
Infect Immun ; 85(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28373357

RESUMEN

The nitrogen phosphotransferase system (PTSNtr) is a regulatory cascade present in many bacteria, where it controls different functions. This system is usually composed of three basic components: enzyme INtr (EINtr), NPr, and EIIANtr (encoded by the ptsP, ptsO, and ptsN genes, respectively). In Legionella pneumophila, as well as in many other Legionella species, the EIIANtr component is missing. However, we found that deletion mutations in both ptsP and ptsO are partially attenuated for intracellular growth. Furthermore, these two PTSNtr components were found to be required for maximal expression of effector-encoding genes regulated by the transcriptional activator PmrA. Genetic analyses which include the construction of single and double deletion mutants and overexpression of wild-type and mutated forms of EINtr, NPr, and PmrA indicated that the PTSNtr components affect the expression of PmrA-regulated genes via PmrA and independently from PmrB and that EINtr and NPr are part of the same cascade and require their conserved histidine residues in order to function. Furthermore, expression of the Legionella micdadei EIINtr component in L. pneumophila resulted in a reduction in the levels of expression of PmrA-regulated genes which was completely dependent on the L. pneumophila PTS components and the L. micdadei EIINtr conserved histidine residue. Moreover, reconstruction of the L. pneumophila PTS in vitro indicated that EINtr is phosphorylated by phosphoenolpyruvate (PEP) and transfers its phosphate to NPr. Our results demonstrate that the L. pneumophila incomplete PTSNtr is functional and involved in the expression of effector-encoding genes regulated by PmrA.


Asunto(s)
Proteínas Bacterianas/genética , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Humanos , Legionella pneumophila/enzimología , Enfermedad de los Legionarios/microbiología , Macrófagos/microbiología , Fosforilación
8.
Microb Pathog ; 110: 359-364, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28711508

RESUMEN

Cronobacter sakazakii is a well-known opportunistic pathogen responsible for necrotizing enterocolitis, meningitis and septicaemia in the premature, immunocompromised infants and neonates. This pathogen possesses various virulence factors and regulatory systems, and pmrA/pmrB regulatory system has been identified in a variety of bacterial species. The current study aims to investigate role of pmrA gene in the pathogenicity and virulence characteristics of Cronobacter sakazakii using whole genome sequencing and RNA-seq. Results demonstrated that the absence of pmrA has the potential to affect Cronobacter sakazakii on its pathogenicity, virulence and resistance abilities by regulating expression of numerous related genes, including CusB, CusC, CusR and ESA_pESA3p05434.


Asunto(s)
Proteínas Bacterianas/genética , Cronobacter sakazakii/genética , Farmacorresistencia Bacteriana/genética , Genoma Bacteriano/genética , Polimixinas/farmacología , Factores de Virulencia/genética , Antibacterianos/farmacología , Proteínas Bacterianas/fisiología , Composición de Base , Cronobacter sakazakii/patogenicidad , ADN Bacteriano/aislamiento & purificación , Infecciones por Enterobacteriaceae/microbiología , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/aislamiento & purificación , Virulencia , Secuenciación Completa del Genoma
9.
Microb Drug Resist ; 30(8): 325-331, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38905152

RESUMEN

Background: The increased incidence of infections due to multidrug-resistant Gram-negative bacteria has led to the renewed interest in the use of 'forgotten' antibiotics such as colistin. In this work, we studied the chromosomal colistin resistance mechanisms among laboratory-induced colistin-resistant Escherichia coli isolates. Methods: Three colistin-susceptible (ColS) clinical isolates of E. coli assigning to ST131, ST405, and ST361 were exposed to successively increasing concentrations of colistin. The nucleotide sequences of pmrA, pmrB, pmrD, phoP, phoQ, and mgrB genes were determined. The fitness burden associated with colistin resistance acquisition was determined by measuring the in vitro growth rate. Results: Colistin resistance induction resulted in 16-64 times increase in colistin MICs in mutants (n = 8) compared with parental isolates. Analysis of chromosomal genes in colistin-resistant mutants compared with those of ColS ancestors revealed genetic alterations confined to PmrAB two-component system and included PmrA G53R/R81S/L105P and PmrB E121K/E121A/A159P/A159V/G302E changes. The PmrB E121 was found as a critical position for colistin resistance development being altered in three mutants with different ancestors. The acquired colistin-resistance phenotype was stable following 10 consecutive passages in the absence of selective pressure of colistin and it did not alter the susceptibility of mutants to other antimicrobial agents. All mutants exhibited growth rates similar to their respective ColS ancestors, except for one isolate, which revealed a significant growth defect. Conclusion: Our results revealed that colistin resistance in E. coli was more related to PmrAB alterations, which did not impose a fitness cost in most cases.


Asunto(s)
Antibacterianos , Colistina , Farmacorresistencia Bacteriana , Escherichia coli , Pruebas de Sensibilidad Microbiana , Colistina/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Proteínas de Escherichia coli/genética , Mutación , Humanos , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/tratamiento farmacológico , Farmacorresistencia Bacteriana Múltiple/genética , Proteínas Bacterianas , Factores de Transcripción
10.
Front Microbiol ; 15: 1293990, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476937

RESUMEN

Introduction: Acinetobacter baumannii PmrAB is a crucial two-component regulatory system (TCS) that plays a vital role in conferring resistance to polymyxin. PmrA, a response regulator belonging to the OmpR/PhoB family, is composed of a C-terminal DNA-binding effector domain and an N-terminal receiver domain. The receiver domain can be phosphorylated by PmrB, a transmembrane sensor histidine kinase that interacts with PmrA. Once phosphorylated, PmrA undergoes a conformational change, resulting in the formation of a symmetric dimer in the receiver domain. This conformational change facilitates the recognition of promoter DNA by the DNA-binding domain of PmrA, leading to the activation of adaptive responses. Methods: X-ray crystallography was carried out to solve the structure of PmrA receiver domain. Electrophoretic mobility shift assay and Isothermal titration calorimetry were recruited to validate the interaction between the recombinant PmrA protein and target DNA. Field-emission scanning electron microscopy (FE-SEM) was employed to characterize the surface morphology of A. baumannii in both the PmrA knockout and mutation strains. Results: The receiver domain of PmrA follows the canonical α5ß5 response regulator assembly, which undergoes dimerization upon phosphorylation and activation. Beryllium trifluoride is utilized as an aspartate phosphorylation mimic in this process. Mutations involved in phosphorylation and dimerization significantly affected the expression of downstream pmrC and naxD genes. This impact resulted in an enhanced cell surface smoothness with fewer modifications, ultimately contributing to a decrease in colistin (polymyxin E) and polymyxin B resistance. Additionally, a conservative direct-repeat DNA PmrA binding sequence TTTAAGNNNNNTTTAAG was identified at the promoter region of the pmrC and naxD gene. These findings provide structural insights into the PmrA receiver domain and reveal the mechanism of polymyxin resistance, suggesting that PmrA could be a potential drug target to reverse polymyxin resistance in Acinetobacter baumannii.

11.
Microb Genom ; 10(3)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38502064

RESUMEN

Two-component regulatory systems (TCSs) are a major mechanism used by bacteria to sense and respond to their environments. Many of the same TCSs are used by biologically diverse organisms with different regulatory needs, suggesting that the functions of TCS must evolve. To explore this topic, we analysed the amino acid sequence divergence patterns of a large set of broadly conserved TCS across different branches of Enterobacteriaceae, a family of Gram-negative bacteria that includes biomedically important genera such as Salmonella, Escherichia, Klebsiella and others. Our analysis revealed trends in how TCS sequences change across different proteins or functional domains of the TCS, and across different lineages. Based on these trends, we identified individual TCS that exhibit atypical evolutionary patterns. We observed that the relative extent to which the sequence of a given TCS varies across different lineages is generally well conserved, unveiling a hierarchy of TCS sequence conservation with EnvZ/OmpR as the most conserved TCS. We provide evidence that, for the most divergent of the TCS analysed, PmrA/PmrB, different alleles were horizontally acquired by different branches of this family, and that different PmrA/PmrB sequence variants have highly divergent signal-sensing domains. Collectively, this study sheds light on how TCS evolve, and serves as a compendium for how the sequences of the TCS in this family have diverged over the course of evolution.


Asunto(s)
Klebsiella , Alelos , Secuencia de Aminoácidos
12.
Infect Drug Resist ; 16: 6451-6462, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37789836

RESUMEN

Purpose: Colistin resistance mechanisms involving mutations in chromosomal genes associated with LPS modification are not completely understood. Mutations in genes coding for the MgrB regulator frequently account for colistin resistance in Klebsiella pneumoniae, whereas mutations in genes coding for PhoPQ and PmrAB are frequent in E. coli. Our aim was to perform a genetic analysis of chromosomal mutations in colistin-resistant (MIC ≥4 µg/mL) clinical isolates of K. pneumoniae (n = 8) and E. coli (n = 7) of different STs. Methods: Isolates were obtained in a 3-year period in a university hospital in Santiago, Chile. Susceptibility to colistin, aminoglycosides, cephalosporins, carbapenems and ciprofloxacin was determined through broth microdilution. Whole genome sequencing was performed for all isolates and chromosomal gene sequences were compared with sequences of colistin-susceptible isolates of the same sequence types. Results: None of the isolates carried mcr genes. Most of the isolates were susceptible to all the antibiotics analyzed. E. coli isolates were ST69, ST127, ST59, ST131 and ST14, and K. pneumoniae isolates were ST454, ST45, ST6293, ST380 and ST25. All the isolates had mutations in chromosomal genes analyzed. K. pneumoniae had mutations mainly in mgrB gene, whereas E. coli had mutations in pmrA, pmrB and pmrE genes. Most of the amino acid changes in LPS-modifying enzymes of colistin-resistant isolates were found in colistin-susceptible isolates of the same and/or different ST. Eleven of them were found only in colistin-resistant isolates. Conclusion: Colistin resistance mechanisms depend on genetic background, and are due to chromosomal mutations, which implies a lower risk of transmission than plasmid-mediated genes. Colistin resistance is not associated with multidrug-resistance, nor to high-risk sequence types.

13.
J Biochem ; 170(6): 787-800, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34585233

RESUMEN

Acinetobacter baumannii is an insidious emerging nosocomial pathogen that has developed resistance to all available antimicrobials, including the last resort antibiotic, colistin. Colistin resistance often occurs due to mutations in the PmrAB two-component regulatory system. To better understand the regulatory mechanisms contributing to colistin resistance, we have biochemically characterized the A. baumannii PmrA response regulator. Initial DNA-binding analysis shows that A. baumannii PmrA bound to the Klebsiella pneumoniae PmrA box motif. This prompted analysis of the putative A. baumannii PmrAB regulon that indicated that the A. baumannii PmrA consensus box is 5'-HTTAAD N5 HTTAAD. Additionally, we provide the first structural information for the A. baumannii PmrA N-terminal domain through X-ray crystallography and we present a full-length model using molecular modelling. From these studies, we were able to infer the effects of two critical PmrA mutations, PmrA::I13M and PmrA::P102R, both of which confer increased colistin resistance. Based on these data, we suggest structural and dynamic reasons for how these mutations can affect PmrA function and hence encourage resistive traits. Understanding these mechanisms will aid in the development of new targeted antimicrobial therapies. Graphical Abstract.


Asunto(s)
Acinetobacter baumannii/química , Proteínas Bacterianas/química , Colistina , ADN Bacteriano/química , Farmacorresistencia Bacteriana , Mutación , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Dominios Proteicos
14.
Pathogens ; 11(9)2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36145454

RESUMEN

Wild animals may act as efficient antimicrobial-resistance reservoirs and epidemiological links between humans, livestock, and natural environments. By using phenotypic and genotypic characterization, the present study highlighted the occurrence of an antimicrobial-resistant (i.e., amoxicillin, amoxicillin-clavulanic acid, cephalothin, and colistin) Enterobacter hormaechei subsp. steigerwaltii strain in wild boar (Sus scrofa) from France. The molecular analysis conducted showed non-synonymous mutations in the pmrA/pmrB and phoQ/phoP operons and the phoP/Q regulator mgrB gene, leading to colistin resistance. The present data highlight the need for continuous monitoring of multidrug-resistant bacteria in wild animals to limit the spread of these threatening pathogens.

15.
Microb Drug Resist ; 27(5): 628-636, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33085934

RESUMEN

Background: Colistin resistance in Acinetobacter baumannii, the last resort drug for serious infections, is emerging worldwide. There has been paucity of data on this aspect from India, which is one of the largest producers of colistin. We studied colistin resistance in A. baumannii and characterized the isolates with respect to resistance mechanisms and virulence. Methods: A total of 365 A. baumannii isolates were studied. Antimicrobial susceptibility testing was performed as per standards. Colistin resistance mechanisms were studied by mutation detection in pmrA/B and lpxA/C/D genes, phenotypic loss of lipopolysaccharide, presence of mcr1-5 genes, and carbonyl cyanide 3-chlorophenylhydrazone (CCCP) effects. Biofilm formation, desiccation survival, and growth kinetics were studied and statistically analyzed for colistin-resistant and colistin-susceptible isolates. Results: All the colistin-resistant isolates (9, 2.5%) showed multiple mutations at novel sites in pmrA/B and/or lpxA/D genes with reversion of resistance with CCCP. Majority of these isolates (6, 66.6%) were from patients without prior colistin therapy. All received prior carbapenems. The resistant isolates demonstrated no significant difference in biofilm formation and desiccation survival but were slow growers. Conclusion: Mutations in pmrA/B and/or lpxA/D genes were the main resistance mechanism in these colistin-resistant isolates that showed no reduction in fitness.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Hidrazonas/farmacología , beta-Lactamasas/genética , Acinetobacter baumannii/enzimología , Acinetobacter baumannii/genética , Biopelículas , Colistina , Infección Hospitalaria/microbiología , Genes Bacterianos/genética , Humanos , India/epidemiología , Pruebas de Sensibilidad Microbiana , Mutación , Plásmidos , Centros de Atención Terciaria
16.
Pediatr Rep ; 13(4): 650-657, 2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34941638

RESUMEN

BACKGROUND: Nonsyndromic cleft lip with or without palate (NSCL/P) is a multifactorial and common birth malformation caused by genetic and environmental factors, as well as by teratogens. Genome-wide association studies found genetic variations with modulatory effects of NSCL/P formation in Chinese and Iranian populations. We aimed to identify the susceptibility of single-nucleotide polymorphisms (SNPs) to nonsyndromic cleft lip with or without palate in the Indian population. MATERIAL AND METHODS: The present study was conducted on NSCL/P cases and controls. Genomic DNA was extracted from peripheral blood and Axiom- Precision Medicine Research Array (PMRA) was performed. The Axiom-PMRA covers 902,527 markers and several thousand novel risk variants. Quality control-passed samples were included for candidate genetic variation identification, gene functional enrichment, and pathway and network analysis. RESULTS: The genome-wide association study identified fourteen novel candidate gene SNPs that showed the most significant association with the risk of NSCL/P, and eight were predicted to have regulatory sequences. CONCLUSION: The GWAS study showed novel candidate genetic variations in NSCL/P formations. These findings contribute to the understanding of genetic predisposition to nonsyndromic cleft lip with or without palate.

17.
Front Microbiol ; 11: 903, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655500

RESUMEN

Cronobacter sakazakii is an opportunistic Gram-negative pathogen that could cause meningitis and necrotizing enterocolitis. Several Gram-negative bacteria use the PmrA/PmrB system to sense and adapt to environmental change by resistance to cationic antimicrobial peptides of host immune systems. The PmrA/PmrB two-component system regulates several genes to modify LPS structure in the bacterial outer membrane. The role of PmrA/PmrB of C. sakazakii has been studied within the current study. The results suggest that PmrA/PmrB plays a crucial role in modifying LPS structure, cationic antimicrobial peptide susceptibility, cell membrane permeability and hydrophobicity, and invading macrophage.

18.
Int J Antimicrob Agents ; 55(3): 105903, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31954832

RESUMEN

This study characterizes four KPC-2-producing Klebsiella pneumoniae isolates from neonates belonging to a single sequence type 147 (ST147) in relation to carbapenem resistance and explores probable mechanisms of differential colistin resistance among the clonal cluster. Whole genome sequencing (WGS) revealed that the isolates were nearly 100% identical and harbored resistance genes (blaKPC-2,OXA-9,CTX-M-15,SHV-11,OXA-1,TEM-1B, oqxA, oqxB, qnrB1, fosA, arr-2, sul1, aacA4, aac(6')Ib-cr, aac(6')Ib), and several virulence genes. blaKPC-2 was the only carbapenem-resistant gene found, bracketed between ISKpn7 and ISKpn6 of Tn4401b on a non-conjugative IncFII plasmid. Remarkably, one of the clonal isolates was resistant to colistin, the mechanistic basis of which was not apparent from comparative genomics. The transmissible colistin resistance gene, mcr, was absent. Efflux pump inhibitor, carbonyl cyanide 3-chlorophenylhydrazone (CCCP) rendered a 32-fold decrease in the minimum inhibitory concentration (MIC) of colistin in the resistant isolate only. acrB, tolC, ramA, and soxS genes of the AcrAB-TolC pump system overexpressed exclusively in the colistin-resistant isolate, although the corresponding homologs of the AcrAB-TolC pump, regulators and promoters were mutually identical. No change was observed in the expression of other efflux genes (kpnE/F and kpnG/H) or two-component system (TCS) genes (phoP/phoQ, pmrA/pmrB). Colistin resistance in one of the clonal KPC-2-producing isolates is postulated to be due to overexpression of the AcrAB-TolC pump. This study is probably the first to report clinical clonal K. pneumoniae isolates with differences in colistin susceptibility. The presence of carbapenem-resistant isolates with differential behavior in the expression of a genomically identical pump system indicates the nuances of the resistance mechanisms and the difficulty of treatment thereof.


Asunto(s)
Antibacterianos/farmacología , Colistina/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , beta-Lactamasas/biosíntesis , Proteínas Bacterianas/biosíntesis , Proteínas Portadoras/biosíntesis , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Humanos , Recién Nacido , Unidades de Cuidado Intensivo Neonatal , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/metabolismo
19.
mBio ; 10(4)2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31311879

RESUMEN

Nosocomial infections with Acinetobacter baumannii are a global problem in intensive care units with high mortality rates. Increasing resistance to first- and second-line antibiotics has forced the use of colistin as last-resort treatment, and increasing development of colistin resistance in A. baumannii has been reported. We evaluated the transcriptional regulator PmrA as potential drug target to restore colistin efficacy in A. baumannii Deletion of pmrA restored colistin susceptibility in 10 of the 12 extensively drug-resistant A. baumannii clinical isolates studied, indicating the importance of PmrA in the drug resistance phenotype. However, two strains remained highly resistant, indicating that PmrA-mediated overexpression of the phosphoethanolamine (PetN) transferase PmrC is not the exclusive colistin resistance mechanism in A. baumannii A detailed genetic characterization revealed a new colistin resistance mechanism mediated by genetic integration of the insertion element ISAbaI upstream of the PmrC homolog EptA (93% identity), leading to its overexpression. We found that eptA was ubiquitously present in clinical strains belonging to the international clone 2, and ISAbaI integration upstream of eptA was required to mediate the colistin-resistant phenotype. In addition, we found a duplicated ISAbaI-eptA cassette in one isolate, indicating that this colistin resistance determinant may be embedded in a mobile genetic element. Our data disprove PmrA as a drug target for adjuvant therapy but highlight the importance of PetN transferase-mediated colistin resistance in clinical strains. We suggest that direct targeting of the homologous PetN transferases PmrC/EptA may have the potential to overcome colistin resistance in A. baumanniiIMPORTANCE The discovery of antibiotics revolutionized modern medicine and enabled us to cure previously deadly bacterial infections. However, a progressive increase in antibiotic resistance rates is a major and global threat for our health care system. Colistin represents one of our last-resort antibiotics that is still active against most Gram-negative bacterial pathogens, but increasing resistance is reported worldwide, in particular due to the plasmid-encoded protein MCR-1 present in pathogens such as Escherichia coli and Klebsiella pneumoniae Here, we showed that colistin resistance in A. baumannii, a top-priority pathogen causing deadly nosocomial infections, is mediated through different avenues that result in increased activity of homologous phosphoethanolamine (PetN) transferases. Considering that MCR-1 is also a PetN transferase, our findings indicate that PetN transferases might be the Achilles heel of superbugs and that direct targeting of them may have the potential to preserve the activity of polymyxin antibiotics.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Colistina/farmacología , Farmacorresistencia Bacteriana , Infecciones por Acinetobacter/tratamiento farmacológico , Acinetobacter baumannii/genética , Acinetobacter baumannii/aislamiento & purificación , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Orden Génico , Humanos , Mutación
20.
Artículo en Inglés | MEDLINE | ID: mdl-31263682

RESUMEN

Bacteria alter gene expression in response to changes in their environment through various mechanisms that include signal transduction systems. These signal transduction systems use membrane histidine kinase with sensing domains to mediate phosphotransfer to DNA-binding proteins that alter the level of gene expression. Such regulators are called two-component systems (TCSs). TCSs integrate external signals and information from stress pathways, central metabolism and other global regulators, thus playing an important role as part of the overall regulatory network. This review will focus on the knowledge of TCSs in the Gram-negative bacterium, Francisella tularensis, a biothreat agent with a wide range of potential hosts and a significant ability to cause disease. While TCSs have been well-studied in several bacterial pathogens, they have not been well-studied in non-model organisms, such as F. tularensis and its subspecies, whose canonical TCS content surprisingly ranges from few to none. Additionally, of those TCS genes present, many are orphan components, including KdpDE, QseC, QseB/PmrA, and an unnamed two-component system (FTN_1452/FTN_1453). We discuss recent advances in this field related to the role of TCSs in Francisella physiology and pathogenesis and compare the TCS genes present in human virulent versus. environmental species and subspecies of Francisella.


Asunto(s)
Francisella tularensis/genética , Francisella tularensis/metabolismo , Tularemia/microbiología , Animales , Proteínas Bacterianas/genética , Proteínas de Unión al ADN , Francisella tularensis/patogenicidad , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/genética , Histidina Quinasa/metabolismo , Humanos , Dominios Proteicos , Transducción de Señal , Tularemia/transmisión , Virulencia/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda