Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 20(7): e1012376, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39008531

RESUMEN

Antimicrobial resistance is an ongoing "one health" challenge of global concern. The acyl-ACP synthetase (termed AasS) of the zoonotic pathogen Vibrio harveyi recycles exogenous fatty acid (eFA), bypassing the requirement of type II fatty acid synthesis (FAS II), a druggable pathway. A growing body of bacterial AasS-type isoenzymes compromises the clinical efficacy of FAS II-directed antimicrobials, like cerulenin. Very recently, an acyl adenylate mimic, C10-AMS, was proposed as a lead compound against AasS activity. However, the underlying mechanism remains poorly understood. Here we present two high-resolution cryo-EM structures of AasS liganded with C10-AMS inhibitor (2.33 Å) and C10-AMP intermediate (2.19 Å) in addition to its apo form (2.53 Å). Apart from our measurements for C10-AMS' Ki value of around 0.6 µM, structural and functional analyses explained how this inhibitor interacts with AasS enzyme. Unlike an open state of AasS, ready for C10-AMP formation, a closed conformation is trapped by the C10-AMS inhibitor. Tight binding of C10-AMS blocks fatty acyl substrate entry, and therefore inhibits AasS action. Additionally, this intermediate analog C10-AMS appears to be a mixed-type AasS inhibitor. In summary, our results provide proof of principle that inhibiting salvage of eFA by AasS reverses the FAS II bypass. This facilitates the development of next-generation of anti-bacterial therapeutics, esp. the dual therapy consisting of C10-AMS scaffold derivatives combined with certain FAS II inhibitors.

2.
PLoS Pathog ; 19(1): e1011110, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36689471

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen that predominantly causes nosocomial and community-acquired lung infections. As a member of ESKAPE pathogens, carbapenem-resistant P. aeruginosa (CRPA) compromises the limited therapeutic options, raising an urgent demand for the development of lead compounds against previously-unrecognized drug targets. Biotin is an important cofactor, of which the de novo synthesis is an attractive antimicrobial target in certain recalcitrant infections. Here we report genetic and biochemical definition of P. aeruginosa BioH (PA0502) that functions as a gatekeeper enzyme allowing the product pimeloyl-ACP to exit from fatty acid synthesis cycle and to enter the late stage of biotin synthesis pathway. In relative to Escherichia coli, P. aeruginosa physiologically requires 3-fold higher level of cytosolic biotin, which can be attributed to the occurrence of multiple biotinylated enzymes. The BioH protein enables the in vitro reconstitution of biotin synthesis. The repertoire of biotin abundance is assigned to different mouse tissues and/or organ contents, and the plasma biotin level of mouse is around 6-fold higher than that of human. Removal of bioH renders P. aeruginosa biotin auxotrophic and impairs its intra-phagosome persistence. Based on a model of CD-1 mice mimicking the human environment, lung challenge combined with systemic infection suggested that BioH is necessary for the full virulence of P. aeruginosa. As expected, the biotin synthesis inhibitor MAC13772 is capable of dampening the viability of CRPA. Notably, MAC13772 interferes the production of pyocyanin, an important virulence factor of P. aeruginosa. Our data expands our understanding of P. aeruginosa biotin synthesis relevant to bacterial infectivity. In particular, this study represents the first example of an extracellular pathogen P. aeruginosa that exploits biotin cofactor as a fitness determinant, raising the possibility of biotin synthesis as an anti-CRPA target.


Asunto(s)
Biotina , Infecciones por Pseudomonas , Animales , Humanos , Ratones , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Vías Biosintéticas , Biotina/química , Biotina/metabolismo , Pseudomonas aeruginosa/metabolismo
3.
PLoS Pathog ; 18(7): e1010615, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35816546

RESUMEN

Tuberculosis (TB) is one of the leading infectious diseases of global concern, and one quarter of the world's population are TB carriers. Biotin metabolism appears to be an attractive anti-TB drug target. However, the first-stage of mycobacterial biotin synthesis is fragmentarily understood. Here we report that three evolutionarily-distinct BioH isoenzymes (BioH1 to BioH3) are programmed in biotin synthesis of Mycobacterium smegmatis. Expression of an individual bioH isoform is sufficient to allow the growth of an Escherichia coli ΔbioH mutant on the non-permissive condition lacking biotin. The enzymatic activity in vitro combined with biotin bioassay in vivo reveals that BioH2 and BioH3 are capable of removing methyl moiety from pimeloyl-ACP methyl ester to give pimeloyl-ACP, a cognate precursor for biotin synthesis. In particular, we determine the crystal structure of dimeric BioH3 at 2.27Å, featuring a unique lid domain. Apart from its catalytic triad, we also dissect the substrate recognition of BioH3 by pimeloyl-ACP methyl ester. The removal of triple bioH isoforms (ΔbioH1/2/3) renders M. smegmatis biotin auxotrophic. Along with the newly-identified Tam/BioC, the discovery of three unusual BioH isoforms defines an atypical 'BioC-BioH(3)' paradigm for the first-stage of mycobacterial biotin synthesis. This study solves a long-standing puzzle in mycobacterial nutritional immunity, providing an alternative anti-TB drug target.


Asunto(s)
Antituberculosos , Biotina , Biotina/química , Biotina/metabolismo , Escherichia coli/metabolismo , Ésteres/metabolismo , Isoenzimas/metabolismo
4.
Trends Biochem Sci ; 44(11): 973-988, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31279652

RESUMEN

Polymyxins are a group of detergent-like antimicrobial peptides that are the ultimate line of defense against carbapenem-resistant pathogens in clinical settings. Polymyxin resistance primarily originates from structural remodeling of lipid A anchored on bacterial surfaces. We integrate genetic, structural, and biochemical aspects of three major types of lipid A modifiers that have been shown to confer intrinsic colistin resistance. Namely, we highlight ArnT, a glycosyltransferase, EptA, a phosphoethanolamine transferase, and the AlmEFG tripartite system, which is restricted to EI Tor biotype of Vibrio cholerae O1. We also discuss the growing family of mobile colistin resistance (MCR) enzymes, each of which is analogous to EptA, and which pose great challenges to global public health.


Asunto(s)
Antibacterianos/química , Lípido A/metabolismo , Polimixinas/química , Antibacterianos/farmacología , Proteínas Bacterianas/química , Farmacorresistencia Bacteriana , Etanolaminas/metabolismo , Regulación Bacteriana de la Expresión Génica , Glicosiltransferasas/metabolismo , Humanos , Modelos Moleculares , Fosfotransferasas/metabolismo , Polimixinas/farmacología , Unión Proteica , Conformación Proteica
5.
Appl Environ Microbiol ; 89(10): e0095623, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37815340

RESUMEN

Klebsiella pneumoniae is a ubiquitous human pathogen, and its clinical treatment faces two major challenges: multidrug resistance and the pathogenesis of hypervirulent K. pneumoniae. The discovery and study of conditionally essential (CE) genes that can function as potential antimicrobial targets has always been a research concern due to their restriction in the development of novel antibiotics. However, the lack of essential functional genomic data has hampered the study of the mechanisms of essential genes related to antimicrobial susceptibility. In this study, we developed a pooled CE genes mobile clustered regularly interspaced short palindromic repeat (CRISPR) interference screening method (Mobile-CRISPRi-seq) for K. pneumoniae to identify genes that play critical roles in antimicrobial fitness in vitro and host immunity in vivo. Targeting 870 predicted CE genes in K. pneumoniae, Mobile-CRISPRi-seq uncovered the depletion of tetrahydrofolate synthesis pathway genes folB and folP under trimethoprim pressure. Our screening also identified genes waaE and fldA related to polymyxin and ß-lactam susceptibility by applying a screening strategy based on Mobile-CRISPRi-seq and comparative genomics. Furthermore, using a mouse infection model and Mobile-CRISPRi-seq, multiple virulence genes were identified, and among these genes, pal, yciS, and ribB were demonstrated to contribute to the pathogenesis of K. pneumoniae. This study provides a simple, rapid, and effective platform for screening potential antimicrobial targets and virulence genes in K. pneumoniae, and this broadly applicable system can be expanded for high-throughput functional gene study in multiple pathogenic bacteria, especially in gram-negative bacteria. IMPORTANCE The discovery and investigation of conditionally essential (CE) genes that can function as potential antimicrobial targets has always been a research concern because of the restriction of antimicrobial targets in the development of novel antibiotics. In this study, we developed a pooled CE gene-wide mobile clustered regularly interspaced short palindromic repeat (CRISPR) interference sequencing (Mobile-CRISPRi-seq) strategy in Klebsiella pneumoniae to identify genes that play critical roles in the fitness of antimicrobials in vitro and host immunity in vivo. The data suggest a robust tool to screen for loss-of-function phenotypes in a pooled gene knockdown library in K. pneumoniae, and Mobile-CRISPRi-seq may be expanded to multiple bacteria for screening and identification of genes with crucial roles in the fitness of antimicrobials and hosts.


Asunto(s)
Genes Esenciales , Klebsiella pneumoniae , Humanos , Klebsiella pneumoniae/genética , Virulencia/genética , Técnicas de Silenciamiento del Gen , Bacterias/genética , Antibacterianos/farmacología
6.
Environ Sci Technol ; 56(21): 15074-15083, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35608924

RESUMEN

Antibiotics exert selective pressures on clinically relevant antibiotic resistance. It is critical to understand how antibiotic resistance evolves in environmental microbes exposed to subinhibitory concentrations of antibiotics and whether evolutionary dynamics and emergence of resistance are predictable. In this study, Comamonas testosteroni isolated from wastewater activated sludge were subcultured in a medium containing 10 ng/mL cefepime for 40 days (∼300 generations). Stepwise mutations were accumulated, leading to an ultimate 200-fold increase in the minimum inhibitory concentration (MIC) of cefepime. Early stage mutation in DNA polymerase-encoding gene dnaE2 played an important role in antibiotic resistance evolution. Diverse resistance mechanisms were employed and validated experimentally, including increased efflux, biofilm formation, reduced antibiotic uptake, and drug inactivation. The cefepime minimal selective concentrations (MSCs) and relative fitness of susceptible, intermediate, and resistant mutants were determined. Agent-based modeling of the modified Moran process enabled simulations of resistance evolution and predictions of the emergence time and frequency of resistant mutants. The unraveled cefepime resistance mechanisms could be employed by broader bacteria, and the newly developed model is applicable to the predictions of general resistance evolution. The improved knowledge facilitates the assessment, prediction, and mitigation of antibiotic resistance progression in antibiotic-polluted environments.


Asunto(s)
Antibacterianos , Bacterias , Cefepima/farmacología , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Farmacorresistencia Microbiana/genética , Farmacorresistencia Bacteriana/genética
7.
Environ Microbiol ; 23(12): 7445-7464, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33559156

RESUMEN

Tigecycline and colistin are few of 'last-resort' antibiotic defences used in anti-infection therapies against carbapenem-resistant bacterial pathogens. The successive emergence of plasmid-borne tet(X) tigecycline resistance mechanism and mobile colistin resistance (mcr) determinant, renders them clinically useless. Here, we report that co-carriage of tet(X6) and mcr-1 gives co-resistance to both classes of antibiotics by a single plasmid in Escherichia coli. Tet(X6), the new tigecycline resistance enzyme is functionally defined. Both Tet(X6) and MCR-1 robustly interfere accumulation of antibiotic-induced reactive oxygen species (ROS). Unlike that mcr-1 exerts fitness cost in E. coli, tet(X6) does not. In the tet(X6)-positive strain that co-harbors mcr-1, tigecycline resistance is independently of colistin resistance caused by MCR-1-mediated lipid A remodelling, and vice versa. In general consistency with that of MCR-1, Tet(X6) leads to the failure of tigecycline treatment in the infection model of G. mellonella. Taken together, the co-production of Tet(X) and MCR-1 appears as a major clinic/public health concern.


Asunto(s)
Farmacorresistencia Bacteriana , Proteínas de Escherichia coli , Escherichia coli , Antibacterianos/farmacología , Colistina/farmacología , Farmacorresistencia Bacteriana/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Pruebas de Sensibilidad Microbiana , Plásmidos/genética , Tigeciclina/farmacología
8.
Environ Microbiol ; 23(2): 844-860, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32686285

RESUMEN

MCR-4 and MCR-8 are two recently identified members of an ongoing MCR family of colistin resistance. Although that aquatic reservoir for MCR-4 is proposed, the origin and mechanism of MCR-8 is poorly understood. Here we report a previously unrecognized non-mobile colistin resistance enzyme, termed NMCR-2, originating from the plant pathogen Kosakonia pseudosacchari. NMCR-2 (551aa) gives 67.3% identity to MCR-8 (565aa). NMCR-2 is placed as a progenitor/ancestor for MCR-8 in phylogeny of MCR members. Genetic study reveals that nmcr-2 is comparable to mcr-8 in the ability of producing phenotypic colistin resistance. Biochemical analyses determine that these two enzymes catalyse the transfer of PEA from the donor PE lipid substrate to the recipient lipid A molecule by a putative 'ping-pong' trade-off. Further experiment of protein engineering demonstrates that the two motifs (TM region and catalytic domain) of NMCR-2 are functionally exchangeable with that of MCR-8, rather than MCR-1. Physiological impacts of nmcr-2 and/or mcr-8 are detected in Escherichia coli, featuring with fitness cost. Evidently, the action and mechanism of NMCR-2 is analogous to that of MCR-8. Therefore, our finding underlines that NMCR-2 might be a possible progenitor of MCR-8.


Asunto(s)
Antibacterianos/farmacología , Colistina/farmacología , Farmacorresistencia Bacteriana , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Enterobacteriaceae/clasificación , Enterobacteriaceae/efectos de los fármacos , Enterobacteriaceae/genética , Enterobacteriaceae/metabolismo , Escherichia coli/clasificación , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Pruebas de Sensibilidad Microbiana , Familia de Multigenes , Filogenia , Plásmidos/genética , Plásmidos/metabolismo
9.
Environ Microbiol ; 23(12): 7465-7482, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34098588

RESUMEN

The transferability of bacterial resistance to tigecycline, the 'last-resort' antibiotic, is an emerging challenge of global health concern. The plasmid-borne tet(X) that encodes a flavin-dependent monooxygenase represents a new mechanism for tigecycline resistance. Natural source for an ongoing family of Tet(X) resistance determinants is poorly understood. Here, we report the discovery of 26 new variants [tet(X18) to tet(X44)] from the poultry pathogen Riemerella anatipestifer, which expands extensively the current Tet(X) family. R. anatipestifer appears as a natural reservoir for tet(X), of which the chromosome harbours varied copies of tet(X) progenitors. Despite that an inactive ancestor rarely occurs, the action and mechanism of Tet(X2/4)-P, a putative Tet(X) progenitor, was comprehensively characterized, giving an intermediate level of tigecycline resistance. The potential pattern of Tet(X) dissemination from ducks to other animals and humans was raised, in the viewpoint of ecological niches. Therefore, this finding defines a large pool of natural sources for Tet(X) tigecycline resistance, heightening the need of efficient approaches to manage the inter-species transmission of tet(X) resistance determinants.


Asunto(s)
Enfermedades de las Aves de Corral , Riemerella , Animales , Antibacterianos/farmacología , Patos , Pruebas de Sensibilidad Microbiana , Aves de Corral , Riemerella/genética , Tigeciclina/farmacología
10.
Microb Pathog ; 150: 104697, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33347964

RESUMEN

Tigecycline and carbapenem are last-resort antibiotics for serious infections caused by pathogens with multi-drug resistance (MDR). Whereas, bacterial pathogens with co-resistance to tigecycline and carbapenem are poorly addressed. Here we report a tigecycline- and carbapenem-resistant Acinetobacter indicus strain HY20 of duck origin, which co-produces Tet(X5) and NDM-3. Tet(X5) is harbored by a novel plasmid pAI01 (116,992 bp long), which carries 10 antimicrobial resistance genes (AMRs), and heavy metal resistance system cobalt-zinc-cadmium (czc) gene cluster. Unlike that tet(X5) is located in the res-tet(X5)-xerD segment of plasmid, the chromosomal blaNDM-3 is flanked by insertion ISAba125. Collectively, our result represents an example of co-carriage of tet(X5) and blaNDM-3, heightening the importance of AMR surveillance needed in poultry production.


Asunto(s)
Acinetobacter , Patos , Acinetobacter/genética , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Resistencia a Múltiples Medicamentos , Pruebas de Sensibilidad Microbiana , beta-Lactamasas/genética
11.
PLoS Genet ; 14(5): e1007389, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29758020

RESUMEN

Polymyxin is the last line of defense against severe infections caused by carbapenem-resistant gram-negative pathogens. The emergence of transferable MCR-1/2 polymyxin resistance greatly challenges the renewed interest in colistin (polymyxin E) for clinical treatments. Recent studies have suggested that Moraxella species are a putative reservoir for MCR-1/2 genetic determinants. Here, we report the functional definition of ICR-Mo from M. osloensis, a chromosomally encoded determinant of colistin resistance, in close relation to current MCR-1/2 family. ICR-Mo transmembrane protein was prepared and purified to homogeneity. Taken along with an in vitro enzymatic detection, MALDI-TOF mass spectrometry of bacterial lipid A pools determined that the ICR-Mo enzyme might exploit a possible "ping-pong" mechanism to accept the phosphoethanolamine (PEA) moiety from its donor phosphatidylethanolamine (PE) and then transfer it to the 1(or 4')-phosphate position of lipid A via an ICR-Mo-bound PEA adduct. Structural decoration of LPS-lipid A by ICR-Mo renders the recipient strain of E. coli resistant to polymyxin. Domain swapping assays indicate that the two domains of ICR-Mo cannot be functionally-exchanged with its counterparts in MCR-1/2 and EptA, validating its phylogenetic position in a distinct set of MCR-like genes. Structure-guided functional mapping of ICR-Mo reveals a PE lipid substrate recognizing cavity having a role in enzymatic catalysis and the resultant conference of antibiotic resistance. Expression of icr-Mo in E. coli significantly prevents the formation of reactive oxygen species (ROS) induced by colistin. Taken together, our results define a member of a group of intrinsic colistin resistance genes phylogenetically close to the MCR-1/2 family, highlighting the evolution of transferable colistin resistance.


Asunto(s)
Proteínas Bacterianas/genética , Colistina/farmacología , Farmacorresistencia Bacteriana/genética , Proteínas de la Membrana/genética , Moraxella/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/metabolismo , Etanolaminas/metabolismo , Proteínas de la Membrana/clasificación , Proteínas de la Membrana/metabolismo , Simulación del Acoplamiento Molecular , Moraxella/enzimología , Moraxella/genética , Fosfatidiletanolaminas/metabolismo , Filogenia , Unión Proteica , Especificidad por Sustrato
12.
FASEB J ; 33(5): 6055-6068, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30759348

RESUMEN

NAD+ is an enzyme cofactor required for the 3 domains of life. However, little is known about the NAD+ biosynthesis and salvage pathways in the opportunistic pathogen Streptococcus suis. A genome-wide search allows us to identify the NAD+ salvage pathway encoded by an operon of nadR-pnuC-nrtR (from SSU05_1973 to SSU05_1971 on the reverse strand) in the S. suis 05ZYH33 that causes streptococcal toxin shock-like syndrome. The regulator of this pathway is Nudix-related transcriptional regulator (NrtR), a transcription regulator of the Nudix family comprising an N-terminal Nudix-like effector domain, and a C-terminal DNA-binding winged helix-turn-helix-like domain. Intriguingly, the S. suis NrtR naturally contains a single amino acid substitution (K92E) in the catalytic site of its Nudix domain that renders it catalytically inactive but does not influence its ability to bind DNA. Despite its lack of enzymatic activity, DNA-binding activity of NrtR is antagonized by the effector ADP-ribose. Furthermore, nrtR knockout in S. suis serotype 2 reduces its capacity to form biofilms and attenuates its virulence in a mouse infection model. Genome mining indicates that nrtR appears in a strain-specific manner whose occupancy is correlated to bacterial infectivity. Unlike the paradigmatic member of NrtR family having 2 unrelated functions (Nudix hydrolase and DNA binding), S. suis 2 retains a single regulatory role in the modulation of NAD+ salvage. This control of NAD+ homeostasis contributes to S. suis virulence.-Wang, Q., Hassan, B. H., Lou, N., Merritt, J., Feng, Y. Functional definition of NrtR, a remnant regulator of NAD+ homeostasis in the zoonotic pathogen Streptococcus suis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Homeostasis , NAD/metabolismo , Streptococcus suis/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biopelículas , Ratones , Operón , Dominios Proteicos , Pirofosfatasas/genética , Pirofosfatasas/metabolismo , Streptococcus suis/genética , Streptococcus suis/patogenicidad , Factores de Transcripción/química , Factores de Transcripción/genética , Virulencia/genética , Hidrolasas Nudix
13.
J Biol Chem ; 293(12): 4350-4365, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29462787

RESUMEN

Polymyxins such as colistin are antibiotics used as a final line of defense in the management of infections with multidrug-resistant Gram-negative bacteria. Although natural resistance to polymyxins is rare, the discovery of a mobilized colistin resistance gene (mcr-1) in gut bacteria has raised significant concern. As an intramembrane enzyme, MCR-1 catalyzes the transfer of phosphoethanolamine (PEA) to the 1 (or 4')-phosphate group of the lipid A moiety of lipopolysaccharide, thereby conferring colistin resistance. However, the structural and biochemical mechanisms used by this integral membrane enzyme remain poorly understood. Here, we report the modeled structure of the full-length MCR-1 membrane protein. Together with molecular docking, our structural and functional dissection of the complex of MCR-1 with its phosphatidylethanolamine (PE) substrate suggested the presence of a 12 residue-containing cavity for substrate entry, which is critical for both enzymatic activity and its resultant phenotypic resistance to colistin. More importantly, two periplasm-facing helices (PH2 and PH2') of the trans-membrane domain were essential for MCR-1 activity. MALDI-TOF MS and thin-layer chromatography assays provide both in vivo and in vitro evidence that MCR-1 catalyzes the transfer of PEA from the PE donor substrate to its recipient substrate lipid A. Also, the chemical modification of lipid A species was detected in clinical species of bacteria carrying mcr-1 Our results provide mechanistic insights into transferable MCR-1 polymyxin resistance, raising the prospect of rational design of small molecules that reverse bacterial polymyxin resistance, as a last-resort clinical option to combat pathogens with carbapenem resistance.


Asunto(s)
Antibacterianos/farmacología , Colistina/farmacología , Farmacorresistencia Bacteriana , Proteínas de Escherichia coli/metabolismo , Escherichia coli/crecimiento & desarrollo , Tracto Gastrointestinal/microbiología , Polimixinas/farmacología , Cristalografía por Rayos X , Escherichia coli/efectos de los fármacos , Escherichia coli/aislamiento & purificación , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Etanolaminas/química , Etanolaminas/metabolismo , Lípido A/química , Lípido A/metabolismo , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mutación , Filogenia
14.
Mol Microbiol ; 109(5): 642-662, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29995988

RESUMEN

Biotin (vitamin B7), a sulfur-containing fatty acid derivative, is a nutritional virulence factor in certain mycobacterial species. Tight regulation of biotin biosynthesis is important because production of biotin is an energetically expensive process requiring 15-20 equivalents of ATP. The Escherichia coli bifunctional BirA is a prototypical biotin regulatory system. In contrast, mycobacterial BirA is an unusual biotin protein ligase without DNA-binding domain. Recently, we established a novel two-protein paradigm of BioQ-BirA. However, structural and molecular mechanism for BioQ is poorly understood. Here, we report crystal structure of the M. smegmatis BioQ at 1.9 Å resolution. Structure-guided functional mapping defined a seven residues-requiring motif for DNA-binding activity. Western blot and MALDI-TOF MS allowed us to unexpectedly discover that the K47 acetylation activates crosstalking of BioQ to its cognate DNA. More intriguingly, excess of biotin augments the acetylation status of BioQ in M. smegmatis. It seems likely that BioQ acetylation proceeds via a non-enzymatic mechanism. Mutation of this acetylation site K47 in BioQ significantly impairs its regulatory role in vivo. This explains in part (if not all) why BioQ has no detectable requirement of the presumable bio-5'-AMP effecter, which is a well-known ligand for the paradigm E. coli BirA regulator system. Unlike the scenario seen with E. coli carrying a single biotinylated protein, AccB, genome-wide search and Streptavidin blot revealed that no less than seven proteins require the rare post-translational modification, biotinylation in M. smegmatis, validating its physiological demand for biotin at relatively high level. Taken together, our finding defines a novel biotin regulatory machinery by BioQ, posing a possibility that development of new antibiotics targets biotin, the limited nutritional virulence factor in certain pathogenic mycobacterial species.


Asunto(s)
Proteínas Bacterianas/química , Biotina/biosíntesis , Mycobacterium smegmatis/enzimología , Factores de Transcripción/química , Acetilación , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/química , Adenosina Monofosfato/genética , Adenosina Monofosfato/metabolismo , Proteínas Bacterianas/genética , Secuencia de Bases , Biotina/análogos & derivados , Biotina/química , Biotina/genética , Biotina/metabolismo , Biotinilación , Cristalografía por Rayos X , Modelos Moleculares , Mycobacterium smegmatis/genética , Plásmidos , Conformación Proteica , Factores de Transcripción/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-30038043

RESUMEN

We describe the first report of a clinical colistin-resistant ST84 Enterobacter cloacae isolate coharboring mcr-4.3 (previously named mcr-4.2) and blaNDM-1 from a patient in China. The blaNDM-1-harboring IncX3 plasmid and the novel mcr-4.3-harboring ColE plasmid were completely sequenced. Although this isolate showed a high level of resistance to colistin, mcr-4.3 plasmid transformation, gene subcloning, susceptibility testing, and lipid A matrix-assisted laser desorption ionization mass spectrometry analysis indicated that mcr-4.3 itself does not confer resistance to colistin.


Asunto(s)
Enterobacter cloacae/enzimología , beta-Lactamasas/metabolismo , Carbapenémicos/farmacología , China , Colistina/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Enterobacter cloacae/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Plásmidos/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , beta-Lactamasas/genética
16.
PLoS Pathog ; 12(11): e1005957, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27893854

RESUMEN

Polymyxins are the last line of defense against lethal infections caused by multidrug resistant Gram-negative pathogens. Very recently, the use of polymyxins has been greatly challenged by the emergence of the plasmid-borne mobile colistin resistance gene (mcr-1). However, the mechanistic aspects of the MCR-1 colistin resistance are still poorly understood. Here we report the comparative genomics of two new mcr-1-harbouring plasmids isolated from the human gut microbiota, highlighting the diversity in plasmid transfer of the mcr-1 gene. Further genetic dissection delineated that both the trans-membrane region and a substrate-binding motif are required for the MCR-1-mediated colistin resistance. The soluble form of the membrane protein MCR-1 was successfully prepared and verified. Phylogenetic analyses revealed that MCR-1 is highly homologous to its counterpart PEA lipid A transferase in Paenibacili, a known producer of polymyxins. The fact that the plasmid-borne MCR-1 is placed in a subclade neighboring the chromosome-encoded colistin-resistant Neisseria LptA (EptA) potentially implies parallel evolutionary paths for the two genes. In conclusion, our finding provids a first glimpse of mechanism for the MCR-1-mediated colistin resistance.


Asunto(s)
Colistina/farmacología , Farmacorresistencia Bacteriana/genética , Proteínas de Escherichia coli/genética , Secuencia de Bases , Farmacorresistencia Bacteriana/efectos de los fármacos , Resistencia a Múltiples Medicamentos/fisiología , Humanos , Pruebas de Sensibilidad Microbiana , Filogenia , Plásmidos
17.
Cell Microbiol ; 19(7)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28092693

RESUMEN

Pathogenic streptococcal species are responsible for some of the most lethal and prevalent animal and human infections. Previous reports have identified a candidate pathogenicity island (PAI) in two highly virulent clinical isolates of Streptococcus suis type 2, a causative agent of high-mortality streptococcal toxic shock syndrome. This PAI contains a type-IVC secretion system C subgroup (type-IVC secretion system) that is involved in the secretion of unknown pathogenic effectors that are responsible for streptococcal toxic shock syndrome caused by highly virulent strains of S. suis. Both virulence protein B4 and virulence protein D4 were demonstrated to be key components of this type-IVC secretion system. In this study, we identify a new PAI family across 3 streptococcal species; Streptococcus genomic island contains type-IV secretion system, which contains a genomic island type-IVC secretion system and a novel PPIase molecule, SP1. SP1 is shown to interact with a component of innate immunity, peptidoglycan recognition protein (PGLYRP-1) and to perturb the PGLYRP-1-mediated bacteriostatic effect by interacting with protein PGLYRP-1. Our study elucidates a novel mechanism by which bacteria escape by components of the innate immune system by secretion of the SP1 protein in pathogenic Streptococci, which then interacts with PGLYRP-1 from the host. Our results provide potential targets for the development of new antimicrobial drugs against bacteria with resistance to innate host immunity.


Asunto(s)
Citocinas/metabolismo , Islas Genómicas/genética , Isomerasa de Peptidilprolil/genética , Infecciones Estreptocócicas/inmunología , Streptococcus suis/patogenicidad , Sistemas de Secreción Tipo IV/genética , Animales , Línea Celular , Femenino , Células HEK293 , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos BALB C , Isomerasa de Peptidilprolil/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus suis/inmunología
18.
J Antimicrob Chemother ; 72(6): 1723-1730, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28333193

RESUMEN

Objectives: Colistin and carbapenem are two lines of last-resort antibiotics against lethal infections caused by MDR Gram-negative pathogens. The emergence of carbapenemase-positive Escherichia coli with colistin resistance poses a serious threat to public health worldwide. Here we report, for the first time (to the best of our knowledge), a novel combination therapy used for the treatment of E. coli co-producing MCR-1 and NDM-5. Methods: The MICs of colistin were determined alone and with 1-4 mg/L amikacin. A 7-by-4 time-kill array of colistin (0, 0.5, 1, 2, 4, 8 and 16 mg/L) and amikacin (0, 1, 2 and 4 mg/L) over 48 h was designed to characterize the in vitro activity of these agents alone and in combination against each E. coli isolate at an inoculum of 10 6 and 10 8 cfu/mL. The sigmoid E max model was utilized for better delineation of the concentration-effect relationship of each combination. In vivo effectiveness was investigated using a mouse model (combination therapy with intraperitoneal colistin plus amikacin compared with monotherapy). Results: For colistin-resistant isolates, the addition of amikacin demonstrated augmented susceptibility, reducing colistin MICs below the current susceptibility breakpoint. A concentration-dependent decrease in the EC 50 values of colistin was observed for all study isolates in the presence of increasing amikacin concentrations. Further in vivo treatment experiments demonstrated that this combination could achieve 1.5-2.8 log 10 killing after 24 h of therapy, while monotherapy was unable to achieve such a killing effect. Conclusions: The combination of colistin and amikacin may be a promising therapeutic option for the treatment of lethal infections caused by NDM-5-bearing MCR-1-positive superbugs.


Asunto(s)
Amicacina/farmacología , Antibacterianos/farmacología , Colistina/farmacología , Proteínas de Escherichia coli/biosíntesis , Escherichia coli/efectos de los fármacos , beta-Lactamasas/biosíntesis , Amicacina/administración & dosificación , Amicacina/uso terapéutico , Animales , Antibacterianos/administración & dosificación , Antibacterianos/uso terapéutico , Colistina/administración & dosificación , Colistina/uso terapéutico , Farmacorresistencia Bacteriana Múltiple , Sinergismo Farmacológico , Quimioterapia Combinada , Escherichia coli/enzimología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Ratones , Pruebas de Sensibilidad Microbiana
19.
BMC Microbiol ; 17(1): 67, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28292261

RESUMEN

BACKGROUND: It was previously reported in China that two recent large-scale outbreaks of Streptococcus suis serotype 2 (S. suis 2) infections in human were caused by two highly virulent S. suis 2 strains, from which a novel genomic island (GEI), associated with disease onset and progression and designated 89 K, was identified. Here, an avirulent strain, 05HAS68, was isolated from a clinically healthy pig. RESULTS: By comparing the genomes of this avirulent strain with virulent strains, it was found that massive genomic rearrangements occurred, resulting in alterations in gene expression that caused enormous single gene gain and loss. Important virulent genes were lost, such as extracellular protein factor (ef) and suilysin (sly) and larger mutants, such as muramidase-released protein (mrp). Piglets vaccinated with the avirulent strain, 05HAS68, had increased TNF-α and IFN-γ levels in the peripheral blood and were fully protected from challenge infection with the most virulent S. suis 2 strain, 05ZYH33. Transfusion of T cells and plasma from vaccinated pigs resulted in protection of recipient animals against the 05ZYH33 challenge. CONCLUSION: These results suggest that analysis genome of the avirulent strains are instrumental in the development of vaccines and for the functional characterization of important of genetic determinants.


Asunto(s)
Genoma Bacteriano/genética , Serogrupo , Infecciones Estreptocócicas/microbiología , Streptococcus suis/genética , Streptococcus suis/inmunología , Streptococcus suis/patogenicidad , Virulencia/genética , Pruebas de Aglutinación , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , China , ADN Bacteriano , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Proteínas Hemolisinas/genética , Interferón gamma/sangre , Masculino , Microscopía Electrónica de Transmisión , Proteoma/análisis , Análisis de Secuencia de ADN , Serotipificación , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas , Streptococcus suis/aislamiento & purificación , Sus scrofa/microbiología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Linfocitos T , Factor de Necrosis Tumoral alfa/sangre
20.
Antimicrob Agents Chemother ; 60(11): 6899-6902, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27550364

RESUMEN

Two Escherichia coli clones (sequence type 648 [ST648] and ST156) that coproduce NDM-5 and MCR-1 were detected from a single fowl in China. The blaNDM-5 gene was found on the two indistinguishable IncX3 plasmids from the two different E. coli isolates, whereas the mcr-1 gene was located on IncHI2 and IncI2 plasmids, respectively, suggesting that blaNDM-5 and mcr-1 have spread in avian intestinal flora. Also, the two strains harbor blaTEM-1, blaCTX-M-55, fosA3, and aac(6')-Ib The multiresistant E. coli strains (especially the epidemic clone ST648) might raise a potential threat to human health via food chain transmission.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Proteínas de Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , beta-Lactamasas/genética , Animales , China , Farmacorresistencia Bacteriana Múltiple/genética , Patos/microbiología , Escherichia coli/aislamiento & purificación , Pruebas de Sensibilidad Microbiana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA