Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 718
Filtrer
1.
Acta Microbiol Immunol Hung ; 71(2): 134-139, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38837239

RÉSUMÉ

The rate of pandrug-resistant Acinetobacter baumannii strains is on the rise in all continents. This bacterium can acquire resistance to all antibiotics, even to colistin. Alterations in the lipid A or/and the two-component pmrAB were earlier detected in colistin resistance. We investigated and analyzed two strains of A. baumannii (ABRC1 and ABRC2) isolated from two patients admitted to intensive care unit with a septic shock. Both strains were resistant to all tested antibiotics including colistin with a MIC >256 mg L-1. Colistin resistance genes (pmrA, pmrB, lpxA, lpxC, lpxD, and lpsB) of two strains (ABRC1 and ABRC2) were investigated by PCR and sequencing. Obtained nucleic acid sequences were aligned with reference sequences of ATCC 19606 and 17987. In this study two amino acid mutations, N287D in the lpxC gene and E117K in the lpxD gene, were detected in both ABRC1 and ABRC2 strains. ABRC1 had an additional H200L mutation in the pmrA gene. Both colistin resistant strains harbored the same A138T mutation in the pmrB gene. The ABRC2 strain also had an alteration in the kinase domain, specifically an R263S substitution of the histidine kinase domain. Three identical mutations were found in the lpsB gene of both A. baumannii strains: Q216K + H218G + S219E. As a result, a newly deduced protein sequence in both ABRC1 and ABRC2 strains differed from those described in ATCC 17978 and 19606 strains was determined. Colistin resistance is multifactorial in A. baumannii. In our study we detected novel mutations in colistin resistant A. baumannii clinical isolates.


Sujet(s)
Infections à Acinetobacter , Acinetobacter baumannii , Antibactériens , Protéines bactériennes , Lipide A , Tests de sensibilité microbienne , Acinetobacter baumannii/génétique , Acinetobacter baumannii/effets des médicaments et des substances chimiques , Acinetobacter baumannii/métabolisme , Humains , Lipide A/génétique , Lipide A/métabolisme , Lipide A/biosynthèse , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Antibactériens/pharmacologie , Infections à Acinetobacter/microbiologie , Résistance bactérienne aux médicaments/génétique , Polymyxines/pharmacologie , Colistine/pharmacologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Mutation
2.
Genes (Basel) ; 15(6)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38927720

RÉSUMÉ

Many enzymes in the Raetz pathway for lipid A biosynthesis in Escherichia coli are essential. A homologous protein Pa1792|LpxH in Pseudomonas aeruginosa is known to complement the loss of LpxH in E. coli. Genome-wide transposon-insertion sequencing analysis indicates that lpxH is essential in P. aeruginosa. However, genetic analysis of lpxH in P. aeruginosa has not been carried out, partly because the conditional alleles of essential genes are not readily constructed. In this study, we first constructed a plasmid-based temperature-sensitive mutant ΔlpxH/pTS-lpxH or lpxH(Ts) in P. aeruginosa PAO1. Spot-plating assay indicated that lpxH(Ts) was lethal at a restrictive temperature, confirming its essentiality for growth. Microscopic analysis revealed that lpxH(Ts) exhibited an oval-shaped morphology, suggesting that lpxH was required for rod-shape formation. SDS-PAGE and Western blotting analysis showed that lpxH(Ts) failed to synthesize lipid A, consistent with its function in lipid A biosynthesis. Strong expression of lpxH but not the non-homologous isoenzyme lpxI or lpxG impeded growth and caused cell lysis, implying that lpxH-specific cofactors were required for this toxic effect in P. aeruginosa. Together, our results demonstrate that lpxH is essential for lipid A biosynthesis, rod-shaped growth, and viability in P. aeruginosa. We propose that this plasmid-based conditional allele is a useful tool for the genetic study of essential genes in P. aeruginosa.


Sujet(s)
Protéines bactériennes , Plasmides , Pseudomonas aeruginosa , Pseudomonas aeruginosa/génétique , Plasmides/génétique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Température , Mutation , Lipide A/génétique , Lipide A/métabolisme , Escherichia coli/génétique , Escherichia coli/métabolisme
3.
Nat Commun ; 15(1): 4733, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830951

RÉSUMÉ

Polymyxins are gram-negative antibiotics that target lipid A, the conserved membrane anchor of lipopolysaccharide in the outer membrane. Despite their clinical importance, the molecular mechanisms underpinning polymyxin activity remain unresolved. Here, we use surface plasmon resonance to kinetically interrogate interactions between polymyxins and lipid A and derive a phenomenological model. Our analyses suggest a lipid A-catalyzed, three-state mechanism for polymyxins: transient binding, membrane insertion, and super-stoichiometric cluster accumulation with a long residence time. Accumulation also occurs for brevicidine, another lipid A-targeting antibacterial molecule. Lipid A modifications that impart polymyxin resistance and a non-bactericidal polymyxin derivative exhibit binding that does not evolve into long-lived species. We propose that transient binding to lipid A permeabilizes the outer membrane and cluster accumulation enables the bactericidal activity of polymyxins. These findings could establish a blueprint for discovery of lipid A-targeting antibiotics and provide a generalizable approach to study interactions with the gram-negative outer membrane.


Sujet(s)
Antibactériens , Lipide A , Polymyxine B , Résonance plasmonique de surface , Polymyxine B/pharmacologie , Polymyxine B/métabolisme , Lipide A/métabolisme , Lipide A/composition chimique , Antibactériens/pharmacologie , Antibactériens/composition chimique , Antibactériens/métabolisme , Tests de sensibilité microbienne , Membrane bactérienne externe/métabolisme , Membrane bactérienne externe/effets des médicaments et des substances chimiques , Cinétique
4.
Mol Biol Rep ; 51(1): 675, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38787484

RÉSUMÉ

BACKGROUND: Bioscaffolds and cells are two main components in the regeneration of damaged tissues via cell therapy. Umbilical cord stem cells are among the most well-known cell types for this purpose. The main objective of the present study was to evaluate the effect of the pretreatment of the foreskin acellular matrix (FAM) by monophosphoryl lipid A (MPLA) and Lactobacillus casei supernatant (LCS) on the attraction of human umbilical cord mesenchymal stem cells (hucMSC). METHODS AND RESULTS: The expression of certain cell migration genes was studied using qRT-PCR. In addition to cell migration, transdifferentiation of these cells to the epidermal-like cells was evaluated via immunohistochemistry (IHC) and immunocytochemistry (ICC) of cytokeratin 19 (CK19). The hucMSC showed more tissue tropism in the presence of MPLA and LCS pretreated FAM compared to the untreated control group. We confirmed this result by scanning electron microscopy (SEM) analysis, glycosaminoglycan (GAG), collagen, and DNA content. Furthermore, IHC and ICC data demonstrated that both treatments increase the protein expression level of CK19. CONCLUSION: Pretreatment of acellular bioscaffolds by MPLA or LCS can increase the migration rate of cells and also transdifferentiation of hucMSC to epidermal-like cells without growth factors. This strategy suggests a new approach in regenerative medicine.


Sujet(s)
Lacticaseibacillus casei , Lipide A , Cellules souches mésenchymateuses , Humains , Cellules souches mésenchymateuses/métabolisme , Lacticaseibacillus casei/métabolisme , Lipide A/métabolisme , Lipide A/analogues et dérivés , Mouvement cellulaire/effets des médicaments et des substances chimiques , Peau/métabolisme , Structures d'échafaudage tissulaires/composition chimique , Mâle , Cordon ombilical/cytologie , Cordon ombilical/métabolisme , Prépuce/cytologie , Transdifférenciation cellulaire/effets des médicaments et des substances chimiques , Ingénierie tissulaire/méthodes , Matrice extracellulaire/métabolisme , Kératine-19/métabolisme , Kératine-19/génétique
5.
Microbiol Res ; 283: 127712, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38593580

RÉSUMÉ

Lipid A plays a crucial role in Vibrio parahaemolyticus. Previously we have reported the diversity of secondary acylation of lipid A in V. parahaemolyticus and four V. parahaemolyticus genes VP_RS08405, VP_RS01045, VP_RS12170, and VP_RS00880 exhibiting homology to the secondary acyltransferases in Escherichia coli. In this study, the gene VP_RS12170 was identified as a specific lipid A secondary hydroxy-acyltransferase responsible for transferring a 3-hydroxymyristate to the 2'-position of lipid A. Four E. coli mutant strains WHL00, WHM00, WH300, and WH001 were constructed, and they would synthesize lipid A with different structures due to the absence of genes encoding lipid A secondary acyltransferases or Kdo transferase. Then V. parahaemolyticus VP_RS12170 was overexpressed in W3110, WHL00, WHM00, WH300, and WH001, and lipid A was isolated from these strains and analyzed by using thin-layer chromatography and high-performance liquid chromatography-tandem mass spectrometry. The detailed structural changes of lipid A in these mutant strains with and without VP_RS12170 overexpression were compared and conclude that VP_RS12170 can specifically transfer a 3-hydroxymyristate to the 2'-position of lipid A. This study also demonstrated that the function of VP_RS12170 is Kdo-dependent and its favorite substrate is Kdo-lipid IVA. These findings give us better understanding the biosynthetic pathway and the structural diversity of V. parahaemolyticus lipid A.


Sujet(s)
Lipide A , Vibrio parahaemolyticus , Lipide A/composition chimique , Lipide A/métabolisme , Escherichia coli/génétique , Escherichia coli/métabolisme , Vibrio parahaemolyticus/génétique , Vibrio parahaemolyticus/métabolisme , Acyltransferases/génétique , Acyltransferases/métabolisme , Spectrométrie de masse
6.
Int J Biol Macromol ; 267(Pt 2): 131327, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38574903

RÉSUMÉ

The emergence of multidrug resistance has provided a great challenge to treat nosocomial infections, which have become a major health threat around the globe. Lipid A (an active endotoxin component), the final product of the Raetz lipid A metabolism pathway, is a membrane anchor of lipopolysaccharide (LPS) of the gram-negative bacterial outer membrane. It shields bacterial cells and serves as a protective barrier from antibiotics, thereby eliciting host response and making it difficult to destroy. UDP-2,3-diacylglucosamine pyrophosphate hydrolase (LpxH), a crucial peripheral membrane enzyme of the Raetz pathway, turned out to be the potential target to inhibit the production of Lipid A. This review provides a comprehensive compilation of information regarding the structural and functional aspects of LpxH, as well as its analogous LpxI and LpxG. In addition, apart from by providing a broader understanding of the enzyme-inhibitor mechanism, this review facilitates the development of novel drug candidates that can inhibit the pathogenicity of the lethal bacterium.


Sujet(s)
Bactéries à Gram négatif , Bactéries à Gram négatif/enzymologie , Bactéries à Gram négatif/effets des médicaments et des substances chimiques , Pyrophosphatases/métabolisme , Pyrophosphatases/composition chimique , Lipide A/composition chimique , Lipide A/métabolisme , Antienzymes/pharmacologie , Antienzymes/composition chimique , Humains
7.
PLoS Biol ; 22(3): e3002558, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38478588

RÉSUMÉ

Polyphosphates (polyP) are chains of inorganic phosphates that can reach over 1,000 residues in length. In Escherichia coli, polyP is produced by the polyP kinase (PPK) and is thought to play a protective role during the response to cellular stress. However, the molecular pathways impacted by PPK activity and polyP accumulation remain poorly characterized. In this work, we used label-free mass spectrometry to study the response of bacteria that cannot produce polyP (Δppk) during starvation to identify novel pathways regulated by PPK. In response to starvation, we found 92 proteins significantly differentially expressed between wild-type and Δppk mutant cells. Wild-type cells were enriched for proteins related to amino acid biosynthesis and transport, while Δppk mutants were enriched for proteins related to translation and ribosome biogenesis, suggesting that without PPK, cells remain inappropriately primed for growth even in the absence of the required building blocks. From our data set, we were particularly interested in Arn and EptA proteins, which were down-regulated in Δppk mutants compared to wild-type controls, because they play a role in lipid A modifications linked to polymyxin resistance. Using western blotting, we confirm differential expression of these and related proteins in K-12 strains and a uropathogenic isolate, and provide evidence that this mis-regulation in Δppk cells stems from a failure to induce the BasRS two-component system during starvation. We also show that Δppk mutants unable to up-regulate Arn and EptA expression lack the respective L-Ara4N and pEtN modifications on lipid A. In line with this observation, loss of ppk restores polymyxin sensitivity in resistant strains carrying a constitutively active basR allele. Overall, we show a new role for PPK in lipid A modification during starvation and provide a rationale for targeting PPK to sensitize bacteria towards polymyxin treatment. We further anticipate that our proteomics work will provide an important resource for researchers interested in the diverse pathways impacted by PPK.


Sujet(s)
Escherichia coli , Lipopolysaccharides , Phosphotransferases (Phosphate Group Acceptor) , Escherichia coli/métabolisme , Lipopolysaccharides/métabolisme , Lipide A/métabolisme , Polyphosphates/métabolisme
8.
J Glob Antimicrob Resist ; 37: 108-121, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38552872

RÉSUMÉ

OBJECTIVES: A concern with the ESKAPE pathogen, Enterobacter bugandensis, and other species of the Enterobacter cloacae complex, is the frequent appearance of multidrug resistance against last-resort antibiotics, such as polymyxins. METHODS: Here, we investigated the responses to polymyxin B (PMB) in two PMB-resistant E. bugandensis clinical isolates by global transcriptomics and deletion mutagenesis. RESULTS: In both isolates, the genes of the CrrAB-regulated operon, including crrC and kexD, displayed the highest levels of upregulation in response to PMB. ∆crrC and ∆kexD mutants became highly susceptible to PMB and lost the heteroresistant phenotype. Conversely, heterologous expression of CrrC and KexD proteins increased PMB resistance in a sensitive Enterobacter ludwigii clinical isolate and in the Escherichia coli K12 strain, W3110. The efflux pump, AcrABTolC, and the two component regulators, PhoPQ and CrrAB, also contributed to PMB resistance and heteroresistance. Additionally, the lipid A modification with 4-L-aminoarabinose (L-Ara4N), mediated by the arnBCADTEF operon, was critical to determine PMB resistance. Biochemical experiments, supported by mass spectrometry and structural modelling, indicated that CrrC is an inner membrane protein that interacts with the membrane domain of the KexD pump. Similar interactions were modeled for AcrB and AcrD efflux pumps. CONCLUSION: Our results support a model where drug efflux potentiated by CrrC interaction with membrane domains of major efflux pumps combined with resistance to PMB entry by the L-Ara4N lipid A modification, under the control of PhoPQ and CrrAB, confers the bacterium high-level resistance and heteroresistance to PMB.


Sujet(s)
Antibactériens , Protéines bactériennes , Enterobacter , Lipide A , Tests de sensibilité microbienne , Polymyxine B , Polymyxine B/pharmacologie , Enterobacter/génétique , Enterobacter/effets des médicaments et des substances chimiques , Enterobacter/métabolisme , Antibactériens/pharmacologie , Lipide A/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Arabinose/métabolisme , Arabinose/pharmacologie , Arabinose/analogues et dérivés , Humains , Régulation de l'expression des gènes bactériens , Opéron , Multirésistance bactérienne aux médicaments/génétique , Infections à Enterobacteriaceae/microbiologie , Résistance bactérienne aux médicaments , Protéines de transport membranaire/génétique , Protéines de transport membranaire/métabolisme
9.
mSphere ; 9(3): e0063323, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38421175

RÉSUMÉ

Kate Hummels works in the field of bacterial cell envelope biosynthesis and studies the regulation of the metabolic pathways needed to build the Gram-negative cell envelope. In this mSphere of Influence article, she reflects on how the papers "A penicillin-binding protein inhibits selection of colistin-resistant, lipopoligosaccharide-deficient Acinetobacter baumannii" by Boll et al. and "Caulobacter lipid A is conditionally dispensable in the absence of fur and in the presence of anionic sphingolipids" by Zik et al. made an impact on her by studying organisms that deviate from accepted norms to highlight the plethora of unanswered questions in cell envelope biology.


Sujet(s)
Lipide A , Lipopolysaccharides , Lipide A/métabolisme , Lipopolysaccharides/métabolisme , Colistine/pharmacologie , Protéines de liaison aux pénicillines , Membrane cellulaire/métabolisme
10.
J Lipid Res ; 65(3): 100509, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38295984

RÉSUMÉ

Alcohol binge drinking allows the translocation of bacterial lipopolysaccharide (LPS) from the gut to the blood, which activates the peripheral immune system with consequences in neuroinflammation. A possible access/direct signaling of LPS to/in the brain has not yet been described under alcohol abuse conditions. Apolipoproteins are compounds altered by alcohol with high affinity to LPS which may be involved in its transport to the brain or in its elimination. Here, we explored the expression of small components of LPS, in its free form or bound to apolipoproteins, in the brain of female and male rats exposed to alcohol binges. Animals received ethanol oral gavages (3 g/kg every 8 h) for 4 days. LPS or its components (Lipid A and core), LPS-binding protein, corticosterone, lipoproteins (HDL, LDL), apolipoproteins (ApoAI, ApoB, and ApoE), and their receptors were measured in plasma and/or in nonperfused prefrontal cortex (PFC) and cerebellum. Brain LipidA-apolipoprotein aggregates were determined by Western blotting and confirmed by co-immunoprecipitation. In animals exposed to alcohol binges: 1) plasma LPS-binding protein was elevated in both sexes; 2) females showed elevations in plasma ApoAI and corticosterone levels; 3) Lipid A formed aggregates with ApoAI in the female PFC and with ApoB in males, the latter showing Toll-like receptor 4 upregulation in PFC but not females. These results suggest that small bacterial components are present within the brain, forming aggregates with different apolipoproteins, depending on the sex, after alcohol binge intoxications. Results may have implications for the crosstalk between alcohol, LPS, and neuroinflammation.


Sujet(s)
Éthanol , Lipopolysaccharides , Rats , Mâle , Femelle , Animaux , Lipopolysaccharides/pharmacologie , Lipopolysaccharides/métabolisme , Maladies neuro-inflammatoires , Lipide A/métabolisme , Corticostérone/métabolisme , Apolipoprotéines/métabolisme , Apolipoprotéines E/métabolisme , Encéphale/métabolisme , Apolipoprotéines B/métabolisme
11.
mBio ; 15(2): e0282323, 2024 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-38131669

RÉSUMÉ

Pseudomonas aeruginosa can survive in a myriad of environments, partially due to modifications of its lipid A, the membrane anchor of lipopolysaccharide. We previously demonstrated that divergent late acyltransferase paralogs, HtrB1 and HtrB2, add acyloxyacyl laurate to lipid A 2- and 2'-acyl chains, respectively. The genome of P. aeruginosa also has genes which encode two dioxygenase enzymes, LpxO1 and LpxO2, that individually hydroxylate a specific secondary laurate. LpxO1 acts on the 2'-acyloxyacyl laurate (added by HtrB2), whereas LpxO2 acts on the 2-acyloxyacyl laurate (added by HtrB1) in a site-specific manner. Furthermore, while both enzyme pairs are evolutionarily linked, phylogenomic analysis suggests the LpxO1/HtrB2 enzyme pair as being of ancestral origin, present throughout the Pseudomonas lineage, whereas the LpxO2/HtrB1 enzyme pair likely arose via horizontal gene transfer and has been retained in P. aeruginosa over time. Using a murine pulmonary infection model, we showed that both LpxO1 and LpxO2 enzymes are functional in vivo, as direct analysis of in vivo lipid A structure from bronchoalveolar lavage fluid revealed 2-hydroxylated lipid A. Gene expression analysis reveals increased lpxO2 but unchanged lpxO1 expression in vivo, suggesting differential regulation of these enzymes during infection. We also demonstrate that loss-of-function mutations arise in lpxO1 and lpxO2 during chronic lung infection in people with cystic fibrosis (CF), indicating a potential role for pathogenesis and airway adaptation. Collectively, our study characterizes lipid A 2-hydroxylation during P. aeruginosa airway infection that is regulated by two distinct lipid A dioxygenase enzymes.IMPORTANCEPseudomonas aeruginosa is an opportunistic pathogen that causes severe infection in hospitalized and chronically ill individuals. During infection, P. aeruginosa undergoes adaptive changes to evade host defenses and therapeutic interventions, increasing mortality and morbidity. Lipid A structural alteration is one such change that P. aeruginosa isolates undergo during chronic lung infection in CF. Investigating genetic drivers of this lipid A structural variation is crucial in understanding P. aeruginosa adaptation during infection. Here, we describe two lipid A dioxygenases with acyl-chain site specificity, each with different evolutionary origins. Further, we show that loss of function in these enzymes occurs in CF clinical isolates, suggesting a potential pathoadaptive phenotype. Studying these bacterial adaptations provides insight into selection pressures of the CF airway on P. aeruginosa phenotypes that persist during chronic infection. Understanding these adaptive changes may ultimately provide clinicians better control over bacterial populations during chronic infection.


Sujet(s)
Mucoviscidose , Dioxygenases , Infections à Pseudomonas , Humains , Animaux , Souris , Pseudomonas aeruginosa/métabolisme , Lipide A/métabolisme , Infection persistante , Laurate/métabolisme , Hydroxylation , Mucoviscidose/microbiologie , Infections à Pseudomonas/microbiologie , Dioxygenases/métabolisme
12.
Int J Mol Sci ; 24(24)2023 Dec 09.
Article de Anglais | MEDLINE | ID: mdl-38139140

RÉSUMÉ

Previously developed whole-cell vaccines against Bordetella pertussis, the causative agent of whooping cough, appeared to be too reactogenic due to their endotoxin content. Reduction in endotoxicity can generally be achieved through structural modifications in the lipid A moiety of lipopolysaccharides (LPS). In this study, we found that dephosphorylation of lipid A in B. pertussis through the heterologous production of the phosphatase LpxE from Francisella novicida did, unexpectedly, not affect Toll-like receptor 4 (TLR4)-stimulating activity. We then focused on the inner core of LPS, whose synthesis has so far not been studied in B. pertussis. The kdtA and kdkA genes, responsible for the incorporation of a single 3-deoxy-D-manno-oct-2-ulosonic acid (Kdo) residue in the inner core and its phosphorylation, respectively, appeared to be essential. However, the Kdo-bound phosphate could be replaced by a second Kdo after the heterologous production of Escherichia coli kdtA. This structural change in the inner core affected outer-core and lipid A structures and also bacterial physiology, as reflected in cell filamentation and a switch in virulence phase. Furthermore, the eptB gene responsible for the non-stoichiometric substitution of Kdo-bound phosphate with phosphoethanolamine was identified and inactivated. Interestingly, the constructed inner-core modifications affected TLR4-stimulating activity. Whereas endotoxicity studies generally focus on the lipid A moiety, our data demonstrate that structural changes in the inner core can also affect TLR4-stimulating activity.


Sujet(s)
Bordetella pertussis , Lipopolysaccharides , Récepteur de type Toll-4 , Humains , Bordetella pertussis/génétique , Bordetella pertussis/métabolisme , Division cellulaire , Endotoxines/métabolisme , Escherichia coli/métabolisme , Lipide A/métabolisme , Lipopolysaccharides/génétique , Lipopolysaccharides/métabolisme , Mutation , Phosphates/métabolisme , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Coqueluche
13.
Biochemistry ; 62(20): 2970-2981, 2023 10 17.
Article de Anglais | MEDLINE | ID: mdl-37782650

RÉSUMÉ

Covalent modification of lipid A with 4-deoxy-4-amino-l-arabinose (Ara4N) mediates resistance to cationic antimicrobial peptides and polymyxin antibiotics in Gram-negative bacteria. The proteins required for Ara4N biosynthesis are encoded in the pmrE and arnBCADTEF loci, with ArnT ultimately transferring the amino sugar from undecaprenyl-phospho-4-deoxy-4-amino-l-arabinose (C55P-Ara4N) to lipid A. However, Ara4N is N-formylated prior to its transfer to undecaprenyl-phosphate by ArnC, requiring a deformylase activity downstream in the pathway to generate the final C55P-Ara4N donor. Here, we show that deletion of the arnD gene in an Escherichia coli mutant that constitutively expresses the arnBCADTEF operon leads to accumulation of the formylated ArnC product undecaprenyl-phospho-4-deoxy-4-formamido-l-arabinose (C55P-Ara4FN), suggesting that ArnD is the downstream deformylase. Purification of Salmonella typhimurium ArnD (stArnD) shows that it is membrane-associated. We present the crystal structure of stArnD revealing a NodB homology domain structure characteristic of the metal-dependent carbohydrate esterase family 4 (CE4). However, ArnD displays several distinct features: a 44 amino acid insertion, a C-terminal extension in the NodB fold, and sequence divergence in the five motifs that define the CE4 family, suggesting that ArnD represents a new family of carbohydrate esterases. The insertion is responsible for membrane association as its deletion results in a soluble ArnD variant. The active site retains a metal coordination H-H-D triad, and in the presence of Co2+ or Mn2+, purified stArnD efficiently deformylates C55P-Ara4FN confirming its role in Ara4N biosynthesis. Mutations D9N and H233Y completely inactivate stArnD implicating these two residues in a metal-assisted acid-base catalytic mechanism.


Sujet(s)
Lipide A , Polymyxines , Polymyxines/pharmacologie , Polymyxines/métabolisme , Lipide A/métabolisme , Arabinose/métabolisme , Osamines/composition chimique , Antibactériens/pharmacologie , Antibactériens/métabolisme , Escherichia coli/génétique , Escherichia coli/métabolisme , Glucides , Protéines bactériennes/composition chimique
14.
Microbiol Spectr ; 11(4): e0085223, 2023 08 17.
Article de Anglais | MEDLINE | ID: mdl-37432123

RÉSUMÉ

Polymyxins are last-line antibiotics employed against multidrug-resistant (MDR) Klebsiella pneumoniae. Worryingly, polymyxin resistance is rapidly on the rise globally. Polymyxins initially target lipid A of lipopolysaccharides (LPSs) in the cell outer membrane (OM), causing disorganization and cell lysis. While most studies focus on how genetic variations confer polymyxin resistance, the mechanisms of membrane remodeling and metabolic changes in polymyxin-resistant strains remain unclear, thus hampering the development of effective therapies to treat severe K. pneumoniae infections. In the present study, lipid A profiling, OM lipidomics, genomics, and metabolomics were integrated to elucidate the global mechanisms of polymyxin resistance and metabolic adaptation in a polymyxin-resistant strain (strain S01R; MIC of >128 mg/L) obtained from K. pneumoniae strain S01, a polymyxin-susceptible (MIC of 2 mg/L), New Delhi metallo-ß-lactamase (NDM)-producing MDR clinical isolate. Genomic analysis revealed a novel in-frame deletion at position V258 of PhoQ in S01R, potentially leading to lipid A modification with 4-amino-4-deoxy-l-arabinose (L-Ara4N) despite the absence of polymyxin B. Comparative metabolomic analysis revealed slightly elevated levels of energy production and amino acid metabolism in S01R compared to their levels in S01. Exposure to polymyxin B (4 mg/L for S01 and 512 mg/L for S01R) substantially altered energy, nucleotide, and amino acid metabolism and resulted in greater accumulation of lipids in both strains. Furthermore, the change induced by polymyxin B treatment was dramatic at both 1 and 4 h in S01 but only significant at 4 h in S01R. Overall, profound metabolic adaptation was observed in S01R following polymyxin B treatment. These findings contribute to our understanding of polymyxin resistance mechanisms in problematic NDM-producing K. pneumoniae strains and may facilitate the discovery of novel therapeutic targets. IMPORTANCE Antimicrobial resistance (AMR) is a major threat to global health. The emergence of resistance to the polymyxins that are the last line of defense in so-called Gram-negative "superbugs" has further increased the urgency to develop novel therapies. There are frequent outbreaks of K. pneumoniae infections in hospitals being reported, and polymyxin usage is increasing remarkably. Importantly, the polymyxin-resistant K. pneumoniae strains are imposing more severe consequences to health systems. Using metabolomics, lipid A profiling, and outer membrane lipidomics, our findings reveal (i) changes in the pentose phosphate pathway and amino acid and nucleotide metabolism in a susceptible strain following polymyxin treatment and (ii) how cellular metabolism, lipid A modification, and outer membrane remodeling were altered in K. pneumoniae following the acquisition of polymyxin resistance. Our study provides, for the first time, mechanistic insights into metabolic responses to polymyxin treatment in a multidrug-resistant, NDM-producing K. pneumoniae clinical isolate with acquired polymyxin resistance. Overall, these results will assist in identifying new therapeutic targets to combat and prevent polymyxin resistance.


Sujet(s)
Infections à Klebsiella , Polymyxines , Humains , Polymyxines/pharmacologie , Polymyxines/métabolisme , Polymyxine B/pharmacologie , Klebsiella pneumoniae , Lipide A/métabolisme , Antibactériens/pharmacologie , Antibactériens/métabolisme , Métabolisme lipidique , Infections à Klebsiella/traitement médicamenteux , Tests de sensibilité microbienne
15.
Int J Antimicrob Agents ; 62(2): 106873, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37276893

RÉSUMÉ

Colistin resistance in carbapenem-resistant Klebsiella pneumoniae (CRKP) poses health challenges. To investigate the prevalence and molecular characteristics of colistin-resistant CRKP, 708 isolates were collected consecutively from 28 tertiary hospitals in China from 2018 to 2019, and 14 colistin-resistant CRKP were identified. Two-component systems (TCSs) related to colistin resistance (PmrA/B, PhoP/Q, and CrrA/B), the negative regulator mgrB gene and mcr genes, were analysed using genomic sequencing. The relative expression of TCSs genes along with their downstream pmrC and pmrK genes was determined using quantitative real-time PCR (qRT‒PCR). A novel point mutation in PhoQ was confirmed by site-directed mutagenesis, and the subsequent transcriptome changes were analysed by RNA sequencing (RNA-Seq). Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) was used to detect modifications in lipid A. The results showed that only one isolate carried the mcr-8.1 gene, nine exhibited MgrB inactivation or absence, and three exhibited mutations in PmrB. One novel point mutation, L247P, in PhoQ was found to lead to a 64-fold increase in the minimum inhibitory concentration (MIC) of colistin. qRT‒PCR revealed overexpression of phoP/Q and pmrK in isolates with or without MgrB inactivation, and pmrB mutation resulted in overexpression of pmrA and pmrC. Furthermore, transcriptome analysis revealed that the PhoQ L247P novel point mutation caused upregulated expression of phoP/Q and its downstream operon pmrHFIJKLM. Meanwhile, the pmrA/B regulatory pathway did not evolve colistin resistance. Mass spectrometry analysis showed the addition of 4-amino-4-deoxy-L-arabinose (L-Ara4N) to lipid A in colistin-resistant isolates with absence of MgrB. These findings illustrate that the molecular mechanisms of colistin resistance in CRKP isolates are complex, and that MgrB inactivation or absence is the predominant molecular mechanism. Interventions should be initiated to monitor and control colistin resistance.


Sujet(s)
Colistine , Infections à Klebsiella , Humains , Colistine/pharmacologie , Colistine/métabolisme , Antibactériens/pharmacologie , Antibactériens/métabolisme , Klebsiella pneumoniae , Prévalence , Lipide A/métabolisme , Protéines bactériennes/métabolisme , Résistance bactérienne aux médicaments/génétique , Carbapénèmes/pharmacologie , Carbapénèmes/métabolisme , Tests de sensibilité microbienne , Infections à Klebsiella/épidémiologie
16.
Microbiol Spectr ; 11(4): e0483222, 2023 08 17.
Article de Anglais | MEDLINE | ID: mdl-37278652

RÉSUMÉ

Pseudomonas aeruginosa is a versatile pathogen that resists environmental stress, such as suboptimal pH. As a result of exposure to environmental stress, P. aeruginosa shows an altered virulence-related phenotype. This study investigated the modifications that P. aeruginosa undertakes at a mildly low pH (pH 5.0) compared with the bacteria grown in a neutral medium (pH 7.2). Results indicated that in a mildly acidic environment, expression of two-component system genes (phoP/phoQ and pmrA/pmrB), lipid A remodeling genes such as arnT and pagP and virulence genes, i.e., pqsE and rhlA, were induced. Moreover, lipid A of the bacteria grown at a mildly low pH is modified by adding 4-amino-arabinose (l-Ara4N). Additionally, the production of virulence factors such as rhamnolipid, alginate, and membrane vesicles is significantly higher in a mildly low-pH environment than in a neutral medium. Interestingly, at a mildly low pH, P. aeruginosa produces a thicker biofilm with higher biofilm biomass. Furthermore, studies on inner membrane viscosity and permeability showed that a mildly low pH causes a decrease in the inner membrane permeability and increases its viscosity. Besides, despite the importance of PhoP, PhoQ, PmrA, and PmrB in Gram-negative bacteria for responding to low pH stress, we observed that the absence of each of these two-component systems does not meaningfully impact the remodeling of the P. aeruginosa envelope. Given that P. aeruginosa is likely to encounter mildly acidic environments during infection in its host, the alterations that the bacterium undertakes under such conditions must be considered in designing antibacterial strategies against P. aeruginosa. IMPORTANCE P. aeruginosa encounters environments with acidic pH when establishing infections in hosts. The bacterium develops an altered phenotype to tolerate a moderate decrease in the environmental pH. At the level of the bacterial envelope, modified lipid A composition and a reduction of the bacterial inner membrane permeability and fluidity are among the changes P. aeruginosa undergoes at a mildly low pH. Also, the bacterium is more likely to form biofilm in a mildly acidic environment. Overall, these alterations in the P. aeruginosa phenotype put obstacles in the way of antibacterial activities. Thus, considering physiological changes in the bacterium at low pH helps design and implement antimicrobial approaches against this hostile microorganism.


Sujet(s)
Lipide A , Pseudomonas aeruginosa , Virulence/génétique , Pseudomonas aeruginosa/métabolisme , Lipide A/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Antibactériens/pharmacologie , Régulation de l'expression des gènes bactériens
17.
J Med Microbiol ; 72(6)2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37261959

RÉSUMÉ

Background. Klebsiella variicola was once recognised as a benign plant-endosymbiont but recent case reports suggest that it is a newly emerging Gram-negative pathogen related to opportunistic infection of multiple sites in humans.Methods. Antimicrobial susceptibility testing was performed using broth microdilution method. To identify colistin resistance mechanisms, phoPQ, pmrAB, and mgrB were sequenced and their mRNA expression was analysed using quantitative real-time PCR. In addition, we tried to detect crrAB and mcr. The lipid A moieties of colistin-susceptible and -resistant isolates were analysed using MALDI-TOF.Results. Among the two K. variicola isolates, one is colistin-resistant, and another is colistin-susceptible. The colistin-resistant K. variicola isolate showed no mutations in phoPQ, pmrAB, and mgrB, and crrAB and mcr were not identified. However, its phoQ and pbgP expression was significantly higher and amino-arabinosylated lipid A with hexa-acylated species in lipopolysaccharide was identified.Conclusions. We found that colistin resistance in K. variicola was mediated by the modification of lipid A. Although the isolate was obtained from faecal samples of healthy adults, colistin-resistant K. variicola challenges public health as an opportunistic pathogen.


Sujet(s)
Colistine , Infections à Klebsiella , Humains , Adulte , Colistine/pharmacologie , Antibactériens/pharmacologie , Lipide A/métabolisme , Protéines bactériennes/génétique , Klebsiella pneumoniae/génétique , Résistance bactérienne aux médicaments/génétique , Tests de sensibilité microbienne
18.
ChemMedChem ; 18(11): e202300023, 2023 06 01.
Article de Anglais | MEDLINE | ID: mdl-37014664

RÉSUMÉ

Despite the widespread emergence of multidrug-resistant nosocomial Gram-negative bacterial infections and the major public health threat it brings, no new class of antibiotics for Gram-negative pathogens has been approved over the past five decades. Therefore, there is an urgent medical need for developing effective novel antibiotics against multidrug-resistant Gram-negative pathogens by targeting previously unexploited pathways in these bacteria. To fulfill this crucial need, we have been investigating a series of sulfonyl piperazine compounds targeting LpxH, a dimanganese-containing UDP-2,3-diacylglucosamine hydrolase in the lipid A biosynthetic pathway, as novel antibiotics against clinically important Gram-negative pathogens. Inspired by a detailed structural analysis of our previous LpxH inhibitors in complex with K. pneumoniae LpxH (KpLpxH), here we report the development and structural validation of the first-in-class sulfonyl piperazine LpxH inhibitors, JH-LPH-45 (8) and JH-LPH-50 (13), that achieve chelation of the active site dimanganese cluster of KpLpxH. The chelation of the dimanganese cluster significantly improves the potency of JH-LPH-45 (8) and JH-LPH-50 (13). We expect that further optimization of these proof-of-concept dimanganese-chelating LpxH inhibitors will ultimately lead to the development of more potent LpxH inhibitors for targeting multidrug-resistant Gram-negative pathogens.


Sujet(s)
Lipide A , Pyrophosphatases , Domaine catalytique , Pyrophosphatases/métabolisme , Lipide A/métabolisme , Antibactériens/pharmacologie , Antibactériens/composition chimique , Pipérazine , Métaux , Bactéries à Gram négatif , Multirésistance bactérienne aux médicaments , Tests de sensibilité microbienne
19.
Vet Microbiol ; 282: 109759, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37104940

RÉSUMÉ

This study presents the engineering of a less endotoxic Salmonella Typhimurium strain by manipulating the lipid-A structure of the lipopolysaccharide (LPS) component. Salmonella lipid A was dephosphorylated by using lpxE from Francisella tularensis. The 1-phosphate group from lipid-A was removed selectively, resulting in a close analog of monophosphoryl lipid A. We observed a significant impact of ∆pagL on major virulence factors such as biofilm formation, motility, persistency, and immune evasion. In correlation with biofilm and motility retardation, adhesion and invasion were elevated but with reduced intracellular survival, a favorable phenotype prospect of a vaccine strain. Western blotting and silver staining confirmed the absence of the O-antigen and truncated lipid-A core in the detoxified Salmonella mutant. In vitro and in vivo studies demonstrated that the dephosphorylated Salmonella mutant mediated lower pro-inflammatory cytokine secretion than the wild-type strain. The vaccine strains were present in the spleen and liver for five days and were cleared from the organs by day seven. However, the wild-type strain persisted in the spleen, liver, and brain, leading to sepsis-induced death. Histological evaluations of tissue samples further confirmed the reduced endotoxic activity of the detoxified Salmonella mutant. The detoxification strategy did not compromise the level of protective immunity, as the vaccine strain could enhance humoral and cellular immune responses and protect against the wild-type challenge in immunized mice.


Sujet(s)
Salmonelloses , Vaccins antisalmonella , Salmonella typhimurium , Salmonella typhimurium/génétique , Salmonella typhimurium/pathogénicité , Salmonella typhimurium/physiologie , Femelle , Animaux , Souris , Souris de lignée BALB C , Lipide A/métabolisme , Vaccins antisalmonella/effets indésirables , Vaccins antisalmonella/génétique , Vaccins antisalmonella/immunologie , Lipopolysaccharides/métabolisme , Immunité humorale , Immunité cellulaire , Biofilms , Salmonelloses/immunologie , Salmonelloses/microbiologie , Carboxylic ester hydrolases/génétique
20.
J Chem Inf Model ; 63(5): 1592-1601, 2023 03 13.
Article de Anglais | MEDLINE | ID: mdl-36802606

RÉSUMÉ

Capsule is one of the common virulence factors in Gram-negative bacteria protecting pathogens from host defenses and consists of long-chain capsular polysaccharides (CPS) anchored in the outer membrane (OM). Elucidating structural properties of CPS is important to understand its biological functions as well as the OM properties. However, the outer leaflet of the OM in current simulation studies is represented exclusively by LPS due to the complexity and diversity of CPS. In this work, representative Escherichia coli CPS, KLPS (a lipid A-linked form) and KPG (a phosphatidylglycerol-linked form), are modeled and incorporated into various symmetric bilayers with co-existing LPS in different ratios. All-atom molecular dynamics simulations of these systems have been conducted to characterize various bilayer properties. Incorporation of KLPS makes the acyl chains of LPS more rigid and ordered, while incorporation of KPG makes them less ordered and flexible. These results are consistent with the calculated area per lipid (APL) of LPS, in which the APL of LPS becomes smaller when KLPS is incorporated, whereas it gets larger when KPG is included. Torsional analysis reveals that the influence of the CPS presence on the conformational distributions of the glycosidic linkages of LPS is small, and minor differences are also detected for the inner and outer regions of the CPS. Combined with previously modeled enterobacterial common antigens (ECAs) in the form of mixed bilayers, this work provides more realistic OM models as well as the basis for characterization of interactions between the OM and OM proteins.


Sujet(s)
Membrane bactérienne externe , Lipopolysaccharides , Lipopolysaccharides/composition chimique , Membrane cellulaire/métabolisme , Protéines de la membrane externe bactérienne/analyse , Protéines de la membrane externe bactérienne/composition chimique , Protéines de la membrane externe bactérienne/métabolisme , Lipide A/métabolisme , Escherichia coli/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...