首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 223 毫秒
1.
Lipid A anchors the lipopolysaccharide (LPS) to the outer membrane and is usually composed of a hexa‐acylated diglucosamine backbone. Burkholderia cenocepacia, an opportunistic pathogen, produces a mixture of tetra‐ and penta‐acylated lipid A. “Late” acyltransferases add secondary acyl chains to lipid A after the incorporation of four primary acyl chains to the diglucosamine backbone. Here, we report that B. cenocepacia has only one late acyltransferase, LpxL (BCAL0508), which adds a myristoyl chain to the 2′ position of lipid A resulting in penta‐acylated lipid A. We also identified PagL (BCAL0788), which acts as an outer membrane lipase by removing the primary β ‐hydroxymyristate (3‐OH‐C14:0) chain at the 3 position, leading to tetra‐acylated lipid A. Unlike PagL, LpxL depletion caused reduced cell growth and defects in cell morphology, both of which were suppressed by overexpressing the LPS flippase MsbA (BCAL2408), suggesting that lipid A molecules lacking the fifth acyl chain contributed by LpxL are not good substrates for the flippase. We also show that intracellular B. cenocepacia within macrophages produced more penta‐acylated lipid A, suggesting lipid A penta‐acylation in B. cenocepacia is required not only for bacterial growth and morphology but also for adaptation to intracellular lifestyle.  相似文献   

2.
We previously described enrichment of conditional Escherichia coli msbA mutants defective in lipopolysaccharide export using Ludox density gradients (Doerrler WT (2007) Appl Environ Microbiol 73; 7992-7996). Here, we use this approach to isolate and characterize temperature-sensitive lpxL mutants. LpxL is a late acyltransferase of the pathway of lipid A biosynthesis (The Raetz Pathway). Sequencing the lpxL gene from the mutants revealed the presence of both missense and nonsense mutations. The missense mutations include several in close proximity to the enzyme's active site or conserved residues (E137K, H132Y, G168D). These data demonstrate that Ludox gradients can be used to efficiently isolate conditional E. coli mutants with defects in lipopolysaccharide biosynthesis and provide insight into the enzymatic mechanism of LpxL.  相似文献   

3.
Six DA  Carty SM  Guan Z  Raetz CR 《Biochemistry》2008,47(33):8623-8637
Escherichia coli lipid A is a hexaacylated disaccharide of glucosamine with secondary laurate and myristate chains on the distal unit. Hexaacylated lipid A is a potent agonist of human Toll-like receptor 4, whereas its tetra- and pentaacylated precursors are antagonists. The inner membrane enzyme LpxL transfers laurate from lauroyl-acyl carrier protein to the 2'- R-3-hydroxymyristate moiety of the tetraacylated lipid A precursor Kdo 2-lipid IV A. LpxL has now been overexpressed, solubilized with n-dodecyl beta- d-maltopyranoside (DDM), and purified to homogeneity. LpxL migration on a gel filtration column is consistent with a molecular mass of 80 kDa, suggestive of an LpxL monomer (36 kDa) embedded in a DDM micelle. Mass spectrometry showed that deformylated LpxL was the predominant species, noncovalently bound to as many as 12 DDM molecules. Purified LpxL catalyzed not only the formation in vitro of Kdo 2-(lauroyl)-lipid IV A but also a slow second acylation, generating Kdo 2-(dilauroyl)-lipid IV A. Consistent with the Kdo dependence of crude LpxL in membranes, Kdo 2-lipid IV A is preferred 6000-fold over lipid IV A by the pure enzyme. Sequence comparisons suggest that LpxL shares distant homology with the glycerol-3-phosphate acyltransferase (GPAT) family, including a putative catalytic dyad located in a conserved H(X) 4D/E motif. Mutation of H132 or E137 to alanine reduces specific activity by over 3 orders of magnitude. Like many GPATs, LpxL can also utilize acyl-CoA as an alternative acyl donor, albeit at a slower rate. Our results show that the acyltransferases that generate the secondary acyl chains of lipid A are members of the GPAT family and set the stage for structural studies.  相似文献   

4.
Similar to most Gram-negative bacteria, the outer leaflet of the outer membrane of Vibrio cholerae is comprised of lipopolysaccharide. Previous reports have proposed that V. cholerae serogroups O1 and O139 synthesize structurally different lipid A domains, which anchor lipopolysaccharide within the outer membrane. In the current study, intact lipid A species of V. cholerae O1 and O139 were analysed by mass spectrometry. We demonstrate that V. cholerae serogroups associated with human disease synthesize a similar asymmetrical hexa-acylated lipid A species, bearing a myristate (C14:0) and 3-hydroxylaurate (3-OH C12:0) at the 2'- and 3'-positions respectively. A previous report from our laboratory characterized the V. cholerae LpxL homologue Vc0213, which transfers a C14:0 to the 2'-position of the glucosamine disaccharide. Our current findings identify V. cholerae Vc0212 as a novel lipid A secondary hydroxy-acyltransferase, termed LpxN, responsible for transferring the 3-hydroxylaurate (3-OH C12:0) to the V. cholerae lipid A domain. Importantly, the presence of a 3-hydroxyl group on the 3'-linked secondary acyl chain was found to promote antimicrobial peptide resistance in V. cholerae; however, this functional group was not required for activation of the innate immune response.  相似文献   

5.
The synthesis of “typical” hexa-acylated lipid A occurs via a nine-step enzymatic pathway, which is generally well conserved throughout all gram-negative bacteria. One exception to the rule is Helicobacter pylori, which has only eight homologs to the nine lipid A biosynthetic enzymes. The discrepancy occurs toward the end of the pathway, with H. pylori containing only a single putative secondary acyltransferase encoded by jhp0265. In Escherichia coli K-12, two late acyltransferases, termed LpxL and LpxM, are required for the biosynthesis of hexa-acylated lipid A. Detailed biochemical and genetic analyses reveal that H. pylori Jhp0265 (the protein encoded by jhp0265) is in fact an LpxL homolog, capable of transferring a stearoyl group to the hydroxyl group of the 2′ linked fatty acyl chain of lipid A. Despite the lack of a homolog to LpxM in the H. pylori genome, the organism synthesizes a hexa-acylated lipid A species, suggesting that an equivalent enzyme exists. Using radiolabeled lipid A substrates and acyl-acyl carrier protein as the fatty acyl donor, we were able to confirm the presence of a second H. pylori late acyl transferase by biochemical assays. After synthesis of the hexa-acylated lipid A species, several modification enzymes then function to produce the major lipid A species of H. pylori that is tetra-acylated. Jhp0634 was identified as an outer membrane deacylase that removes the 3′-linked acyl chains of H. pylori lipid A. Together, this work elucidates the biochemical machinery required for the acylation and deacylation of the lipid A domain of H. pylori lipopolysaccharide.  相似文献   

6.
The lipopolysaccharide of Vibrio cholerae has been reported to contain a single 3-deoxy-d-manno-octulosonic acid (Kdo) residue that is phosphorylated. The phosphorylated Kdo sugar further links the hexa-acylated V. cholerae lipid A domain to the core oliogosaccharide and O-antigen. In this report, we confirm that V. cholerae possesses the enzymatic machinery to synthesize a phosphorylated Kdo residue. Further, we have determined that the presence of the phosphate group on the Kdo residue is necessary for secondary acylation in V. cholerae. The requirement for a secondary substituent on the Kdo residue (either an additional Kdo sugar or a phosphate group) was also found to be critical for secondary acylation catalyzed by LpxL proteins from Bordetella pertussis, Escherichia coli, and Haemophilus influenzae. Although three putative late acyltransferase orthologs have been identified in the V. cholerae genome (Vc0212, Vc0213, and Vc1577), only Vc0213 appears to be functional. Vc0213 functions as a myristoyl transferase acylating lipid A at the 2′-position of the glucosamine disaccharide. Generally acyl-ACPs serve as fatty acyl donors for the acyltransferases required for lipopolysaccharide biosynthesis; however, in vitro assays indicate that Vc0213 preferentially utilizes myristoyl-CoA as an acyl donor. This is the first report to biochemically characterize enzymes involved in the biosynthesis of the V. cholerae Kdo-lipid A domain.Lipopolysaccharide (LPS),2 the major surface molecule in the outer membrane of Gram-negative bacteria, is composed of three domains: lipid A, core oligosaccharide, and O-antigen (1). The core oligosaccharide is further divided into two distinct regions: inner and outer core. The inner core consists of the Kdo sugars, which are responsible for linking the core region to the lipid A moiety of LPS. Lipid A is the hydrophobic anchor of LPS and is the only portion of LPS required for activating the host innate immune response by interacting with Toll-like receptor 4 and the accessory molecule, MD2.Kdo-lipid A biosynthesis is a well conserved and ordered process among Gram-negative bacteria; however, not all Gram-negative bacteria produce similar lipid A structures (2). In Escherichia coli, the biosynthesis of the Kdo-lipid A domain occurs via a nine-step process, resulting in the production of a hexa-acylated lipid A structure known as Kdo2-lipid A. Kdo2-lipid A has long been thought to be essential for the viability of E. coli; however, viable suppressor strains have been isolated that lack the Kdo sugar (3). The late steps of the biosynthetic pathway involve the addition of the Kdo sugars and the secondary or “late” acyl chains. The enzyme responsible for the addition of the Kdo sugars is the Kdo transferase (WaaA). In E. coli, this enzyme is bifunctional, transferring two Kdo sugars to the lipid A precursor, lipid IVA (4); however, other Gram-negative bacteria have been shown to possess a monofunctional or trifunctional WaaA, as is the case in Haemophilus influenzae (5) or Chlamydia trachomatis (6), respectively.Previous reports have shown that in E. coli, the addition of the Kdo sugars is critical for the functionality of the secondary acyltransferases (LpxL, LpxM, and LpxP). The E. coli late acyltransferase LpxL catalyzes the transfer of laurate (C12:0) to the acyl chain linked at the 2′-position of Kdo2-lipid IVA (7). LpxM then catalyzes the addition of a myristate (C14:0) to the 3′-linked acyl chain of the penta-acylated lipid A precursor (8). When E. coli experience cold shock conditions (temperatures below 20 °C), the late acyltransferase LpxP transfers a palmitoleate (C16:1) to the 2′-position of Kdo2-lipid IVA, replacing the C12:0 acyl chain transferred by LpxL (9). Lipid A secondary acyltransferases have been shown to primarily utilize acyl-acyl carrier proteins (acyl-ACPs) as their acyl chain donor; however, a recent report by Six et al. (10) has shown that purified E. coli LpxL is capable of utilizing acyl-coenzyme A (acyl-CoA) as an alternative acyl donor at a lesser rate.The Gram-negative bacteria Vibrio cholerae is the causative agent of the severe diarrheal disease cholera. Cholera is transmitted via the fecal-oral route by ingestion of contaminated drinking water or food. The World Health Organization reported ∼130,000 cases of cholera in 2005 with the majority occurring in Africa. There are two serogroups of V. cholerae capable of epidemic and pandemic disease: O1 and O139 (11). Previous structural analyses have revealed that these serogroups possess very different lipid A structures. The V. cholerae O1 lipid A structure was reported as hexa-acylated, bearing secondary acyl chains at the 2- and 2′-positions of phosphorylated Kdo-lipid A (1113); however, V. cholerae O139 was reported as having an octa-acylated lipid A (see Fig. 1) (11, 14).Open in a separate windowFIGURE 1.Comparison of E. coli K12 lipid A species to V. cholerae O1 and V. cholerae O139 lipid A species. The covalent modifications of lipid A are indicated with dashed bonds, and the lengths of the acyl chains are indicated below each structure. The lipid A of E. coli K12 is a hexa-acylated structure, bearing two secondary acyl chains at the 2′- and 3′-positions. The E. coli lipid A structure is glycosylated at the 6′-position with two Kdo moieties and is phosphorylated at the 1- and 4′-positions of the disaccharide backbone. Similar to E. coli, the lipid A species of V. cholerae serogroup O1 is hexa-acylated, but with a symmetrical acyl chain distribution. The proposed lipid A structure of V. cholerae O139 is the octa-acylated structure. Both V. cholerae serogroups O1 and O139 reported lipid A species have a single Kdo sugar that is phosphorylated (red) and a phosphoethanolamine (magenta) attached to the 1-phosphate.Our report focuses on V. cholerae O1 El Tor, which is the predominant disease-causing strain worldwide. Because little attention has been given to the Kdo-lipid A domain of V. cholerae, we investigated the assembly of the inner core structure of V. cholerae O1 LPS and the late acylation steps. This report demonstrates the importance of a secondary negative charge on the primary Kdo sugar of lipid A for late acyltransferase activity in V. cholerae and in other Gram-negative bacteria. Also, we have identified the putative V. cholerae late acyltransferase, Vc0213 as the LpxL homolog, transferring a myristate (C14:0) to the 2′-position of V. cholerae lipid A. These initial findings provide us with the groundwork needed to investigate the modifications of the V. cholerae Kdo-lipid A structure, which may serve as attractive vaccine targets in future research.  相似文献   

7.
Most Gram‐negative organisms produce lipopolysaccharide (LPS), a complex macromolecule anchored to the bacterial membrane by the lipid A moiety. Lipid A is synthesized via the Raetz pathway, a conserved nine‐step enzymatic process first characterized in Escherichia coli. The Epsilonproteobacterium Helicobacter pylori uses the Raetz pathway to synthesize lipid A; however, only eight of nine enzymes in the pathway have been identified in this organism. Here, we identify the missing acyltransferase, Jhp0255, which transfers a secondary acyl chain to the 3′‐linked primary acyl chain of lipid A, an activity similar to that of E. coli LpxM. This enzyme, reannotated as LpxJ due to limited sequence similarity with LpxM, catalyses addition of a C12:0 or C14:0 acyl chain to the 3′‐linked primary acyl chain of lipid A, complementing an E. coli LpxM mutant. Enzymatic assays demonstrate that LpxJ and homologues in Campylobacter jejuni and Wolinella succinogenes can act before the 2′ secondary acyltransferase, LpxL, as well as the 3‐deoxy‐d ‐manno‐octulosonic acid (Kdo) transferase, KdtA. Ultimately, LpxJ is one member of a large class of acyltransferases found in a diverse range of organisms that lack an E. coli LpxM homologue, suggesting that LpxJ participates in lipid A biosynthesis in place of an LpxM homologue.  相似文献   

8.
The chemical structures of six lipid A species (A, B, C, D-1, D-2, and E) purified from Rhizobium etli CE3 were investigated by one- and two-dimensional NMR spectroscopy. The R. etli lipid A subtypes each contain an unusual acyloxyacyl residue at position 2' as part of a conserved distal glucosamine moiety but differ in their proximal units. All R. etli lipid A species lack phosphate groups. However, they are derivatized with an alpha-linked galacturonic acid group at position 4', as shown by nuclear Overhauser effect spectroscopy. Component B, which had been not been reported in previous studies, features a beta, 1'-6 linked disaccharide of glucosamine acylated at positions 2, 3, 2', and 3' in a pattern that is typical of lipid A found in other Gram-negative bacteria. D-1 contains an acylated aminogluconate unit in place of the proximal glucosamine residue of B. C and E lack ester-linked beta-hydroxyacyl chains at position 3, as judged by their H-3 chemical shifts, and may be synthesized from B and D-1, respectively, by the R. etli 3-O-deacylase. D-2 is an isomer of D-1 that forms nonenzymatically by acyl chain migration. A may be an elimination product derived from D-1 during hydrolysis at 100 degrees C (pH 4.5), a step needed to release lipid A from lipopolysaccharide. Based on these findings, we propose a biosynthetic scheme for R. etli lipid A in which B is generated first by a variation of the E. coli pathway. The aminogluconate unit of D-1 could then be made from B by enzymatic oxidation of the proximal glucosamine. As predicted by our hypothesis, enzyme(s) can be demonstrated in extracts of R. etli that convert (14)C-labeled B to D-1.  相似文献   

9.
The facultative intracellular pathogen Bartonella henselae is responsible for a broad range of clinical manifestations, including the formation of vascular tumors as a result of increased proliferation and survival of colonized endothelial cells. This remarkable interaction with endotoxin-sensitive endothelial cells and the apparent lack of septic shock are considered to be due to a reduced endotoxic activity of the B. henselae lipopolysaccharide. Here, we show that B. henselae ATCC 49882(T) produces a deep-rough-type lipopolysaccharide devoid of O-chain and report on its complete structure and Toll-like receptor-dependent biological activity. The major short-chain lipopolysaccharide was studied by chemical analyses, electrospray ionization, and matrix-assisted laser desorption/ionization mass spectrometry, as well as by NMR spectroscopy after alkaline deacylation. The carbohydrate portion of the lipopolysaccharide consists of a branched trisaccharide containing a glucose residue attached to position 5 of an alpha-(2-->4)-linked 3-deoxy-d-manno-oct-2-ulosonic acid disaccharide. Lipid A is a pentaacylated beta-(1'-->6)-linked 2,3-diamino-2,3-dideoxy-glucose disaccharide 1,4'-bisphosphate with two amide-linked residues each of 3-hydroxydodecanoic and 3-hydroxyhexadecanoic acids and one residue of either 25-hydroxyhexacosanoic or 27-hydroxyoctacosanoic acid that is O-linked to the acyl group at position 2'. The lipopolysaccharide studied activated Toll-like receptor 4 signaling only to a low extent (1,000-10,000-fold lower compared with that of Salmonella enterica sv. Friedenau) and did not activate Toll-like receptor 2. Some unusual structural features of the B. henselae lipopolysaccharide, including the presence of a long-chain fatty acid, which are shared by the lipopolysaccharides of other bacteria causing chronic intracellular infections (e.g. Legionella and Chlamydia), may provide the molecular basis for low endotoxic potency.  相似文献   

10.
Gao S  Peng D  Zhang W  Muszyński A  Carlson RW  Gu XX 《The FEBS journal》2008,275(20):5201-5214
Lipid A is a biological component of the lipo-oligosaccharide of a human pathogen, Moraxella catarrhalis. No other acyltransferases except for UDP-GlcNAc acyltransferase, responsible for lipid A biosynthesis in M. catarrhalis, have been identified. By bioinformatics, two late acyltransferase genes, lpxX and lpxL, responsible for lipid A biosynthesis were identified, and knockout mutants of each gene in M. catarrhalis strain O35E were constructed and named O35ElpxX and O35ElpxL. Structural analysis of lipid A from the parental strain and derived mutants showed that O35ElpxX lacked two decanoic acids (C10:0), whereas O35ElpxL lacked one dodecanoic (lauric) acid (C12:0), suggesting that lpxX encoded decanoyl transferase and lpxL encoded dodecanoyl transferase. Phenotypic analysis revealed that both mutants were similar to the parental strain in their toxicity in vitro. However, O35ElpxX was sensitive to the bactericidal activity of normal human serum and hydrophobic reagents. It had a reduced growth rate in broth and an accelerated bacterial clearance at 3 h (P < 0.01) or 6 h (P < 0.05) after an aerosol challenge in a murine model of bacterial pulmonary clearance. O35ElpxL presented similar patterns to those of the parental strain, except that it was slightly sensitive to the hydrophobic reagents. These results indicate that these two genes, particularly lpxX, encoding late acyltransferases responsible for incorporation of the acyloxyacyl-linked secondary acyl chains into lipid A, are important for the biological activities of M. catarrhalis.  相似文献   

11.
Wang Z  Li J  Altman E 《Carbohydrate research》2006,341(17):2816-2825
The lipid A components of Aeromonas salmonicida subsp. salmonicida from strains A449, 80204-1 and an in vivo rough isolate were isolated by mild acid hydrolysis of the lipopolysaccharide. Structural studies carried out by a combination of fatty acid, electrospray ionization-mass spectrometry and nuclear magnetic resonance analyses confirmed that the structure of lipid A was conserved among different isolates of A. salmonicida subsp. salmonicida. All analyzed strains contained three major lipid A molecules differing in acylation patterns corresponding to tetra-, penta- and hexaacylated lipid A species and comprising 4'-monophosphorylated beta-2-amino-2-deoxy-d-glucopyranose-(1-->6)-2-amino-2-deoxy-d-glucopyranose disaccharide, where the reducing end 2-amino-2-deoxy-d-glucose was present primarily in the alpha-pyranose form. Electrospray ionization-tandem mass spectrometry fragment pattern analysis, including investigation of the inner-ring fragmentation, allowed the localization of fatty acyl residues on the disaccharide backbone of lipid A. The tetraacylated lipid A structure containing 3-(dodecanoyloxy)tetradecanoic acid at N-2',3-hydroxytetradecanoic acid at N-2 and 3-hydroxytetradecanoic acid at O-3, respectively, was found. The pentaacyl lipid A molecule had a similar fatty acid distribution pattern and, additionally, carried 3-hydroxytetradecanoic acid at O-3'. In the hexaacylated lipid A structure, 3-hydroxytetradecanoic acid at O-3' was esterified with a secondary 9-hexadecenoic acid. Interestingly, lipid A of the in vivo rough isolate contained predominantly tetra- and pentaacylated lipid A species suggesting that the presence of the hexaacyl lipid A was associated with the smooth-form lipopolysaccharide.  相似文献   

12.
To elucidate the minimal lipopolysaccharide (LPS) structure needed for the viability of Escherichia coli, suppressor-free strains lacking either the 3-deoxy-d-manno-oct-2-ulosonic acid transferase waaA gene or derivatives of the heptosyltransferase I waaC deletion with lack of one or all late acyltransferases (lpxL/M/P) and/or various outer membrane biogenesis factors were constructed. Δ(waaC lpxL lpxM lpxP) and waaA mutants exhibited highly attenuated growth, whereas simultaneous deletion of waaC and surA was lethal. Analyses of LPS of suppressor-free waaA mutants grown at 21 °C, besides showing accumulation of free lipid IVA precursor, also revealed the presence of its pentaacylated and hexaacylated derivatives, indicating in vivo late acylation can occur without Kdo. In contrast, LPS of Δ(waaC lpxL lpxM lpxP) strains showed primarily Kdo2-lipid IVA, indicating that these minimal LPS structures are sufficient to support growth of E. coli under slow-growth conditions at 21/23 °C. These lipid IVA derivatives could be modified biosynthetically by phosphoethanolamine, but not by 4-amino-4-deoxy-l-arabinose, indicating export defects of such minimal LPS. ΔwaaA and Δ(waaC lpxL lpxM lpxP) exhibited cell-division defects with a decrease in the levels of FtsZ and OMP-folding factor PpiD. These mutations led to strong constitutive additive induction of envelope responsive CpxR/A and σE signal transduction pathways. Δ(lpxL lpxM lpxP) mutant, with intact waaC, synthesized tetraacylated lipid A and constitutively incorporated a third Kdo in growth medium inducing synthesis of P-EtN and l-Ara4N. Overexpression of msbA restored growth of Δ(lpxL lpxM lpxP) under fast-growing conditions, but only partially that of the Δ(waaC lpxL lpxM lpxP) mutant. This suppression could be alleviated by overexpression of certain mutant msbA alleles or the single-copy chromosomal MsbA-498V variant in the vicinity of Walker-box II.Lipopolysacharides (LPS)4 are the major amphiphilic constituents of the outer leaflet of the outer membrane (OM) of Gram-negative bacteria, including Escherichia coli. LPS share a common architecture composed of a membrane-anchored phosphorylated and acylated β(1→6)-linked GlcN disaccharide, termed lipid A, to which a carbohydrate moiety of varying size is attached (1, 2). The latter may be divided into a lipid A proximal core oligosaccharide and, in smooth-type bacteria, a distal O-antigen. LPS always contain 3-deoxy-α-d-manno-oct-2-ulosonic acid (Kdo) linked to the lipid A.The physiological importance of the Kdo/lipid A region is reflected by its specific position within the pathway of LPS biosynthesis. In E. coli K-12, a bisphosphorylated lipid A precursor molecule with two amide and two ester-bound (R)-3-hydroxymyristate residues (lipid IVA) is synthesized from UDP-GlcNAc, following 6 distinct enzyme reactions (1). This intermediate serves as an acceptor for the Kdo transferase (WaaA), which transfers two Kdo residues from CMP-Kdo to yield an α(2→4)-linked Kdo disaccharide-attached α(2→6) to the non-reducing GlcN residue of lipid IVA (3). The latter reaction product, termed Kdo2-lipid IVA, comprises a key intermediate of LPS biosynthesis that acts 2-fold as a specific substrate: (i) for glycosyltransferases catalyzing further steps of the core oligosaccharide biosynthesis (4) and (ii) for acyltransferases that complete the lipid A moiety by the transfer of 2 additional fatty acids to the (R)-3-hydroxyl groups of both acyl chains, which are directly bound to position 2′ and 3′ of the non-reducing GlcN residue (1). Three acyltransferases, encoded by paralogous genes, have been described in E. coli K-12, which catalyze the latter enzyme reactions using acyl carrier protein-activated fatty acids as co-substrates (510). At ambient temperatures, a lauroyl residue is first transferred by LpxL (6) to the OH group of the amide-bound (R)-3-hydroxymyristate residue at position 2′. This catalytic step is partially replaced at low temperature (12 °C) by LpxP, which transfers palmitoleate to the same position in ∼80% of the LPS molecules (7). The free OH group of the ester-bound (R)-3-hydroxymyristate residue at position 3′ within both pentaacylated intermediates is then myristoylated by LpxM to give a hexaacylated lipid A moiety (Fig. 3) (5).Open in a separate windowFIGURE 3.Chemical structure of tetraacylated lipid IVA precursor (A) and Kdo2-lipid IVA (B). R1 represents C12:0 or C16:1; R2, C14:0; R3 and R4 are under LPS-modifying conditions P-EtN and l-Ara4N, respectively, and R5, C16:0.Consistent with the essentiality of LPS in E. coli, all the genes, whose products are required for committed steps of biosynthesis of lipid IVA and subsequent transfer of Kdo to it, are essential (1, 2). However, individually neither the subsequent steps of addition of the secondary lauroyl and myristoyl residues to the distal glucosoamine unit by LpxL and LpxM to synthesize hexaacylated lipid A nor the later glycosylation of hexaacylated Kdo2-lipid A is essential for viability of bacteria like E. coli K-12 under defined growth conditions (8). Although Re mutants that possess LPS with only hexaacylated Kdo2-lipid A or mutants that synthesize complete LPS core with only lipid IVA are viable, they are impaired in several growth properties, including constitutive induction of RpoE signal transduction in Re mutants (8, 1113). A triple null mutant, which lacks all 3 late acyltransferases, is viable but only in slow-growth conditions in accordance with lipid IVA being a poor substrate of the lipid A transporter MsbA (8). Mutants impaired in the synthesis of Kdo, which synthesize only lipid IVA lacking any glycosylation, can be constructed, but they require additional suppressor mutations either in msbA, or the yhjD gene (14, 15). Strains that potentially can only synthesize Kdo2-lipid IVA have not been reported up to now. Thus, suppressor-free minimal LPS structures that can support growth of E. coli K-12 bacteria known up to now have genetic compositions of Δ(lpxL lpxM lpxP) or Re mutants.We describe the construction and characterization of suppressor-free ΔwaaA and Δ(waaC lpxL lpxM lpxP) mutants, synthesizing either free lipid IVA derivatives or Kdo2-lipid IVA LPS, respectively. Analyses of lipid A of ΔwaaA also revealed the presence of free penta- and hexaacylated lipid A derivatives, arising due to incorporation of secondary acyl chains. Such suppressor-free strains could be constructed only in slow-growth conditions at lower temperatures. Growth of Δ(waaC lpxL lpxM lpxP) could be restored by extragenic chromosomal MsbA-D498V suppressor mutation or by the overexpression of the msbA wild-type gene product. The LPS of Δ(waaC lpxL lpxM lpxP) and lipid IVA precursor of ΔwaaA was found to be substituted by P-EtN, but not l-Ara4N, under LPS-modifying growth conditions. Deletion of late acyltransferases in ΔwaaC or deletion of the waaA gene resulted in constitutively elevated levels of periplasmic protease HtrA, due to additive induction of the envelope stress responsive CpxR/A two-component system and σE pathway.  相似文献   

13.
The chemical structure of lipid A isolated from Porphyromonas gingivalis lipopolysaccharide was elucidated by compositional analysis, mass spectrometry, and nuclear magnetic resonance spectroscopy. The hydrophilic backbone of free lipid A was found to consisted of beta(1,6)-linked D-glucosamine disaccharide 1-phosphate. (R)-3-Hydroxy-15-methylhexadecanoic acid and (R)-3-hydroxyhexadecanoic acid are attached at positions 2 and 3 of the reducing terminal residue, respectively, and positions 2' and 3' of the nonreducing terminal unit are acylated with (R)-3-O-(hexadecanoyl)-15-methylhexadecanoic acid and (R)-3-hydroxy-13-methyltetradecanoic acid, respectively. The hydroxyl group at position 4' is partially replaced by another phosphate group, and the hydroxyl groups at positions 4 and 6' are unsubstituted. Considerable heterogeneity in the fatty acid chain length and the degree of acylation and phosphorylation was detected by liquid secondary ion-mass spectrometry (LSI-MS). A significant pseudomolecular ion of lipid A at m/z 1,769.6 [M-H]- corresponding to a diphosphorylated GlcN backbone bearing five acyl groups described above was detected in the negative mode of LSI-MS. Predominant ions, however, were observed at m/z 1,434.9 [M-H]- and m/z 1,449.0 [M-H]-, each representing monophosphoryl lipid A lacking (R)-3-hydroxyhexadecanoic and (R)-3-hydroxy-13-methyltetradecanoic acids, respectively. The presence of mono- and diphosphorylated lipid A species was also confirmed by LSI-MS of de-O-acylated lipid A (m/z 955.3 and 1,035.2, respectively).  相似文献   

14.
Lipopolysaccharide is one of the major constituents of the Gram-negative bacterial outer membrane and is, due to its endotoxic activity, responsible for the relatively high reactogenicity of whole-cell vaccines. In addition, lipopolysaccharide has strong immune stimulating properties, which makes it, potentially, an interesting vaccine component. In a previous study, we have shown that expression of two lipopolysaccharide-modifying enzymes, i.e., PagP and PagL, modulates the endotoxic activity of the Gram-negative bacterium Bordetella pertussis, the causative agent of whooping cough. To assess the consequences of PagP and PagL expression on the efficacy and reactogenicity of whole-cell pertussis vaccines, we have immunised mice and challenged them intranasally with wild-type B. pertussis. Vaccine efficacy, B. pertussis-specific antibody responses, and cytokine profiles were evaluated. The results show that expression of PagL, but not of PagP, significantly increases vaccine efficacy without altering vaccine reactogenicity. Therefore, PagL-expressing B. pertussis strains may form a basis for the development of a new and safer whole-cell pertussis vaccine, as higher vaccine efficacies may allow a reduced vaccine dosage. These data show, for the first time, that lipopolysaccharide composition is an important determinant for the efficacy of whole-cell pertussis vaccines.  相似文献   

15.
The chemical structure of the lipopolysaccharide of a deep-rough mutant (strain I-69 Rd-/b+) of Haemophilus influenzae was investigated. The hydrophilic backbone of lipid A was shown to consist of a beta-(1',6)-linked D-glucosamine disaccharide with phosphate groups at C-1 of the reducing D-glucosamine and at C-4' of the non-reducing one. Four molecules of (R)-3-hydroxytetradecanoic acid were found directly linked to the lipid A backbone, two by amide and two by ester linkage (positions 2,2' and 3,3', respectively). Laser-desorption mass spectrometry showed that both 3-hydroxytetradecanoic acids linked to the non-reducing glucosamine carry tetradecanoic acid at their 3-hydroxyl group, so that altogether six molecules of fatty acid are present in lipid A. The lipopolysaccharide was the first described to contain only one sugar unit linked to lipid A. This, sugar in accordance with a previous report [Zamze et al. (1987) Biochem. J. 245, 583-587], was shown to be a dOclA phosphate. The phosphate group was found at position 4, but the analytical procedures employed (permethylation and methanolysis followed by gas-liquid chromatography/mass spectrometry) also revealed dOclA 5-phosphate. Since a cyclic 4,5-phosphate could be ruled out by 31P-NMR, we conclude that, in this lipopolysaccharide, a mixture of dOclA 4- and 5-phosphate is present. By methylation analysis of the dephosphorylated, deacylated and reduced lipopolysaccharide the attachment site of the dOclA was assigned to position C-6' of the non-reducing glucosamine of lipid A. The anomeric linkages present in the lipopolysaccharide were assessed by 1H-NMR and 13C-NMR of deacylated lipopolysaccharide. The saccharide backbone of this Haemophilus influenzae lipopolysaccharide possesses the following structure: (Formula; see text)  相似文献   

16.
Lipid A isolated by mild acid hydrolysis from lipopolysaccharides of 22 nontypeable and 2 type f Haemophilus influenzae strains was investigated using electrospray ionization coupled to quadrupole ion trap mass spectrometry. The lengths, positions, and number of acyl chains in the lipid A molecule were determined using multiple-step tandem mass spectrometry (MSn). All of the analyzed strains showed a major lipid A molecule comprising beta-2-amino-2-deoxy-D-glucopyranose-(1-->6)-alpha-2-amino-2-deoxy-D-glucopyranose phosphorylated at the C4' and C1 positions. The C2/C2' and C3/C3' positions were substituted by amide-linked and ester-linked 3-hydroxytetradecanoic acid chains, respectively. The fatty acid chains on C3' and C2' were further esterified by tetradecanoic acid chains. In all strains, minor amounts of lipid A molecules with different acylation patterns were identified. Thus, structures comprising the hexaacylated lipid A with the C2 or C3 position being substituted by 3-hydroxydecanoic acid, and hexaacylated lipid A with the C3 and C3' positions being substituted by 3-hydroxydodecanoic or dodecanoyloxytetradecanoic acid, respectively, were found. In addition, lipid A with an acetyl group attached to the 3-hydroxytetradecanoic acid groups attached to the C2 or C3 position was detected in two nontypeable H. influenzae strains.  相似文献   

17.
The chemical structure of lipid A from the lipopolysaccharide of the mushroom-associated bacterium Pseudomonas reactans, a pathogen of cultivated mushroom, was elucidated by compositional analysis and spectroscopic methods (MALDI-TOF and two-dimensional NMR). The sugar backbone was composed of the beta-(1'-->6)-linked d-glucosamine disaccharide 1-phosphate. The lipid A fraction showed remarkable heterogeneity with respect to the fatty acid and phosphate composition. The major species are hexacylated and pentacylated lipid A, bearing the (R)-3-hydroxydodecanoic acid [C12:0 (3OH)] in amide linkage and a (R)-3-hydroxydecanoic [C10:0 (3OH)] in ester linkage while the secondary fatty acids are present as C12:0 and/or C12:0 (2-OH). A nonstoichiometric phosphate substitution at position C-4' of the distal 2-deoxy-2-amino-glucose was detected. Interestingly, the pentacyl lipid A is lacking a primary fatty acid, namely the C10:0 (3-OH) at position C-3'. The potential biological meaning of this peculiar lipid A is also discussed.  相似文献   

18.
The structure of the lipid A component of lipopolysaccharides isolated from two wild-type strains (Fisher 2 and 7) and one rough mutant (PAC 605) of Pseudomonas aeruginosa was investigated using chemical analysis, methylation analysis, combined gas-liquid chromatography/mass spectrometry, laser-desorption mass spectrometry and NMR spectroscopy. The lipid A backbone was found to consist of a pyranosidic beta 1,6-linked D-glucosamine disaccharide [beta-D-GlcpN-(1----6)-D-GlcpN], phosphorylated in positions 4' and 1. Position 6' of the beta-D-GlcpN-(1----6)-D-GlcpN disaccharide was identified as the attachment site of the core oligosaccharide and the hydroxyl group at C-4 was not substituted. Lipid A of the three P. aeruginosa strains expressed heterogeneity with regard to the degree of acylation: a hexaacyl as well as a pentaacyl component were structurally characterized. The hexaacyl lipid A contains two amide-bound 3-O-acylated (R)-3-hydroxydodecanoic acid groups [12:0(3-OH)] at positions 2 and 2' of the GlcN dissacharide and two ester-bound (R)-3-hydroxydecanoic acid groups [10:0(3-OH)] at positions 3 and 3'. The pentaacyl species, which represents the major lipid A component, lacks one 10:0(3-OH) residue, the hydroxyl group in position 3 of the reducing GlcN residue being free. In both hexa- and pentaacyl lipid A the 3-hydroxyl group of the two amide-linked 12:0(3-OH) residues are acylated by either dodecanoic (12:0) or (S)-2-hydroxydodecanoic acid [12:0(2-OH)], the lipid A species with two 12:0(2-OH) residues, however, being absent. The presence of only five acyl residues in the major lipid A fraction may account for the low endotoxic activity observed with P. aeruginosa lipopolysaccharide.  相似文献   

19.
Lipopolysaccharide is a major constituent of the outer membrane of Gram-negative bacteria and important in the induction of pro-inflammatory responses. Recently, novel LPS species derived from Neisseria meningitidis H44/76 by insertional inactivation of the lpxL1 and lpxL2 genes have been created with a lipid A portion consisting of five (penta-acylated lpxL1) or four (tetra-acylated lpxL2) fatty acids connected to the glucosamine backbone instead of six fatty acids in the wild-type LPS. We show that these mutant LPS-types are poor inducers of cytokines (tumor-necrosis factor-α, IL-1β, IL-10, IL-RA) in human mononuclear cells. Both penta- and tetra-acylated meningococcal LPSs were able to inhibit cytokine production by wild-type Escherichia coli or meningococcal LPS. Binding of FITC-labelled E. coli LPS TLR4 transfected Chinese hamster ovary (CHO) cells was inhibited by both mutant LPS-types. Experiments with CHO fibroblasts transfected with human CD14 and TLR4 showed that the antagonizing effect was dependent on the expression of human TLR4. In contrast to the situation in humans, lpxL1 LPS has agonistic activity for cytokine production in peritoneal macrophages of DBA mice, and exacerbated arthritis in murine collagen induced arthritis model. N. meningitidis lipid A mutant LPSs lpxL1 and lpxL2 function as LPS antagonists in humans by inhibiting TLR4-dependent cytokine production but have agonistic activity in mice.  相似文献   

20.
Recent work within our laboratory has focused on the enzymes we hypothesize are involved in the biosynthesis of bis(monoacylglycerol)phosphate from phosphatidylglycerol. Here we describe a transacylase, active at acidic pH values, isolated from a macrophage-like cell line, RAW 264.7. This enzyme acylates the head group glycerol of sn-3:sn-1' lysophosphatidylglycerol to form sn-3:sn-1' bis(monoacylglycerol)phosphate. Here we demonstrate that this enzyme uses two lysophosphatidylglycerol molecules, one as an acyl donor and another as an acyl acceptor, and that the acyl contributions from all other lipids tested are comparatively minor. This enzyme prefers saturated acyl chains to monounsaturates, 16 and 18 carbon fatty acids over 14 carbon fatty acids, and saturated acyl chains at the sn-1 position to monounsaturated acyl chains on the sn-2 carbon of lysophosphatidylglycerol. We present data which show the transacylase activity depends on the presence of a lipid-water interface and the lipid polymorphic state.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号