Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 3947, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850651

ABSTRACT

UDP-N-acetylglucosamine (UDP-GlcNAc) acyltransferase (LpxA) catalyzes the first step of lipid A biosynthesis, the transfer of an R-3-hydroxyacyl chain from its acyl carrier protein (ACP) to the 3-OH group of UDP-GlcNAc. Essential in the growth of Gram-negative bacteria, LpxA is a logical target for antibiotics design. A pentadecapeptide (Peptide 920) with high affinity towards LpxA was previously identified in a phage display library. Here we created a small library of systematically designed peptides with the length of four to thirteen amino acids using Peptide 920 as a scaffold. The concentrations of these peptides at which 50% of LpxA is inhibited (IC50) range from 50 nM to >100 µM. We determined the crystal structure of E. coli LpxA in a complex with a potent inhibitor. LpxA-inhibitor interaction, solvent model and all contributing factors to inhibitor efficacy were well resolved. The peptide primarily occludes the ACP binding site of LpxA. Interactions between LpxA and the inhibitor are different from those in the structure of Peptide 920. The inhibitory peptide library and the crystal structure of inhibitor-bound LpxA described here may further assist in the rational design of inhibitors with antimicrobial activity that target LpxA and potentially other acyltransferases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Design , Peptides/pharmacology , Uridine Diphosphate N-Acetylglucosamine/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Inhibitory Concentration 50 , Lipid A/antagonists & inhibitors , Lipid A/biosynthesis , Peptide Library , Peptides/chemistry
2.
Article in English | MEDLINE | ID: mdl-29938202

ABSTRACT

Lipid A is an essential basal component of lipopolysaccharide of most Gram-negative bacteria. Inhibitors targeting LpxC, a conserved enzyme in lipid A biosynthesis, are antibiotic candidates against Gram-negative pathogens. Here we report the characterization of the role of lipid A in Coxiella burnetii growth in axenic media, monkey kidney cells (BGMK and Vero), and macrophage-like THP-1 cells by using a potent LpxC inhibitor -LPC-011. We first determined the susceptibility of C. burnetii LpxC to LPC-011 in a surrogate E. coli model. In E. coli, the minimum inhibitory concentration (MIC) of LPC-011 against C. burnetii LpxC is < 0.05 µg/mL, a value lower than the inhibitor's MIC against E. coli LpxC. Considering the inhibitor's problematic pharmacokinetic properties in vivo and Coxiella's culturing time up to 7 days, the stability of LPC-011 in cell cultures was assessed. We found that regularly changing inhibitor-containing media was required for sustained inhibition of C. burnetii LpxC in cells. Under inhibitor treatment, Coxiella has reduced growth yields in axenic media and during replication in non-phagocytic cells, and has a reduced number of productive vacuoles in such cells. Inhibiting lipid A biosynthesis in C. burnetii by the inhibitor was shown in a phase II strain transformed with chlamydial kdtA. This exogenous KdtA enzyme modifies Coxiella lipid A with an α-Kdo-(2 → 8)-α-Kdo epitope that can be detected by anti-chlamydia genus antibodies. In inhibitor-treated THP-1 cells, Coxiella shows severe growth defects characterized by poor vacuole formation and low growth yields. Coxiella progenies prepared from inhibitor-treated cells retain the capability of normally infecting all tested cells in the absence of the inhibitor, which suggests a dispensable role of lipid A for infection and early vacuole development. In conclusion, our data suggest that lipid A has significance for optimal development of Coxiella-containing vacuoles, and for robust multiplication of C. burnetii in macrophage-like THP-1 cells. Unlike many bacteria, C. burnetii replication in axenic media and non-phagocytic cells was less dependent on normal lipid A biosynthesis.


Subject(s)
Axenic Culture/methods , Coxiella burnetii/growth & development , Coxiella burnetii/pathogenicity , Lipid A/antagonists & inhibitors , Macrophages/microbiology , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/genetics , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Chlorocebus aethiops , Coxiella burnetii/drug effects , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli/pathogenicity , Humans , Hydroxamic Acids/pharmacology , Lipid A/genetics , Macrophages/drug effects , THP-1 Cells , Threonine/analogs & derivatives , Threonine/pharmacology , Vacuoles/drug effects , Vacuoles/microbiology , Vero Cells
3.
Nano Lett ; 18(5): 2864-2869, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29589756

ABSTRACT

Endotoxicity originating from a dangerous debris (i.e., lipopolysaccharide, LPS) of Gram-negative bacteria is a challenging clinical problem, but no drugs or therapeutic strategies that can successfully address this issue have been identified yet. In this study, we report a subnanometer gold cluster that can efficiently block endotoxin activity to protect against sepsis. The endotoxin blocker consists of a gold nanocluster that serves as a flakelike substrate and a coating of short alkyl motifs that act as an adhesive to dock with LPS by compacting the intramolecular hydrocarbon chain-chain distance ( d-spacing) of lipid A, an endotoxicity active site that can cause overwhelming cytokine induction resulting in sepsis progression. Direct evidence showed the d-spacing values of lipid A to be decreased from 4.19 Å to either 3.85 or 3.54 Å, indicating more dense packing densities in the presence of subnanometer gold clusters. In terms of biological relevance, the concentrations of key pro-inflammatory NF-κB-dependent cytokines, including plasma TNF-α, IL-6, and IL-1ß, and CXC chemokines, in LPS-challenged mice showed a noticeable decrease. More importantly, we demonstrated that the treatment of antiendotoxin gold nanoclusters significantly prolonged the survival time in LPS-induced septic mice. The ultrasmall gold nanoclusters could target lipid A of LPS to deactivate endotoxicity by compacting its packing density, which might constitute a potential therapeutic strategy for the early prevention of sepsis caused by Gram-negative bacterial infection.


Subject(s)
Gold/therapeutic use , Lipid A/antagonists & inhibitors , Metal Nanoparticles/therapeutic use , Sepsis/therapy , Animals , Cytokines/blood , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred C57BL , Sepsis/blood , Sepsis/chemically induced
4.
Microb Pathog ; 94: 2-11, 2016 May.
Article in English | MEDLINE | ID: mdl-26318142

ABSTRACT

OBJECTIVE: CL(14-25), a dodecapeptide of cyanate lyase from rice, is a novel cationic α-helical antimicrobial peptide. In this study, we examined inhibitory ability of CL(14-25) against endotoxic activities of lipopolysaccharides (LPSs) from Escherichia coli and periodontal pathogenic Aggregatibacter actinomycetemcomitans. METHODS: Endotoxin-neutralizing activity of CL(14-25) was evaluated by inhibition to induction of cytokine and nitric oxide in human aortic endothelial cells (HAECs) and RAW264 mouse macrophage cells, respectively. Protective effect of CL(14-25) was determined in mice against lethal toxicity of LPS. RESULTS: IL-6 in HAECs was induced by stimulation with LPS preparations of A. actinomycetemcomitans and E. coli tested in this study, and addition of CL(14-25) to the medium caused inhibition of their induction in a dose-dependent manner. CL(14-25) inhibited NO induction in RAW264 cells by a smooth type LPS of E. coli O55:B5 and an Rc type LPS of E. coli J5 as well as lipid A of E. coli R515 in a dose-dependent manner. Simultaneous injection of E. coli O55:B5 LPS and CL(14-25) in BALB/c mice resulted in prevention of lethal toxicity of the former. The results of a Limulus amebocyte lysate assay and surface plasmon resonance analysis of interaction between CL(14-25) and E. coli LPS or lipid A showed that CL(14-25) specifically binds to a lipid A moiety of LPS. CONCLUSION: The results of present study suggest that CL(14-25) has a potential to be used as a nutraceutical agent for periodontal therapy.


Subject(s)
Aggregatibacter actinomycetemcomitans/metabolism , Carbon-Nitrogen Lyases/chemistry , Escherichia coli/metabolism , Lipopolysaccharides/antagonists & inhibitors , Peptide Fragments/pharmacology , Aggregatibacter actinomycetemcomitans/chemistry , Animals , Cytokines/biosynthesis , Cytokines/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Endothelial Cells/drug effects , Escherichia coli/chemistry , Humans , Interleukin-6/biosynthesis , Lipid A/antagonists & inhibitors , Lipid A/chemistry , Lipid A/toxicity , Lipopolysaccharides/chemistry , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Oryza/enzymology , Peptide Fragments/chemistry , RAW 264.7 Cells
5.
Biochim Biophys Acta ; 1858(5): 980-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26514603

ABSTRACT

The bacterial membrane provides a target for antimicrobial peptides. There are two groups of bacteria that have characteristically different surface membranes. One is the Gram-negative bacteria that have an outer membrane rich in lipopolysaccharide. Several antimicrobials have been found to inhibit the synthesis of this lipid, and it is expected that more will be developed. In addition, antimicrobial peptides can bind to the outer membrane of Gram-negative bacteria and block passage of solutes between the periplasm and the cell exterior, resulting in bacterial toxicity. In Gram-positive bacteria, the major bacterial lipid component, phosphatidylglycerol can be chemically modified by bacterial enzymes to convert the lipid from anionic to cationic or zwitterionic form. This process leads to increased levels of resistance of the bacteria against polycationic antimicrobial agents. Inhibitors of this enzyme would provide protection against the development of bacterial resistance. There are antimicrobial agents that directly target a component of bacterial cytoplasmic membranes that can act on both Gram-negative as well as Gram-positive bacteria. Many of these are cyclic peptides with a rigid binding site capable of binding a lipid component. This binding targets antimicrobial agents to bacteria, rather than being toxic to host cells. This article is part of a Special Issue entitled: Antimicrobial peptides edited by Karl Lohner and Kai Hilpert.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cell Membrane/drug effects , Lipid A/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Cardiolipins/chemistry , Cardiolipins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/chemistry , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/metabolism , Lipid A/chemistry , Lipid A/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Molecular Targeted Therapy , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Species Specificity
6.
Mar Drugs ; 12(7): 4260-73, 2014 Jul 23.
Article in English | MEDLINE | ID: mdl-25056632

ABSTRACT

LPS molecules of marine bacteria show structures distinct from terrestrial bacteria, due to the different environment that marine bacteria live in. Because of these different structures, lipid A molecules from marine bacteria are most often poor stimulators of the Toll-like receptor 4 (TLR4) pathway. Due to their low stimulatory potential, these lipid A molecules are suggested to be applicable as antagonists of TLR4 signaling in sepsis patients, where this immune response is amplified and unregulated. Antagonizing lipid A molecules might be used for future therapies against sepsis, therapies that currently do not exist. In this review, we will discuss these differences in lipid A structures and their recognition by the immune system. The modifications present in marine lipid A structures are described, and their potential as LPS antagonists will be discussed. Finally, since clinical trials built on antagonizing lipid A molecules have proven unsuccessful, we propose to also focus on different aspects of the TLR4 signaling pathway when searching for new potential drugs. Furthermore, we put forward the notion that bacteria probably already produce inhibitors of TLR4 signaling, making these bacterial products interesting molecules to investigate for future sepsis therapies.


Subject(s)
Lipid A/antagonists & inhibitors , Sepsis/drug therapy , Toll-Like Receptor 4/antagonists & inhibitors , Acylation , Humans , Lipid A/chemistry , Lipid A/immunology , Protein Multimerization , Signal Transduction/drug effects , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/physiology , Water Microbiology
7.
Bioorg Med Chem Lett ; 23(8): 2362-7, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23499237

ABSTRACT

Lipopolysaccharide (LPS) biosynthesis is an attractive antibacterial target as it is both conserved and essential for the survival of key pathogenic bacteria. Lipid A is the hydrophobic anchor for LPS and a key structural component of the outer membrane of Gram-negative bacteria. Lipid A biosynthesis is performed in part by a unique zinc dependent metalloamidase, LpxC (UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase), which catalyzes the first non-reversible step in lipid A biosynthesis. The UDP portion of the LpxC substrate-binding pocket has been relatively unexplored. We have designed and evaluated a series of hydroxamate based inhibitors which explore the SAR of substitutions directed into the UDP pocket with a range of substituted α-amino acid based linkers. We also provide the first wild type structure of Pseudomonas aeruginosa LpxC which was utilized in the design of many of these analogs.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/chemistry , Amino Acids, Cyclic/chemistry , Uridine Diphosphate/chemistry , Amidohydrolases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Binding Sites , Drug Design , Hydrophobic and Hydrophilic Interactions , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Lipid A/antagonists & inhibitors , Lipid A/biosynthesis , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/biosynthesis , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Uridine Diphosphate/metabolism
8.
ACS Chem Biol ; 7(7): 1170-7, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22530734

ABSTRACT

UDP-3-O-(R-3-hydroxyacyl)GlcN N-acyltransferase (LpxD) has been shown to be essential to survival of lipid A producing Gram-negative bacteria. In this study, LpxD-binding peptides 12 amino acids in length were identified from a phage-bound random peptide library screen. Three peptides displayed antibacterial activity when expressed intracellularly, one of which (RJPXD33) represented 15% of the total hits. RJPXD33 binds to E. coli LpxD with a K(d) of 6 µM and is competitive with R-3-hydroxymyristoyl-ACP binding. RJPXD33 can be C-terminally fused in vivo with thioredoxin or N-terminally modified in vitro with ß-alanyl-fluorescein and maintain LpxD binding. The latter was used to develop an LpxD fluorescent binding assay used to evaluate unlabeled ligands and is amenable to small molecule library screening. Furthermore, RJPXD33 also binds to and inhibits E. coli UDP-N-acetylglucosamine acyltransferase (LpxA) with a K(d) of 20 µM, unearthing the possibility for the development of small molecule, dual-binding LpxA/LpxD inhibitors as novel antimicrobials.


Subject(s)
Acyltransferases/antagonists & inhibitors , Anti-Bacterial Agents/administration & dosage , Drug Delivery Systems/methods , Lipid A/antagonists & inhibitors , Lipid A/biosynthesis , Acyltransferases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/metabolism , Binding Sites/physiology , Molecular Sequence Data , Time Factors
9.
Innate Immun ; 18(1): 171-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21382909

ABSTRACT

Lipid A, the active moiety of LPS, exerts its effects through interaction with TLR4, triggering a signalling cascade that results in the release of pro-inflammatory cytokines. Eritoran is a lipid A analogue that competes with LPS for binding to TLR4; however, after intravenous administration, it undergoes a time-dependent deactivation as a consequence of binding to high-density lipoproteins (HDLs). The site of eritoran association with HDL remains unknown. Therefore the aim of this study was to determine if HDL-associated apolipoproteins A1, A2, serum amyloid A (SAA) and C1, inhibit the ability of eritoran to block LPS-induced TNF-α release from whole blood. Eritoran activity after LPS stimulation in human whole blood was assessed in the presence of reconstituted HDL (rHDL) containing different apos. In rHDL, the major apolipoproteins in both the healthy and septic state, A1 and SAA, caused a significant reduction in eritoran antagonistic activity and had a greater effect than minor apolipoproteins A2 and C1. Apolipoproteins associated with HDL are likely to facilitate eritoran deactivation. Apolipoproteins A1 and SAA should be of particular focus as they are the major apos found on HDL in both the healthy and septic state. Further evaluation of the physical association between apolipoproteins and eritoran should be explored.


Subject(s)
Blood Cells/drug effects , Disaccharides/antagonists & inhibitors , Lipid A/antagonists & inhibitors , Lipoproteins, HDL/immunology , Sugar Phosphates/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/chemistry , Apolipoprotein A-II/metabolism , Binding, Competitive , Blood Cells/immunology , Blood Cells/metabolism , Blood Cells/pathology , Disaccharides/pharmacology , Humans , Molecular Structure , Serum Amyloid A Protein/metabolism , Sugar Phosphates/pharmacology , Tumor Necrosis Factor-alpha/metabolism
10.
Biopolymers ; 92(6): 508-17, 2009.
Article in English | MEDLINE | ID: mdl-19637366

ABSTRACT

LPS release from Gram-negative bacteria can result in sepsis, a serious systemic inflammatory response to infection that can lead to septic shock and multiple organ failure. Thus, easy-to-synthesize, effective, and safe LPS-inhibitors are required to develop new agents for the treatment of sepsis. On the basis of the chemical features of the toxic part of LPS, lipid A, here we present peptide-based LPS-neutralizers that can be readily obtained using solid-phase methodologies. The presence of PEG-like moieties yielded the most active compounds, thereby indicating that these functionalities may be of great value in the design of new inhibitors. In this regard, the substitution of several amino acids by PEG-like chains in a previously reported cyclic anti-LPS peptide (the peptide RLKWc) rendered a new derivative that retained the activity of the original peptide. We foresee that this strategy could be successfully applied to other LPS-neutralizing peptides.


Subject(s)
Lipid A/antagonists & inhibitors , Oligopeptides/chemistry , Oligopeptides/chemical synthesis , Polyethylene Glycols , Protein Structure, Secondary
11.
J Endod ; 35(5): 668-72, 2009 May.
Article in English | MEDLINE | ID: mdl-19410080

ABSTRACT

INTRODUCTION: It is important to develop an antimicrobial agent without any damage on dental pulp. In the present study, we examined whether pretreatment of bacterial lipopolysaccharides (LPS) with ozonated water (O(3)aq) improves LPS-induced responses of rat odontoblastic cell line, KN-3. METHODS: After the pretreatment of LPS with O(3)aq, effects of LPS and O(3)aq-treated LPS on cell viability; calcification ability; expression of cyclooxygenase 2 (COX-2), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-alpha); and activation of p38 of KN-3 cells were examined. RESULTS: The formation of mineralized nodules by KN-3 cells was suppressed by LPS, whereas that suppression was inhibited by the pretreatment of LPS with ozonated water. We also found that LPS-induced expression of COX-2, IL-6, and TNF-alpha and p38 activation were markedly suppressed when LPS was pretreated with ozonated water. Furthermore, expression of COX-2, IL-6, and TNF-alpha by LPS were mainly induced through p38 activation. CONCLUSION: These results suggest that odontoblastic cells exhibit inflammatory responses against LPS and that ozonated water has the ability to improve LPS-induced inflammatory responses and suppression of odontoblastic properties of KN-3 cells through direct inhibition of LPS.


Subject(s)
Anti-Infective Agents/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Odontoblasts/drug effects , Oxidants, Photochemical/pharmacology , Ozone/pharmacology , Aggregatibacter actinomycetemcomitans , Animals , Butadienes/pharmacology , Calcification, Physiologic/drug effects , Cell Line , Cell Survival/drug effects , Cyclooxygenase 1/drug effects , Cyclooxygenase 2/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Escherichia coli , Imidazoles/pharmacology , Interleukin-6/analysis , Lipid A/antagonists & inhibitors , Membrane Proteins/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/drug effects , Nitriles/pharmacology , Phosphorylation/drug effects , Pyridines/pharmacology , Rats , Tumor Necrosis Factor-alpha/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/drug effects
12.
Mini Rev Med Chem ; 9(3): 306-17, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19275723

ABSTRACT

Lipopolysaccharide (LPS) or endotoxin, the major constituent of the outer membrane of Gram negative bacteria, has been implicated as the bacterial product responsible for the clinical syndrome of sepsis. LPS binding to the host receptor Toll-like receptor 4 (TLR4) triggers an inflammatory reaction characterised by the release of large number of inflammatory mediators that allow the host to respond to the invading pathogen. When this production becomes un-controlled and excessive, it leads to the development of septic shock. Despite decades of efforts in supporting therapies, sepsis remains the leading cause of death amongst critically ill patients. Unfortunately, the major factor contributing to the high morbidity and mortality of sepsis is the lack of the effective targeted treatment. Indeed, over 30 drugs for the treatment of sepsis have been developed: many of these target specific inflammatory mediators and have thus been, in general, unsuccessful since sepsis relies on the cross talk of several cytokines and the block of a single factor has been proven to be ineffective. More successful strategies include those modulating the early phase of LPS signalling such as the ones that prevent the binding of LPS to host cells and the subsequent cascade of detrimental events. In this light, effective LPS antagonists would represent invaluable tools to efficaciously manage sepsis. This review discusses the evolution of naturally occurring and synthetic LPS antagonists with emphasis on the development of several natural new molecules.


Subject(s)
Lipopolysaccharides/antagonists & inhibitors , Sepsis/drug therapy , Animals , Humans , Lipid A/analogs & derivatives , Lipid A/antagonists & inhibitors , Lipid A/pharmacology , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Molecular Structure , Signal Transduction/drug effects , Signal Transduction/physiology
13.
J Med Chem ; 52(4): 1209-13, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19161283

ABSTRACT

New glycolipids and a benzylammonium lipid were rationally designed by varying the chemical structure of a D-glucose-derived hit compound active as lipid A antagonist. We report the synthesis of these compounds, their in vitro activity as lipid A antagonists on HEK cells, and the capacity to inhibit LPS-induced septic shock in vivo. The lack of toxicity and the good in vivo activity suggest the use of some compounds of the panel as hits for antisepsis drug development.


Subject(s)
Anti-Infective Agents/chemical synthesis , Benzylammonium Compounds/chemical synthesis , Glycolipids/chemical synthesis , Lipids/chemical synthesis , Sepsis/drug therapy , Anti-Infective Agents/pharmacology , Benzylammonium Compounds/pharmacology , Cell Line , Drug Design , Glycolipids/pharmacology , Humans , Lipid A/antagonists & inhibitors , Lipids/pharmacology , Shock, Septic/drug therapy , Structure-Activity Relationship
14.
Biopolymers ; 91(3): 207-20, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19107926

ABSTRACT

The anti-lipopolysaccharide factor ALF-Pm3 is a 98-residue protein identified in hemocytes from the black tiger shrimp Penaeus monodon. It was expressed in Pichia pastoris from the constitutive glyceraldehyde-3-phosphate dehydrogenase promoter as a folded and (15)N uniformly labeled rALF-Pm3 protein. Its 3D structure was established by NMR and consists of three alpha-helices packed against a four-stranded beta-sheet. The C(34)-C(55) disulfide bond was shown to be essential for the structure stability. By using surface plasmon resonance, we demonstrated that rALF-Pm3 binds to LPS, lipid A and to OM-174, a soluble analogue of lipid A. Biophysical studies of rALF-Pm3/LPS and rALF-Pm3/OM-174 complexes indicated rather high molecular sized aggregates, which prevented us to experimentally determine by NMR the binding mode of these lipids to rALF-Pm3. However, on the basis of striking structural similarities to the FhuA/LPS complex, we designed an original model of the possible lipid A-binding site of ALF-Pm3. Such a binding site, located on the ALF-Pm3 beta-sheet and involving seven charged residues, is well conserved in ALF-L from Limulus polyphemus and in ALF-T from Tachypleus tridentatus. In addition, our model is in agreement with experiments showing that beta-hairpin synthetic peptides corresponding to ALF-L beta-sheet bind to LPS. Delineating lipid A-binding site of ALFs will help go further in the de novo design of new antibacterial or LPS-neutralizing drugs.


Subject(s)
Crustacea/chemistry , Invertebrate Hormones/chemistry , Invertebrate Hormones/metabolism , Lipid A/antagonists & inhibitors , Lipid A/metabolism , Models, Molecular , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Invertebrate Hormones/genetics , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Structural Homology, Protein , Surface Plasmon Resonance
15.
Microbes Infect ; 10(5): 571-4, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18403233

ABSTRACT

Lipopolysaccharides, the ubiquitous part of the outer membrane of Gram-negative bacteria, and their derivatives are recognised by plants to trigger or potentiate particular defence responses such as induction of genes encoding pathogenesis-related proteins. The molecular mechanisms of LPS perception that underpin these effects in plants are, however, unknown. Here, lipid A from Halomonas magadiensis, which is an antagonist of lipid A action in human cells, was used to investigate lipid A action in plants. Our findings offer an insight into the different structural requirements for direct induction and potentiation of plant defences by lipid A.


Subject(s)
Arabidopsis/microbiology , Gene Expression Regulation, Plant , Halomonas/chemistry , Lipid A/antagonists & inhibitors , Escherichia coli/chemistry , Escherichia coli Infections/microbiology , Gram-Negative Bacterial Infections/microbiology , Plant Leaves/microbiology , Plant Proteins/metabolism , RNA, Plant/analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Curr Pharm Biotechnol ; 9(1): 9-15, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18289052

ABSTRACT

Multi-drug resistant (MDR), pathogenic Gram-negative bacteria pose a serious health threat, and novel antibiotic targets must be identified to combat MDR infections. One promising target is the zinc-dependent metalloamidase UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC), which catalyzes the committed step of lipid A (endotoxin) biosynthesis. LpxC is an essential, single copy gene that is conserved in virtually all Gram-negative bacteria. LpxC structures, revealed by NMR and X-ray crystallography, demonstrate that LpxC adopts a novel 'beta-alpha-alpha-beta sandwich' fold and encapsulates the acyl chain of the substrate with a unique hydrophobic passage. Kinetic analysis revealed that LpxC functions by a general acid-base mechanism, with a glutamate serving as the general base. Many potent LpxC inhibitors have been identified, and most contain a hydroxamate group targeting the catalytic zinc ion. Although early LpxC-inhibitors were either narrow-spectrum antibiotics or broad-spectrum in vitro LpxC inhibitors with limited antibiotic properties, the recently discovered compound CHIR-090 is a powerful antibiotic that controls the growth of Escherichia coli and Pseudomonas aeruginosa, with an efficacy rivaling that of the FDA-approved antibiotic ciprofloxacin. CHIR-090 inhibits a wide range of LpxC enzymes with sub-nanomolar affinity in vitro, and is a two-step, slow, tight-binding inhibitor of Aquifex aeolicus and E. coli LpxC. The success of CHIR-090 suggests that potent LpxC-targeting antibiotics may be developed to control a broad range of Gram-negative bacteria.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/biosynthesis , Lipid A/biosynthesis , Zinc/metabolism , Animals , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/biosynthesis , Humans , Hydroxamic Acids/pharmacology , Lipid A/antagonists & inhibitors , Threonine/analogs & derivatives , Threonine/pharmacology , Zinc/antagonists & inhibitors
17.
Mol Microbiol ; 67(2): 264-77, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18047581

ABSTRACT

One-third of the lipid A found in the Escherichia coli outer membrane contains an unsubstituted diphosphate unit at position 1 (lipid A 1-diphosphate). We now report an inner membrane enzyme, LpxT (YeiU), which specifically transfers a phosphate group to lipid A, forming the 1-diphosphate species. (32)P-labelled lipid A obtained from lpxT mutants do not produce lipid A 1-diphosphate. In vitro assays with Kdo(2)-[4'-(32)P]lipid A as the acceptor shows that LpxT uses undecaprenyl pyrophosphate as the substrate donor. Inhibition of lipid A 1-diphosphate formation in wild-type bacteria was demonstrated by sequestering undecaprenyl pyrophosphate with the cyclic polypeptide antibiotic bacitracin, providing evidence that undecaprenyl pyrophosphate serves as the donor substrate within whole bacteria. LpxT-catalysed phosphorylation is dependent upon transport of lipid A across the inner membrane by MsbA, a lipid A flippase, indicating a periplasmic active site. In conclusion, we demonstrate a novel pathway in the periplasmic modification of lipid A that is directly linked to the synthesis of undecaprenyl phosphate, an essential carrier lipid required for the synthesis of various bacterial polymers, such as peptidoglycan.


Subject(s)
Lipid A/metabolism , Periplasm/enzymology , Polyisoprenyl Phosphates/biosynthesis , ATP-Binding Cassette Transporters/metabolism , Anti-Bacterial Agents/pharmacology , Bacitracin/pharmacology , Bacterial Proteins/metabolism , Escherichia coli K12/enzymology , Escherichia coli K12/genetics , Lipid A/antagonists & inhibitors , Membrane Lipids/metabolism , Mutation , Peptidyl Transferases/metabolism , Phosphates/metabolism , Phosphorylation/drug effects , Polyisoprenyl Phosphates/antagonists & inhibitors , Polyisoprenyl Phosphates/metabolism , Pyrophosphatases/genetics , Pyrophosphatases/metabolism
18.
J Immunol ; 179(6): 4083-92, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785847

ABSTRACT

Repeated exposure to low doses of endotoxin results in progressive hyporesponsiveness to subsequent endotoxin challenge, a phenomenon known as endotoxin tolerance. In spite of its clinical significance in sepsis and characterization of the TLR4 signaling pathway as the principal endotoxin detection mechanism, the molecular determinants that induce tolerance remain obscure. We investigated the role of the TRIF/IFN-beta pathway in TLR4-induced endotoxin tolerance. Lipid A-induced homotolerance was characterized by the down-regulation of MyD88-dependent proinflammatory cytokines TNF-alpha and CCL3, but up-regulation of TRIF-dependent cytokine IFN-beta. This correlated with a molecular phenotype of defective NF-kappaB activation but a functional TRIF-dependent STAT1 signaling. Tolerance-induced suppression of TNF-alpha and CCL3 expression was significantly relieved by TRIF and IFN regulatory factor 3 deficiency, suggesting the involvement of the TRIF pathway in tolerance. Alternatively, selective activation of TRIF by poly(I:C)-induced tolerance to lipid A. Furthermore, pretreatment with rIFN-beta also induced tolerance, whereas addition of IFN-beta-neutralizing Ab during the tolerization partially alleviated tolerance to lipid A but not TLR2-induced endotoxin homo- or heterotolerance. Furthermore, IFNAR1-/- murine embryonal fibroblast and bone-marrow derived macrophages failed to induce tolerance. Together, these observations constitute evidence for a role of the TRIF/IFN-beta pathway in the regulation of lipid A/TLR4-mediated endotoxin homotolerance.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Immune Tolerance , Lipid A/toxicity , Myeloid Differentiation Factor 88/physiology , Signal Transduction/immunology , Toll-Like Receptor 4/physiology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Cells, Cultured , Chemokines/biosynthesis , Chemokines/genetics , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/immunology , Immune Tolerance/genetics , Immunophenotyping , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/physiology , Interferon-beta/physiology , Ligands , Lipid A/antagonists & inhibitors , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , NF-kappa B/deficiency , NF-kappa B/physiology , Poly I-C/metabolism , Poly I-C/pharmacology , STAT1 Transcription Factor/physiology , Signal Transduction/genetics
19.
Bioorg Med Chem ; 15(14): 4800-12, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17513113

ABSTRACT

The lipid A from nitrogen-fixing bacterial species Rhizobium sin-1 is structurally unusual due to lack of phosphates and the presence of a 2-aminogluconolactone and a very long chain fatty acid, 27-hydroxyoctacosanoic acid (27OHC28:0), moiety. This structurally unusual lipid A can antagonize TNF-alpha production by human monocytes induced by Escherichia coli LPS. To establish the relevance of the unusual long chain 27-hydroxyoctacosanoic acid for antagonistic properties, a highly convergent strategy for the synthesis of several derivatives of the lipid A of R. sin-1 has been developed. Compound 1 is a natural R. sin-1 lipid A having a 27-hydroxyoctacosanoic acid at C-2', compound 2 contains an octacosanoic acid moiety at this position, and compound 3 is modified by a short chain tetradecanoic acid. Cellular activation studies with a human monocytic cell line have shown that the octacosanoic acid is important for optimal antagonistic properties. The hydroxyl of the natural 27-hydroxyoctacosanoic moiety does, however, not account for inhibitory activity. The resulting structure-activity relationships are important for the design of compounds for the treatment of septic shock.


Subject(s)
Fatty Acids/chemistry , Fatty Acids/pharmacology , Lipid A/antagonists & inhibitors , Rhizobium/metabolism , Cell Line , Fatty Acids/chemical synthesis , Humans , Lipid A/metabolism , Molecular Structure , Monocytes/drug effects , Monocytes/metabolism , Structure-Activity Relationship
20.
Chin J Traumatol ; 10(3): 180-3, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17535644

ABSTRACT

OBJECTIVE: To investigate the mechanism of polymyxin B (PMB) antagonizing the biological activity of lipopolysaccharide (LPS). METHODS: The affinity of PMB for LPS and lipid A was assayed by biosensor, and the neutralization of PMB for LPS (2 ng/ml) was detected by kinetic turbidimetric limulus test. The releases of TNF-alpha and IL-6 in murine peritoneal macrophages a (PMphi) after exposure to LPS (100 ng/ml) were detected, and the expression levels of TLR4, TNF-alpha and IL-6 mRNA in PMphi induced by LPS (100 ng/ml) were measured by RT-PCR. RESULTS: PMB had high-affinity to LPS and lipid A with dissociation equilibrium constants of 18.9 nmol/L and 11.1 nmol/L, respectively, and neutralized LPS in a dose-dependent manner. Furthermore, PMB could markedly inhibit the expressions of TLR4, TNF-alpha and IL-6 mRNA and the release of cycokines in LPS-stimulated murine peritoneal macrophages. CONCLUSIONS: PMB neutralizes LPS and inhibites the expression and release of cycokines in macrophages, in which the affinity of PMB for lipid A plays an important role.


Subject(s)
Lipopolysaccharides/antagonists & inhibitors , Polymyxin B/pharmacology , Animals , Cytokines/analysis , Limulus Test , Lipid A/antagonists & inhibitors , Macrophages/chemistry , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...