Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.211
Filtrer
1.
Methods Mol Biol ; 2851: 39-60, 2024.
Article de Anglais | MEDLINE | ID: mdl-39210170

RÉSUMÉ

Gram-positive bacteria, including lactic acid bacteria (LAB), possess lipoteichoic acid (LTA) on the cell surface. LTA is an amphiphilic molecule typically composed of hydrophilic glycerolphosphate polymer and hydrophobic anchor glycolipid moieties. It is involved in physiological properties of the cell surface and also plays roles in interactions with the host. Appropriate preparation procedures, such as extraction and purification, are required to clarify the structure-activity relationship. Structural diversity of LTA has been reported at the bacterial species and strain levels, and structural differences might affect interactions with the host. This chapter introduces techniques for preparation and structural analysis of LTA derived from LAB. It consists of four sections, covering butanol extraction, hydrophobic interaction chromatography, immunoblotting, and structural analysis. Technical notes containing supplemental information about the individual steps are also provided.


Sujet(s)
Membrane cellulaire , Lactobacillales , Lipopolysaccharides , Acides teichoïques , Acides teichoïques/composition chimique , Lipopolysaccharides/composition chimique , Lactobacillales/métabolisme , Membrane cellulaire/métabolisme , Membrane cellulaire/composition chimique , Interactions hydrophobes et hydrophiles
2.
Int J Mol Sci ; 25(16)2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39201412

RÉSUMÉ

Galectins are ß-galactoside-binding animal lectins involved in various biological functions, such as host defense. Galectin-2 and -3 are members of the galectin family that are expressed in the stomach, including the gastric mucosa and surface mucous cells. Galectin-3 exhibits aggregation and bactericidal activity against Helicobacter pylori in a ß-galactoside-dependent manner. We previously reported that galectin-2 has the same activity under neutral pH conditions. In this study, the H. pylori aggregation activity of galectin-2 was examined under weakly acidic conditions, in which H. pylori survived. Galectin-2 agglutinated H. pylori even at pH 6.0, but not at pH 5.0, correlating with its structural stability, as determined using circular dichroism. Additionally, galectin-2 binding to the lipopolysaccharide (LPS) of H. pylori cultured under weakly acidic conditions was investigated using affinity chromatography and Western blotting. Galectin-2 could bind to H. pylori LPS containing H type I, a Lewis antigen, in a ß-galactoside-dependent manner. In contrast, galectin-3 was structurally more stable than galectin-2 under acidic conditions and bound to H. pylori LPS containing H type I and Lewis X. In conclusion, galectin-2 and -3 might function cooperatively in the defense against H. pylori in the stomach under different pH conditions.


Sujet(s)
Galectine 2 , Helicobacter pylori , Lipopolysaccharides , Helicobacter pylori/métabolisme , Lipopolysaccharides/métabolisme , Lipopolysaccharides/composition chimique , Concentration en ions d'hydrogène , Galectine 2/métabolisme , Galectine 2/composition chimique , Humains , Galectine -3/métabolisme , Galectine -3/composition chimique , Liaison aux protéines , Agglutination , Galectines/métabolisme , Galectines/composition chimique
3.
J Chem Theory Comput ; 20(15): 6890-6903, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39008538

RÉSUMÉ

Lipopolysaccharide (LPS) is a complex glycolipid molecule that is the main lipidic component of the outer leaflet of the outer membrane of Gram-negative bacteria. It has very limited lateral motion compared to phospholipids, which are more ubiquitous in biological membranes, including in the inner leaflet of the outer membrane of Gram-negative bacteria. The slow-moving nature of LPS can present a hurdle for molecular dynamics simulations, given that the (pragmatically) accessible timescales to simulations are currently limited to microseconds, during which LPS displays some conformational dynamics but hardly any lateral diffusion. Thus, it is not feasible to observe phenomena such as insertion of molecules, including antibiotics/antimicrobials, directly into the outer membrane from the extracellular side nor to observe LPS dissociating from proteins via molecular dynamics using currently available models at the atomistic and more coarse-grained levels of granularity. Here, we present a model of deep rough LPS compatible with the Martini 2 coarse-grained force field with scaled down nonbonded interactions to enable faster diffusion. We show that the faster-diffusing LPS model is able to reproduce the salient biophysical properties of the standard models, but due to its faster lateral motion, molecules are able to penetrate deeper into membranes containing the faster model. We show that the fast ReLPS model is able to reproduce experimentally determined patterns of interaction with outer membrane proteins while also allowing for LPS to associate and dissociate with proteins within microsecond timescales. We also complete the Martini 3 LPS toolkit for Escherichia coli by presenting a (standard) model of deep rough LPS for this force field.


Sujet(s)
Escherichia coli , Lipopolysaccharides , Simulation de dynamique moléculaire , Lipopolysaccharides/composition chimique , Escherichia coli/composition chimique , Cinétique , Diffusion
4.
J Biol Chem ; 300(8): 107577, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39019214

RÉSUMÉ

The dimeric architecture of tandem-repeat type galectins, such as galectin-4 (Gal-4), modulates their biological activities, although the underlying molecular mechanisms have remained elusive. Emerging evidence show that tandem-repeat galectins play an important role in innate immunity by recognizing carbohydrate antigens present on the surface of certain pathogens, which very often mimic the structures of the human self-glycan antigens. Herein, we have analyzed the binding preferences of the C-domain of Gal-4 (Gal-4C) toward the ABH-carbohydrate histo-blood antigens with different core presentations and their recognition features have been rationalized by using a combined experimental approach including NMR, solid-phase and hemagglutination assays, and molecular modeling. The data show that Gal-4C prefers A over B antigens (two-fold in affinity), contrary to the N-domain (Gal-4N), although both domains share the same preference for the type-6 presentations. The behavior of the full-length Gal-4 (Gal-4FL) tandem-repeat form has been additionally scrutinized. Isothermal titration calorimetry and NMR data demonstrate that both domains within full-length Gal-4 bind to the histo-blood antigens independently of each other, with no communication between them. In this context, the heterodimeric architecture does not play any major role, apart from the complementary A and B antigen binding preferences. However, upon binding to a bacterial lipopolysaccharide containing a multivalent version of an H-antigen mimetic as O-antigen, the significance of the galectin architecture was revealed. Indeed, our data point to the linker peptide domain and the F-face of the C-domain as key elements that provide Gal-4 with the ability to cross-link multivalent ligands, beyond the glycan binding capacity of the dimer.


Sujet(s)
Galectine 4 , Lipopolysaccharides , Oligosaccharides , Humains , Lipopolysaccharides/composition chimique , Galectine 4/métabolisme , Galectine 4/composition chimique , Oligosaccharides/composition chimique , Oligosaccharides/métabolisme , Multimérisation de protéines , Liaison aux protéines , Système ABO de groupes sanguins/composition chimique , Système ABO de groupes sanguins/métabolisme , Domaines protéiques
5.
Int J Mol Sci ; 25(12)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38928052

RÉSUMÉ

Bacterial endotoxins (lipopolysaccharides (LPSs)) are important mediators of inflammatory processes induced by Gram-negative microorganisms. LPSs are the key inducers of septic shock due to a Gram-negative bacterial infection; thus, the structure and functions of LPSs are of specific interest. Often, highly purified bacterial endotoxins must be isolated from small amounts of biological material. Each of the currently available methods for LPS extraction has certain limitations. Herein, we describe a rapid and simple microscale method for extracting LPSs. The method consists of the following steps: ultrasonic destruction of the bacterial material, LPS extraction via heating, LPS purification with organic solvents, and treatment with proteinase K. LPSs that were extracted by using this method contained less than 2-3% protein and 1% total nucleic acid. We also demonstrated the structural integrity of the O-antigen and lipid A via the sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) methods, respectively. We demonstrated the ability of the extracted LPSs to induce typical secretion of cytokines and chemokines by primary macrophages. Overall, this method may be used to isolate purified LPSs with preserved structures of both the O-antigen and lipid A and unchanged functional activity from small amounts of bacterial biomass.


Sujet(s)
Lipopolysaccharides , Spectrométrie de masse MALDI , Lipopolysaccharides/isolement et purification , Lipopolysaccharides/composition chimique , Spectrométrie de masse MALDI/méthodes , Animaux , Souris , Macrophages/métabolisme , Lipide A/composition chimique , Lipide A/isolement et purification , Cytokines/métabolisme , Endopeptidase K/métabolisme , Endopeptidase K/composition chimique , Électrophorèse sur gel de polyacrylamide/méthodes
6.
Anal Chem ; 96(22): 9151-9158, 2024 06 04.
Article de Anglais | MEDLINE | ID: mdl-38758019

RÉSUMÉ

Lipopolysaccharides (LPS) and lipooligosaccharides (LOS) are located in the outer membrane of Gram-negative bacteria and are comprised of three distinctive parts: lipid A, core oligosaccharide (OS), and O-antigen. The structure of each region influences bacterial stability, toxicity, and pathogenesis. Here, we highlight the use of targeted activated-electron photodetachment (a-EPD) tandem mass spectrometry to characterize LPS and LOS from two crucial players in the human gut microbiota, Escherichia coli Nissle and Bacteroides fragilis. a-EPD is a hybrid activation method that uses ultraviolet photoirradiation to generate charge-reduced radical ions followed by collisional activation to produce informative fragmentation patterns. We benchmark the a-EPD method for top-down characterization of triacyl LOS from E. coli R2, then focus on characterization of LPS from E. coli Nissle and B. fragilis. Notably, a-EPD affords extensive fragmentation throughout the backbone of the core OS and O-antigen regions of LPS from E. coli Nissle. This hybrid approach facilitated the elucidation of structural details for LPS from B. fragilis, revealing a putative hexuronic acid (HexA) conjugated to lipid A.


Sujet(s)
Escherichia coli , Lipopolysaccharides , Lipopolysaccharides/composition chimique , Escherichia coli/composition chimique , Bacteroides fragilis/composition chimique , Électrons , Spectrométrie de masse en tandem
7.
Int J Biol Macromol ; 271(Pt 1): 132540, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38782319

RÉSUMÉ

Lipoteichoic acid (LTA) in the gram-positive bacterial cell wall acts as an immunomodulatory factor in host cells. The chemical structures vary among bacterial species and strains, and may be related to biological activities. In our previous work, much higher immunoglobulin A (IgA)-inducing activity was observed in cells of the Apilactobacillus genus (Apilactobacillus kosoi 10HT, Apilactobacillus apinorum JCM 30765T, and Apilactobacillus kunkeei JCM 16173T) than other lactic acid bacteria, and their LTA was responsible for the activity. In the present study, we elucidated the chemical structures of LTA from these Apilactobacillus strains to explore the structure-function relationship of the IgA-inducing activity. The 1H-nuclear magnetic resonance spectra suggested that their LTA structures were similar. All have a poly-glycerolphosphate main chain, which comprised 12 to 20 average number of the repeating units, with partial substitutions of glucose(α1-, glucosyl(α1-2)glucose(α1- (α-linked-kojibiose), and l-lysine at the C-2 hydroxy group of the glycerol residue. l-Lysine is a substituent never seen before in LTA, and is a probable characteristic of the Apilactobacillus genus. Removal of l-lysine residue from LTA by mild alkaline treatment decreased IgA induction in murine Peyer's patch experiments. The novel l-lysine residue in Apilactobacillus LTA plays a crucial role in the remarkably high IgA-inducing activity.


Sujet(s)
Immunoglobuline A , Lipopolysaccharides , Lysine , Acides teichoïques , Acides teichoïques/composition chimique , Lipopolysaccharides/composition chimique , Lipopolysaccharides/pharmacologie , Animaux , Lysine/composition chimique , Souris , Glycérophosphate/composition chimique , Lactobacillaceae/composition chimique
8.
Curr Opin Struct Biol ; 87: 102828, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38723580

RÉSUMÉ

Recent advances in molecular dynamics (MD) simulations have led to rapid improvement in our understanding of the molecular details of the outer membranes (OMs) of Gram-negative bacteria. In this review, we highlight the latest discoveries from MD simulations of OMs, shedding light on the dynamic nature of these bacteria's first line of defense. With the focus on cutting-edge approaches, we explore the OM's sensitivity to structural features, including divalent cations and membrane composition, which have emerged as crucial determinants of antimicrobial passage. Additionally, studies have provided novel insights into outer-membrane proteins (OMPs), revealing their intricate roles in substrate translocation and their distinct interactions with lipopolysaccharides (LPS) in the OM. Finally, we explore the challenging process of ß-barrel membrane protein insertion, showcasing recent findings that have enhanced our grasp of this fundamental biological phenomenon.


Sujet(s)
Protéines de la membrane externe bactérienne , Bactéries à Gram négatif , Simulation de dynamique moléculaire , Bactéries à Gram négatif/métabolisme , Bactéries à Gram négatif/composition chimique , Protéines de la membrane externe bactérienne/composition chimique , Protéines de la membrane externe bactérienne/métabolisme , Membrane bactérienne externe/métabolisme , Membrane bactérienne externe/composition chimique , Lipopolysaccharides/composition chimique , Lipopolysaccharides/métabolisme
9.
Glycoconj J ; 41(2): 119-131, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38642279

RÉSUMÉ

Gram-negative bacteria living in marine waters have evolved peculiar adaptation strategies to deal with the numerous stress conditions that characterize aquatic environments. Among the multiple mechanisms for efficient adaptation, these bacteria typically exhibit chemical modifications in the structure of the lipopolysaccharide (LPS), which is a fundamental component of their outer membrane. In particular, the glycolipid anchor to the membrane of marine bacteria LPSs, i.e. the lipid A, frequently shows unusual chemical structures, which are reflected in equally singular immunological properties with potential applications as immune adjuvants or anti-sepsis drugs. In this work, we determined the chemical structure of the lipid A from Cellulophaga pacifica KMM 3664T isolated from the Sea of Japan. This bacterium showed to produce a heterogeneous mixture of lipid A molecules that mainly display five acyl chains and carry a single phosphate and a D-mannose disaccharide on the glucosamine backbone. Furthermore, we proved that C. pacifica KMM 3664T LPS acts as a weaker activator of Toll-like receptor 4 (TLR4) compared to the prototypical enterobacterial Salmonella typhimurium LPS. Our results are relevant to the future development of novel vaccine adjuvants and immunomodulators inspired by marine LPS chemistry.


Sujet(s)
Lipide A , Lipide A/composition chimique , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/composition chimique , Membrane bactérienne externe/métabolisme , Membrane bactérienne externe/composition chimique , Animaux , Lipopolysaccharides/composition chimique , Souris
10.
Proc Natl Acad Sci U S A ; 121(17): e2403206121, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38630725

RÉSUMÉ

Mycobacterium abscessus is increasingly recognized as the causative agent of chronic pulmonary infections in humans. One of the genes found to be under strong evolutionary pressure during adaptation of M. abscessus to the human lung is embC which encodes an arabinosyltransferase required for the biosynthesis of the cell envelope lipoglycan, lipoarabinomannan (LAM). To assess the impact of patient-derived embC mutations on the physiology and virulence of M. abscessus, mutations were introduced in the isogenic background of M. abscessus ATCC 19977 and the resulting strains probed for phenotypic changes in a variety of in vitro and host cell-based assays relevant to infection. We show that patient-derived mutational variations in EmbC result in an unexpectedly large number of changes in the physiology of M. abscessus, and its interactions with innate immune cells. Not only did the mutants produce previously unknown forms of LAM with a truncated arabinan domain and 3-linked oligomannoside chains, they also displayed significantly altered cording, sliding motility, and biofilm-forming capacities. The mutants further differed from wild-type M. abscessus in their ability to replicate and induce inflammatory responses in human monocyte-derived macrophages and epithelial cells. The fact that different embC mutations were associated with distinct physiologic and pathogenic outcomes indicates that structural alterations in LAM caused by nonsynonymous nucleotide polymorphisms in embC may be a rapid, one-step, way for M. abscessus to generate broad-spectrum diversity beneficial to survival within the heterogeneous and constantly evolving environment of the infected human airway.


Sujet(s)
Mycobacterium abscessus , Humains , Protéines bactériennes/génétique , Lipopolysaccharides/composition chimique , Mutation
11.
Chemphyschem ; 25(14): e202400147, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38625051

RÉSUMÉ

We investigated, by using all-atom molecular dynamics simulations, the effect of the outer membrane of Gram-negative bacteria, composed in the outer leaflet by polar/charged lipopolysaccharides (LPS), on the electrostatic properties of general porins from the Enterobacteriaceae family. General porins constitute the main path for the facilitated diffusion of polar antibiotics through the outer membrane. As model system we selected OmpK36 from Klebsiella pneumoniae, the ortholog of OmpC from Escherichia coli. This species presents high variability of amino acid composition of porins, with the effect to increase its resistance to the penetration of antibiotics. The various properties we analyzed seem to indicate that LPS acts as an independent layer without affecting the internal electrostatic properties of OmpK36. The only apparent effect on the microsecond time scale we sampled is the appearance of calcium ions, when present at moderate concentration in solution, inside the pore. However, we noticed increased fluctuations of the polarization density and only minor changes on its average value.


Sujet(s)
Lipopolysaccharides , Simulation de dynamique moléculaire , Porines , Électricité statique , Lipopolysaccharides/composition chimique , Porines/composition chimique , Porines/métabolisme , Klebsiella pneumoniae/effets des médicaments et des substances chimiques , Klebsiella pneumoniae/composition chimique , Enterobacteriaceae/effets des médicaments et des substances chimiques , Enterobacteriaceae/composition chimique , Enterobacteriaceae/métabolisme , Escherichia coli/effets des médicaments et des substances chimiques , Escherichia coli/composition chimique
12.
Int J Mol Sci ; 25(5)2024 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-38474006

RÉSUMÉ

The lipopolysaccharide (LPS) that resides on the outermost surface and protects Gram-negative bacteria from host defenses is one of the key components leading to Salmonella infection, particularly the endotoxic lipid A domain of LPS. Lipid A modifications have been associated with several genes such as the arnT that encodes 4-amino-4-deoxy-L-arabinose transferase, which can be critical for bacteria to resist cationic antimicrobial peptides and interfere with host immune recognition. However, the association of arnT with virulence is not completely understood. Thus, this study aimed to elucidate the interrelationship of the major lipid A modification gene arnT with Salmonella Typhimurium virulence. We observed that the arnT-deficient S. Typhimurium (JOL2943), compared to the wild type (JOL401), displayed a significant decrease in several virulence phenotypes such as polymyxin B resistance, intracellular survival, swarming, and biofilm and extracellular polymeric substance (EPS) production. Interestingly, the cell-surface hydrophobicity, adhesion, and invasion characteristics remained unaffected. Additionally, LPS isolated from the mutant induced notably lower levels of endotoxicity-related cytokines in RAW and Hela cells and mice, particularly IL-1ß with a nine-fold decrease, than WT. In terms of in vivo colonization, JOL2943 showed diminished presence in internal organs such as the spleen and liver by more than 60%, while ileal infectivity remained similar to JOL401. Overall, the arnT deletion rendered the strain less virulent, with low endotoxicity, maintained gut infectivity, and reduced colonization in internal organs. With these ideal characteristics, it can be further explored as a potential attenuated Salmonella strain for therapeutics or vaccine delivery systems.


Sujet(s)
Lipide A , Salmonella typhimurium , Humains , Animaux , Souris , Salmonella typhimurium/génétique , Lipide A/composition chimique , Lipopolysaccharides/composition chimique , Virulence , Matrice de substances polymériques extracellulaires , Cellules HeLa , Protéines bactériennes/génétique
13.
Carbohydr Res ; 538: 109089, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38513463

RÉSUMÉ

Diazotrophic bacteria of the genus Azospirillum are known widely, because they are ubiquitous in the rhizosphere and can promote the growth and performance of nonlegume plants. Recently, more Azospirillum species have been isolated from sources other than plants or soil. We report the structures of the O polysaccharides (OPSs) from the lipopolysaccharides of the type strains A. thiophilum BV-ST (1) and A. griseum L-25-5w-1T (2), isolated from aquatic environments. Both structures have a common tetrarhamnan in the repeating-unit, which is decorated with a side xylose in the OPS of A. thiophilum BV-ST.


Sujet(s)
Azospirillum , Lipopolysaccharides , Lipopolysaccharides/composition chimique , Azospirillum/composition chimique , Polyosides
14.
Carbohydr Polym ; 332: 121928, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38431400

RÉSUMÉ

Published work has shown that glycoconjugate vaccines, based on truncated detoxified lipopolysaccharides from Moraxella catarrhalis attached through their reducing end to a carrier protein, gave good protection for all three serotypes A, B, and C in mice immunisation experiments. The (from the non-reducing end) truncated LPS structures were obtained from bacterial glycosyl transferase knock-out mutants and contained the de-esterified Lipid A, two Kdo residues and five glucose moieties. This work describes the chemical synthesis of the same outer Moraxella LPS structures, spacer-equipped and further truncated from the reducing end, i.e., without the Lipid A part and containing four or five glucose moieties or four glucose moieties and one Kdo residue, and their subsequent conjugation to a carrier protein via a five­carbon bifunctional spacer to form glycoconjugates. Immunisation experiments both in mice and rabbits of these gave a good antibody response, being 2-7 times that of pre-immune sera. However, the sera produced only recognized the immunizing glycan immunogens and failed to bind to native LPS or whole bacterial cells. Comparative molecular modelling of three alternative antigens shows that an additional (2 â†’ 4)-linked Kdo residue, not present in the synthetic structures, has a significant impact on the shape and volume of the molecule, with implications for antigen binding and cross-reactivity.


Sujet(s)
Lipopolysaccharides , Moraxella catarrhalis , Lapins , Animaux , Souris , Lipopolysaccharides/composition chimique , Lipide A , Anticorps antibactériens , Glycoconjugués , Oligosaccharides/composition chimique , Glucose , Protéines de transport
15.
Org Lett ; 26(10): 2103-2107, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38443201

RÉSUMÉ

A chemical synthesis of a unique nanosaccharide fragment from Helicobacter pylori lipopolysaccharide was achieved via a convergent glycosylation method. Challenges involved in the synthesis include the highly stereoselective construction of ß-3-deoxy-d-manno-oct-2-ulosonic acid (Kdo) and two 1,2-cis-glycosidic linkages, as well as the formation of a branched 2,7-disubstituted heptose subunit. Hydrogen-bond mediated aglycone delivery strategy and benzoyl-directing remote participation effect were employed, respectively, for the efficient generation of the desired ß-Kdo glycoside and 1,2-cis-α-l-fucoside/d-glucoside. Moreover, the key branched framework was successfully established through a [(7 + 1) + 1] assembly approach involving the stepwise glycosylation of the heptasaccharide alcohol with two monosaccharide donors. The synthesized 1 containing a propylamine linker at the reducing end can be covalently bound to a carrier protein for further immunological studies.


Sujet(s)
Hétérosides , Lipopolysaccharides , Lipopolysaccharides/composition chimique , Hétérosides/composition chimique
16.
Int J Mol Sci ; 25(4)2024 Feb 06.
Article de Anglais | MEDLINE | ID: mdl-38396650

RÉSUMÉ

Lipopolysaccharides (LPSs) are major components of the outer membranes of Gram-negative bacteria. In this work, the structure of the O-polysaccharide of Ochrobactrum quorumnocens T1Kr02 was identified by nuclear magnetic resonance (NMR), and the physical-chemical properties and biological activity of LPS were also investigated. The NMR analysis showed that the O-polysaccharide has the following structure: →2)-ß-d-Fucf-(1→3)-ß-d-Fucp-(1→. The structure of the periplasmic glucan coextracted with LPS was established by NMR spectroscopy and chemical methods: →2)-ß-d-Glcp-(1→. Non-stoichiometric modifications were identified in both polysaccharides: 50% of d-fucofuranose residues at position 3 were O-acetylated, and 15% of d-Glcp residues at position 6 were linked with succinate. This is the first report of a polysaccharide containing both d-fucopyranose and d-fucofuranose residues. The fatty acid analysis of the LPS showed the prevalence of 3-hydroxytetradecanoic, hexadecenoic, octadecenoic, lactobacillic, and 27-hydroxyoctacosanoic acids. The dynamic light scattering demonstrated that LPS (in an aqueous solution) formed supramolecular particles with a size of 72.2 nm and a zeta-potential of -21.5 mV. The LPS solution (10 mkg/mL) promoted the growth of potato microplants under in vitro conditions. Thus, LPS of O. quorumnocens T1Kr02 can be recommended as a promoter for plants and as a source of biotechnological production of d-fucose.


Sujet(s)
Lipopolysaccharides , Ochrobactrum , Lipopolysaccharides/composition chimique , Fucose/composition chimique , Antigènes O/composition chimique , Bactéries
17.
ACS Infect Dis ; 10(2): 763-778, 2024 02 09.
Article de Anglais | MEDLINE | ID: mdl-38259029

RÉSUMÉ

Gram-negative bacteria possess a complex structural cell envelope that constitutes a barrier for antimicrobial peptides that neutralize the microbes by disrupting their cell membranes. Computational and experimental approaches were used to study a model outer membrane interaction with an antimicrobial peptide, melittin. The investigated membrane included di[3-deoxy-d-manno-octulosonyl]-lipid A (KLA) in the outer leaflet and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE) in the inner leaflet. Molecular dynamics simulations revealed that the positively charged helical C-terminus of melittin anchors rapidly into the hydrophilic headgroup region of KLA, while the flexible N-terminus makes contacts with the phosphate groups of KLA, supporting melittin penetration into the boundary between the hydrophilic and hydrophobic regions of the lipids. Electrochemical techniques confirmed the binding of melittin to the model membrane. To probe the peptide conformation and orientation during interaction with the membrane, polarization modulation infrared reflection absorption spectroscopy was used. The measurements revealed conformational changes in the peptide, accompanied by reorientation and translocation of the peptide at the membrane surface. The study suggests that melittin insertion into the outer membrane affects its permeability and capacitance but does not disturb the membrane's bilayer structure, indicating a distinct mechanism of the peptide action on the outer membrane of Gram-negative bacteria.


Sujet(s)
Peptides antimicrobiens , Lipopolysaccharides , Lipopolysaccharides/composition chimique , Mélittine/composition chimique , Peptides/composition chimique , Bactéries à Gram négatif/métabolisme
18.
J Colloid Interface Sci ; 659: 397-412, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38183806

RÉSUMÉ

BACKGROUND: Clinical treatments ofgastric infections using antibiotics suffer from the undesired killing of commensal bacteria and emergence of antibiotic resistance. It is desirable to develop pH-responsive antimicrobial peptides (AMPs) that kill pathogenic bacteria such as H. pyloriand resistant E. coli under acidic environment with minimal impact to commensal bacteria whilst not causing antibiotic resistance. EXPERIMENTS: Using a combined approach of cell assays, molecular dynamics (MD) simulations and membrane models facilitating biophysical and biochemical measurements including small angle neutron scattering (SANS), we have characterized the pH-responsive physiochemical properties and antimicrobial performance of two amphiphilic AMPs, GIIKDIIKDIIKDI-NH2 and GIIKKIIDDIIKKI-NH2 (denoted as 3D and 2D, respectively), that were designed by selective substitutions of cationic residues of Lys (K) in the extensively studied AMP G(IIKK)3I-NH2 with anionic residue Asp (D). FINDINGS: Whilst 2D kept non-ordered coils across the entire pH range studied, 3D displayed a range of secondary structures when pH was shifted from basic to acidic, with distinct self-assembly into nanofibers in aqueous environment. Further experimental and modeling studies revealed that the AMPs interacted differently with the inner and outer membranes of Gram-negative bacteria in a pH-responsive manner and that the structural features characterized by membrane leakage and intramembrane nanoaggregates revealed from fluorescence spectroscopy and SANS were well linked to antimicrobial actions. Different antimicrobial efficacies of 2D and 3D were underlined by the interplay between their ability to bind to the outer membrane lipid LPS (lipopolysaccharide), outer membrane permeability change and inner membrane depolarization and leakage. Furthermore, AMP's binding with the inner membrane under acidic condition caused both the dissipation of membrane potential (Δψ) and the continuous dissipation of transmembrane ΔpH, with Δψ and ΔpH being the key components of the proton motive force. Combinations of antibiotic (Minocycline) with the pH-responsive AMP generated the synergistic effects against Gram-negative bacteria only under acidic condition. These features are crucial to target applications to gastric infections, anti-acne and wound healing.


Sujet(s)
Antibactériens , Anti-infectieux , Antibactériens/composition chimique , Peptides antimicrobiens cationiques/composition chimique , Escherichia coli , Bactéries à Gram négatif , Anti-infectieux/pharmacologie , Lipopolysaccharides/composition chimique , Bactéries/métabolisme , Concentration en ions d'hydrogène , Tests de sensibilité microbienne
19.
Int J Biol Macromol ; 261(Pt 1): 129516, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38278393

RÉSUMÉ

The lipopolysaccharides of Herbaspirillum lusitanum P6-12T (HlP6-12T) and H. frisingense GSF30T (HfGSF30T) was isolated by phenol-water extraction from bacterial cells and was characterized using chemical analysis and SDS-PAGE. It was shown that these bacteria produce LPSs that differ in their physicochemical properties and macromolecular organization. In this paper, the lipid A structure of the HlP6-12T LPS, was characterized through chemical analyses and matrix-assisted laser desorption ionization (MALDI) mass spectrometry. To prove the effect of the size of micelles on their bioavailability, we examined the activity of both LPSs toward the morphology of wheat seedlings. Analysis of the HlP6-12T and HfGSF30T genomes showed no significant differences between the operons that encode proteins involved in the biosynthesis of the lipids A and core oligosaccharides. The difference may be due to the composition of the O-antigen operon. HfGSF30T has two copies of the rfb operon, with the main one divided into two fragments. In contrast, the HlP6-12T genome contains only a single rfb-containing operon, and the other O-antigen operons are not comparable at all. The integrity of O-antigen-related genes may also affect LPS variability of. Specifically, we have observed a hairpin structure in the middle of the O-antigen glycosyltransferase gene, which led to the division of the gene into two fragments, resulting in incorrect protein synthesis and potential abnormalities in O-antigen production.


Sujet(s)
Herbaspirillum , Lipopolysaccharides , Lipopolysaccharides/composition chimique , Antigènes O/métabolisme , Interactions hôte-microbes , Herbaspirillum/génétique , Chromatographie gazeuse-spectrométrie de masse , Spectrométrie de masse MALDI
20.
J Chem Theory Comput ; 20(4): 1704-1716, 2024 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-37676287

RÉSUMÉ

The outer lipopolysaccharide (LPS) membrane of Gram-negative bacteria forms the main barrier for transport of antimicrobial molecules into the bacterial cell. In this study we develop coarse-grained models for the outer membrane of Escherichia coli in the Martini-3 framework. The coarse-grained model force field was parametrized and validated using all-atom simulations of symmetric membranes of lipid A and rough LPS as well as a complete asymmetric membrane of LPS with the O-antigen. The bonded parameters were obtained using an iterative refinement procedure with target bonded distributions obtained from all-atom simulations. The membrane thickness, area of the LPS, and density distributions for the different regions as well as the water and ion densities in Martini-3 simulations show excellent agreement with the all-atom data. Additionally the solvent accessible surface area for individual molecules in water was found to be in good agreement. The binding of calcium ions with phosphate and carboxylate moieties of LPS is accurately captured in the Martini-3 model, indicative of the integrity of the highly negatively charged LPS molecules in the outer membranes of Gram-negative bacteria. The melting transition of the coarse-grained lipid A membrane model was found to occur between 300 and 310 K, and the model captured variations in area per LPS, order parameter, and membrane thickness across the melting transition. Our study reveals that the proposed Martini-3 models for LPS are able to capture the physicochemical balance of the complex sugar architecture of the outer membrane of Escherichia coli. The coarse-grained models developed in this study would be useful for determining membrane protein interactions and permeation of potential antimicrobials through bacterial membranes at mesoscopic spatial and temporal scales.


Sujet(s)
Lipide A , Lipopolysaccharides , Lipopolysaccharides/composition chimique , Escherichia coli , Simulation de dynamique moléculaire , Bactéries à Gram négatif/composition chimique , Eau
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE