Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Anim Sci ; 92(8): 3375-87, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24902597

ABSTRACT

A transgenic Cassie (CA) line of Yorkshire (YK) pigs was developed using a transgene composed of the mouse parotid secretory protein promoter linked to the Escherichia coli phytase gene integrated in chromosome 4. Previous studies documented that salivary secretion of phytase was sufficient to enable efficient digestion of plant feed phytate P. In the present study the catalytic properties and tissue distribution of the phytase in CA pigs were determined by a combination of enzymatic assays, immunohistochemistry, and immunoblots of tissue samples. The E. coli phytase had a mass of 44.82 kDa whereas the phytase secreted in CA saliva had a mass of 52.42 kDa as a result of glycosylation of the enzyme in the parotid gland. Despite the difference in size, the 2 enzymes exhibited similar substrate specificities, and substrate affinity ( K: m) and maximum hydrolytic activity ( V: max) catalytic properties. Phytase assays showed that the enzyme was present at high specific activity in the salivary glands with low activity in the soft palate and essentially none in the kidney, lean (muscle), liver, or skin of CA pigs and none in YK pigs. This conclusion was supported by immunoblot analysis using a polyclonal anti-phytase antibody. Immunohistochemical analysis of 83 different tissue locations of CA and YK pigs confirmed the ubiquitous presence of phytase in serous cells of the salivary glands and the localized presence of phytase in both serous and mixed cell types in the submucosal glands of the oropharynx; in the pharynx, tonsils, and esophagus; in some Bowman's glands in the nasal mucosa and eustachian tube; and in the prostate gland of CA boars. Furthermore, it showed the absence of phytase from the kidney, lean, liver, and skin of CA pigs. Phytase was not detected in any of the conventional YK tissues tested. The phytase was found to be glycosylated with the allergenic galactose-α-1,3-galactose (α-gal) epitope by immunoblotting using α-gal specific monoclonal antibodies. Galactose-α-1,3-galactose glycosylation of proteins is a common feature of pork and other red meats. The α-gal epitope was shown to be associated with a few proteins in muscle and skin but with the greatest number of proteins in kidney and parotid tissues of CA and YK pigs. The absence of phytase from the major food tissues and the displacement of other α-gal glycosylated proteins in the parotid glands by α-gal glycosylated phytase in conjunction with previously published data support the contention that expression of the novel phytase has minimal influence on pork quality and safety.


Subject(s)
6-Phytase/metabolism , Animals, Genetically Modified/metabolism , Palate/metabolism , Parotid Gland/metabolism , Saliva/metabolism , Salivary Glands/metabolism , Swine/metabolism , Animals , Animals, Genetically Modified/genetics , Brain/metabolism , Escherichia coli/enzymology , Female , Hydrogen-Ion Concentration , Intestinal Mucosa/metabolism , Male , Muscle, Skeletal/metabolism , Swine/genetics , Tissue Distribution
2.
Phys Rev Lett ; 106(14): 140601, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21561176

ABSTRACT

Generalizations of the three main equations of quantum physics, namely, the Schrödinger, Klein-Gordon, and Dirac equations, are proposed. Nonlinear terms, characterized by exponents depending on an index q, are considered in such a way that the standard linear equations are recovered in the limit q→1. Interestingly, these equations present a common, solitonlike, traveling solution, which is written in terms of the q-exponential function that naturally emerges within nonextensive statistical mechanics. In all cases, the well-known Einstein energy-momentum relation is preserved for arbitrary values of q.

3.
Infect Immun ; 72(7): 3925-31, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15213136

ABSTRACT

The goal of this study was to determine whether Helicobacter pylori lipopolysaccharide (LPS) O-chain polysaccharide contributes to gastritis in a mouse model. C57BL/6J or C57BL/6-Prkdc(scid) (severe combined immunodeficient [SCID]) mice were inoculated with H. pylori strain SS1 or SS1::0826kan, in which a beta-1,4-galactosyltransferase (HP0826), an LPS biosynthetic enzyme, had been disrupted. H. pylori strain SS1::0826kan expresses truncated LPS lacking O chain. Recipient SCID mice were given C57BL/6J splenocytes by intraperitoneal injection. Bacterial colonization, gastric lesions (gastritis, neutrophilic infiltration, and gastric epithelial metaplasia), cellular (delayed-type hypersensitivity) and humoral immune responses to H. pylori sonicate, and gastric gamma interferon (IFN-gamma) mRNA expression were quantified. Recipient SCID mice colonized by H. pylori strain SS1 developed extensive gastritis with loss of normal fundic gland morphology. In contrast, gastric mucosa of recipient SCID mice colonized by H. pylori strain SS1::0826kan was not statistically distinguishable from that of uninfected recipient mice. Delayed-type hypersensitivity and humoral immune responses were detected in infected mice inoculated with wild-type SS1, but not with SS1::0826kan. IFN-gamma transcription was lower in mice infected with SS1::0826kan than in mice infected with SS1. In this model of rapidly progressive gastritis due to H. pylori, the O chain contributed to the extent of gastritis and to the host immune response. These data support a role for H. pylori LPS O chain in direct induction of the host immune response leading to gastritis and gastric damage and are in contrast to protein antigens, such as urease and cag products which do not contribute to gastritis in mice.


Subject(s)
Gastritis/microbiology , Helicobacter pylori/pathogenicity , O Antigens/metabolism , Animals , Gastric Mucosa/metabolism , Gastritis/immunology , Gastritis/metabolism , Helicobacter pylori/genetics , Helicobacter pylori/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , O Antigens/genetics , O Antigens/immunology , Spleen/metabolism
4.
Biochem Cell Biol ; 79(4): 449-59, 2001.
Article in English | MEDLINE | ID: mdl-11527214

ABSTRACT

Helicobacter pylori is a widespread Gram-negative bacterium responsible for the onset of various gastric pathologies and cancers in humans. A familiar trait of H. pylori is the production of cell-surface lipopolysaccharides (LPSs; O-chain --> core --> lipid A) with O-chain structures analogous to some mammalian histo-blood-group antigens, those being the Lewis determinants (Lea, Leb, Lex, sialyl Lex, Ley) and blood groups A and linear B. Some of these LPS antigens have been implicated as autoimmune, adhesion, and colonization components of H. pylori pathogenic mechanisms. This article describes the chemical structures of LPSs from H. pylori isolated from subjects with no overt signs of disease. Experimental data from chemical- and spectroscopic-based studies unanimously showed that these H. pylori manufactured extended heptoglycans composed of 2- and 3-linked D-glycero-alpha-D-manno-heptopyranose units and did not express any blood-group O-antigen chains. The fact that another H. pylori isolate with a similar LPS structure was shown to be capable of colonizing mice indicates that H. pylori histo-blood-group structures are not an absolute prerequisite for colonization in the murine model also. The absence of O-chains with histo-blood groups may cause H. pylori to become inept in exciting an immune response. Additionally, the presence of elongated heptoglycans may impede exposure of disease-causing outer-membrane antigens. These factors may render such H. pylori incapable of creating exogenous contacts essential for pathogenesis of severe gastroduodenal diseases and suggest that histo-blood groups in the LPS may indeed play a role in inducing a more severe H. pylori pathology.


Subject(s)
Helicobacter pylori/metabolism , Lewis Blood Group Antigens , Polysaccharides/metabolism , Carbohydrate Sequence , Electrophoresis, Polyacrylamide Gel , Helicobacter pylori/pathogenicity , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Polysaccharides/chemistry
5.
FEBS Lett ; 499(1-2): 1-5, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11418100

ABSTRACT

The lipid A structure of the Gram-negative bacterium Helicobacter mustelae, a ferret gastric pathogen responsible for the onset of gastric diseases in its host, was investigated. Two variant lipid A structures were found in the same strain. One structure contained a bisphosphorylated beta-(1-->6)-linked D-glucosamine backbone disaccharide with hydroxytetradecanoic acid in amide linkages. Unlike the structure described for the lipid A of the related human Helicobacter pylori gastric pathogen, which contains a C1 phosphate moiety, this lipid A presented phosphate groups at both the C1 and C4' positions, and contained no octadecanoyl fatty acid, which is present in H. pylori. The second lipid A structure had a different fatty acid composition in that 3-OH C(16) replaced most of the amide-linked 3-OH C(14).


Subject(s)
Helicobacter pylori/chemistry , Helicobacter/chemistry , Lipid A/chemistry , Animals , Carbohydrate Conformation , Fatty Acids/analysis , Fatty Acids/chemistry , Ferrets/microbiology , Glucosamine/metabolism , Lipopolysaccharides/pharmacology , Magnetic Resonance Spectroscopy , Myristates/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Trends Microbiol ; 8(12): 565-70, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11115753

ABSTRACT

In mimicry with human gastric epithelial cells, the lipopolysaccharide of Helicobacter pylori expresses Lewis blood group antigens. Recent data suggest that molecular mimicry does not promote immune evasion, nor does it lead to induction of autoantibodies, but that H. pylori Lewis X mediates adhesion to gastric epithelial cells and is essential for colonization.


Subject(s)
Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Lewis Blood Group Antigens/physiology , O Antigens/physiology , Bacterial Adhesion/physiology , Carbohydrate Sequence , Helicobacter pylori/immunology , Helicobacter pylori/physiology , Humans , Lewis Blood Group Antigens/chemistry , Lewis Blood Group Antigens/immunology , Molecular Mimicry , Molecular Sequence Data , O Antigens/chemistry , O Antigens/immunology , Virulence
8.
Carbohydr Res ; 329(1): 121-30, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11086692

ABSTRACT

The chemical and antigenic properties of the cell-surface lipopolysaccharides (LPSs) and capsular polysaccharides (CPSs) of seven representative strains of Actinobacillus suis from healthy and diseased pigs were investigated. Four strains produced a linear (1 --> 6)-beta-D-glucan homopolymer, beta-D-Glcp-(1-[ --> 6)-beta-D-Glcp-(1-]n -->, as a LPS-O-chain (O1) and as a CPS (K1). Polyclonal antisera prepared against a (1 --> 6)-beta-D-glucan-containing strain showed a positive reaction against both LPSs and CPSs derived from the above strains (designated serotype O1/K1). One strain carried the (1 --> 6)-beta-D-glucan solely as a LPS-O-chain (serotype O1) and two strains did not express the (1 --> 6)-beta-D-glucan, but, instead, produced a different O-chain (designated serotype 02); these three strains expressed their own characteristic CPSs. (1 --> 6)-beta-D-Glucan structures are common cell wall components of yeast, fungi and lichens, but, to our knowledge, this is the first time a (1 --> 6)-beta-D-glucan has been described in a prokaryotic organism. Conformational and nuclear magnetic resonance analyses showed that the beta-D-Glcp-(1 --> 6)-beta-D-Glcp linkage was flexible and two distinct glycosidic conformers are described. Cross-reactive antibodies to the A. suis (1 --> 6)-beta-D-glucan could be detected in sera from a variety of species and in sera from specific pathogen free pigs. This cross-reactivity may arise from immuno-stimulation of organisms present in the surrounding environment that contain (1 --> 6)-beta-D-glucan, which may also explain the high incidence of false positive results in previous serological tests for A. suis. In addition, these (1 --> 6)-beta-D-glucan background antibodies may be protective against A. suis infection. The characterization herein of (1 --> 6)-beta-D-glucan is the foundation for the development of a serotyping system for A. suis.


Subject(s)
Actinobacillus/chemistry , Glucans/immunology , Glucans/metabolism , beta-Glucans , Algorithms , Animals , Antibodies/metabolism , Carbohydrate Conformation , Carbohydrate Sequence , Cross Reactions , Gas Chromatography-Mass Spectrometry , Immune Sera/immunology , Immunoblotting , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/immunology , Serotyping , Swine/microbiology , Thermodynamics
9.
J Clin Microbiol ; 38(10): 3759-62, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11015398

ABSTRACT

A cell surface antigen-typing system was devised for the swine pathogen Actinobacillus suis and used to examine the prevalence of different lipopolysaccharide (O) types in healthy and diseased pigs. The strains examined in this study were isolated from a variety of locations in Canada and from Kansas. Lipopolysaccharide preparations of 151 isolates of A. suis were characterized by immunoblotting using polyclonal antisera generated to strains SO4 (O1/K1), H89-1173 (O2/K3), and VSB 3714, a rough strain. Approximately 54% (62 of 114) of A. suis isolates from diseased pigs, all (11 of 11) isolates from healthy pigs, and all (4 of 4) reference strains reacted with O1/K1 antiserum. More than 80% (18 of 22) of A. suis strains used for bacterin production and approximately 41% (47 of 114) of isolates from diseased pigs bound O2/K3 antiserum. One isolate appeared to be rough, and five were untypeable. O1/K1- and O2/K3-reactive strains were equally prevalent in Kansas, whereas O2/K3-reactive strains were more common in Québec and western Canada and O1/K1 strains were most common in Ontario. The fact that virtually all of the strains submitted for bacterin production were O2/K3-reactive strains is consistent with the notion that these strains may be more virulent than O1/K1 strains; alternatively, this may reflect geographic or other biases. In addition, we observed cross-reactivity between A. suis cell surface antigens and swine antisera to several other important pathogens. This finding may explain why previous attempts to develop a simple serodiagnostic test for A. suis have been unsuccessful.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus/classification , Lipopolysaccharides/analysis , Swine Diseases/microbiology , Swine/microbiology , Actinobacillus/isolation & purification , Actinobacillus Infections/microbiology , Animals , Bacterial Typing Techniques , Canada , Cross Reactions , Immunoblotting , Kansas , Rabbits , Reference Values
10.
Infect Immun ; 68(10): 5928-32, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10992504

ABSTRACT

Helicobacter pylori NCTC 11637 lipopolysaccharide (LPS) expresses the human blood group antigens Lewis x (Le(x)), Le(y), and H type I. In this report, we demonstrate that the H type I epitope displays high-frequency phase variation. One variant expressed Le(x) and Le(y) and no H type I as determined by serology; this switch was reversible. Insertional mutagenesis in NCTC 11637 of JHP563 (a poly(C) tract containing an open reading frame homologous to glycosyltransferases) yielded a transformant with a serotype similar to the phase variant. Structural analysis of the NCTC 11637 LPS confirmed the loss of the H type I epitope. Sequencing of JHP563 in strains NCTC 11637, an H type I-negative variant, and an H type I-positive switchback variant showed a C14 (gene on), C13 (gene off), and C14 tract, respectively. Inactivation of strain G27, which expresses Le(x), Le(y), H type I, and Le(a), yielded a transformant that expressed Le(x) and Le(y). We conclude that JHP563 encodes a beta3-galactosyltransferase involved in the biosynthesis of H type I and Le(a) and that phase variation in H type I is due to C-tract changes in this gene. A second H type I-negative variant (variant 3a) expressed Le(x) and Le(a) and had lost both H type I and Le(y) expression. Inactivation of HP093-HP094 resulted in a transformant expressing Le(x) and lacking Le(y) and H type I. Structural analysis of a mutant LPS confirmed the serological data. We conclude that the HP093-HP094 alpha2-fucosyltransferase (alpha2-FucT) gene product is involved in the biosynthesis of both Le(y) and Le(x). Finally, we inactivated HP0379 in strain 3a. The transformant had lost both Le(x) and Le(a) expression, which demonstrates that the HP0379 gene product is both an alpha3- and an alpha4-FucT. Our data provide understanding at the molecular level of how H. pylori is able to diversify in the host, a requirement likely essential for successful colonization and transmission.


Subject(s)
Epitopes , Helicobacter pylori/immunology , Lewis Blood Group Antigens/immunology , Lipopolysaccharides/classification , Lipopolysaccharides/immunology , Antibodies, Monoclonal/immunology , Carbohydrate Sequence , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Humans , Lewis Blood Group Antigens/chemistry , Lipopolysaccharides/chemistry , Molecular Sequence Data , Mutagenesis, Insertional , Polymerase Chain Reaction , Sequence Analysis, DNA
11.
Eur J Biochem ; 267(19): 6059-66, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10998067

ABSTRACT

The identification of Helicobacter pylori isolates that expresses exclusively type I Lewis antigens is necessary to determine the biosynthetic pathway of these antigens. Fast-atom bombardment MS provides evidence that the H. pylori isolate UA1111 expresses predominantly Leb, with H type I and Lea in lesser amounts. Cloning and expression of the H. pylori fucosyltransferases (FucTs) allow comparisons with previously identified H. pylori enzymes and determination of the enzyme specificities. Although all FucTs, one alpha(1,2) FucT and two alpha(1,3/4) FucTs, appear to be functional in this isolate, their activities are lower and enzyme specificities are different to other H. pylori FucTs previously characterized. Studies of the cloned enzyme activities and mutational analysis indicate that Lea acts as the substrate for the synthesis of Leb. This is different from the human Leb biosynthetic pathway, but analogous to the biosynthetic pathway utilized by H. pylori for the production of Ley.


Subject(s)
Antigens, Bacterial/biosynthesis , Bacterial Proteins/metabolism , Fucose/metabolism , Fucosyltransferases/metabolism , Helicobacter pylori/enzymology , Lewis Blood Group Antigens/biosynthesis , Molecular Mimicry , Oligosaccharides/biosynthesis , Amino Acid Sequence , Bacterial Proteins/genetics , Base Sequence , Carbohydrate Sequence , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Frameshift Mutation , Fucosyltransferases/deficiency , Fucosyltransferases/genetics , Gene Targeting , Glycosylation , Helicobacter pylori/genetics , Helicobacter pylori/immunology , Humans , Molecular Sequence Data , Spectrometry, Mass, Fast Atom Bombardment , Galactoside 2-alpha-L-fucosyltransferase
12.
Glycobiology ; 10(7): 701-13, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10910974

ABSTRACT

Past studies have shown that the cell surface lipopolysaccharides (LPSs) of the ubiquitous human gastric pathogen Helicobacter pylori (a type 1 carcinogen) isolated from people residing in Europe and North America express predominantly type 2 Lewis x (Le(x)) and Le(y) epitopes and, infrequently, type 1 Le(a), Le(b), and Le(d) antigens. This production of Lewis blood-group structures by H. pylori LPSs, similar to those found in the surfaces of human gastric cells, allows the bacterium to mimic its human niche. In this study, LPSs of H.pylori strains extracted from patients living in China, Japan, and Singapore were chemically and serologically analyzed. When compared with Western H.pylori LPSs, these Asian strains showed a stronger tendency to produce type 1 blood groups. Of particular interest, and novel observations in H.pylori, the O-chain regions of strains F-58C and R-58A carried type 1 Le(a) without the presence of type 2 Le(x), strains R-7A and H607 were shown to have the capability of producing the type 1 blood group A antigen, and strains CA2, H507, and H428 expressed simultaneously the difucosyl isomeric antigens, type 1 Le(b) and type 2 Le(y). The apparent proclivity for the production of type 1 histo-blood group antigens in Asian H.pylori LPSs, as compared with Western strains, may be an adaptive evolutionary effect in that differences in the gastric cell surfaces of the respective hosts might be significantly dissimilar to select for the formation of different LPS structures on the resident H.pylori strain.


Subject(s)
Helicobacter Infections/epidemiology , Helicobacter Infections/immunology , Helicobacter pylori/chemistry , Lewis Blood Group Antigens/chemistry , Lipopolysaccharides/chemistry , ABO Blood-Group System/chemistry , Asia , Carbohydrate Sequence , Epitopes/chemistry , Humans , Molecular Mimicry , Molecular Sequence Data , Monosaccharides/analysis , Nuclear Magnetic Resonance, Biomolecular , Species Specificity , Spectrometry, Mass, Fast Atom Bombardment , Stomach Neoplasms/microbiology , Stomach Ulcer/microbiology
13.
Mol Microbiol ; 35(6): 1530-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10760152

ABSTRACT

Helicobacter pylori NCTC11637 expresses a lipopolysaccharide (LPS) that comprises an O antigen side-chain with structural homology to the human blood group antigen Lewis X (Le(x)). The role of this molecule in adhesion of H. pylori to gastric epithelial cells was investigated. Mutants expressing truncated LPS structures were generated through insertional mutagenesis of rfbM and galE; genes encode GDP mannose pyrophosphorylase and galactose epimerase respectively. Compositional and structural analysis revealed that the galE mutant expressed a rough LPS that lacked an O antigen side-chain. In contrast, an O antigen side-chain was still synthesized by the rfbM mutant, but it lacked fucose and no longer reacted with anti-Le(x) monoclonal antibodies (Mabs). The ability of these mutants to bind to paraffin-embedded sections from the antrum region of a human stomach was assessed. Adhesion of the wild type was characterized by tropic binding to the apical surface of mucosal epithelial cells and cells lining gastric pits. In contrast, both the rfbM and galE mutants failed to demonstrate tropic binding and adhered to the tissue surface in a haphazard manner. These results indicate that LPS and, more specifically, Le(x) structures in the O antigen side-chain play an important role in targeting H. pylori to specific cell lineages within the gastric mucosa. The role of Le(x) in this interaction was confirmed by the tropic binding of synthetic Le(x), conjugated to latex beads, to gastric tissue. The observed pattern of adhesion was indistinguishable from that of wild-type H. pylori.


Subject(s)
Epithelial Cells/microbiology , Helicobacter pylori/chemistry , Helicobacter pylori/physiology , Lewis X Antigen/chemistry , O Antigens/chemistry , Bacterial Adhesion , Carbohydrate Sequence , Gastric Mucosa/microbiology , Humans , Latex , Lewis X Antigen/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Molecular Sequence Data , Mutation , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism
14.
Mol Microbiol ; 35(5): 1156-67, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712696

ABSTRACT

A previously annotated open reading frame (ORF) (HP0826) from Helicobacter pylori was cloned and expressed in Escherichia coli cells and determined to be a beta-1,4-galactosyltransferase that used GlcNAc as an acceptor. Mutational analysis in H. pylori strains demonstrated that this enzyme plays a key role in the biosynthesis of the type 2 N-acetyl-lactosamine (LacNAc) polysaccharide O-chain backbone, by catalysing the addition of Gal to GlcNAc. To examine the potential role of this O-chain structure in bacterial colonization of the host stomach, the mutation was introduced into H. pylori strain SS1 which is known to be capable of colonizing the gastric mucosa of mice. Compared with the parental strain, mutated SS1 was less efficient at colonizing the murine stomach.


Subject(s)
Genome, Bacterial , Helicobacter pylori/genetics , Lipopolysaccharides/metabolism , Mutagenesis , N-Acetyllactosamine Synthase/metabolism , Alleles , Amino Acid Sequence , Animals , Base Sequence , Carbohydrate Sequence , DNA Primers , Electrophoresis, Polyacrylamide Gel , Female , Helicobacter Infections/enzymology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/enzymology , Helicobacter pylori/metabolism , Helicobacter pylori/pathogenicity , Lipopolysaccharides/chemistry , Mice , Molecular Sequence Data , N-Acetyllactosamine Synthase/chemistry , Sequence Homology, Amino Acid , Spectrometry, Mass, Fast Atom Bombardment , Stomach/microbiology
15.
Mol Microbiol ; 35(4): 718-27, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10692150

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that is notorious for its intrinsic drug resistance. We have used chemical and genetic techniques to characterize three putative kinase genes that are involved in the addition of phosphate to the inner core region of P. aeruginosa lipopolysaccharide. The first gene is a waaP homologue, whereas the other two (wapP and wapQ) are unique to P. aeruginosa. Repeated attempts using a variety of membrane-stabilizing conditions to generate waaP:Gm (Gm, gentamicin) or wapP:Gm mutants were unsuccessful. We were able to generate a chromosomal waaP mutant that had a wild-type copy of either waaPPa or waaPEc in trans, but were unable to cure this plasmid-borne copy of the gene. These results are consistent with the fact that P. aeruginosa mutants lacking inner core heptose (Hep) or phosphate have never been isolated and demonstrate the requirement of Hep-linked phosphate for P. aeruginosa viability. A wapQ:Gm mutant was isolated and it had an unaltered minimum inhibitory concentration (MIC) for novobiocin and only a small decrease in the MIC for sodium dodecyl sulphate (SDS), suggesting that the loss of a phosphate group transferred by WapQ may only be having a small impact on outer-membrane permeability. Nuclear magnetic resonance and methylation linkage analysis showed that WaaPPa could add one phosphate to O4 of HepI in a Salmonella typhimurium waaP mutant. The expression of WaaPPa increased the outer-membrane integrity of these complemented mutants, as evidenced by 35-fold and 75-fold increases in the MIC for novobiocin and SDS respectively. The S. typhimurium waaP mutant transformed with both waaP and wapP had over 250-fold and 1000-fold increases, respectively, in these MICs. The inner core phosphates of P. aeruginosa appear to be playing a key role in the intrinsic drug resistance of this bacterium.


Subject(s)
Drug Resistance, Microbial , Phosphates/metabolism , Polysaccharides, Bacterial/metabolism , Pseudomonas aeruginosa/metabolism , Carbohydrate Sequence , Cell Division , Cell Membrane Permeability , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Genes, Bacterial/genetics , Genes, Essential , Heptoses/metabolism , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Multigene Family , Mutation , Phosphorylation , Phosphotransferases/genetics , Polysaccharides, Bacterial/chemistry , Pseudomonas aeruginosa/cytology , Pseudomonas aeruginosa/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Sequence Analysis, DNA
16.
Eur J Biochem ; 267(2): 305-20, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10632700

ABSTRACT

This study describes the molecular makeup of the cell-wall lipopolysaccharides (LPSs) (O-chain polysaccharide-->core oligosaccharide-->lipid A) from five Helicobacter pylori strains: H. pylori 26695 and J99, the complete genome sequences of which have been published, the established mouse model Sydney strain (SS1), and the symptomatic strains P466 and UA915. All chemical and serological experiments were performed on the intact LPSs. H. pylori 26695 and SS1 possessed either a low-Mr semi-rough-form LPS carrying mostly a single Ley type-2 blood-group determinant in the O-chain region covalently attached to the core oligosaccharide or a high-Mr smooth-form LPS, as did strain J99, with an elongated partially fucosylated type-2 N-acetyllactosamine (polyLacNAc) O-chain polymer, terminated mainly by a Lex blood-group determinant, connected to the core oligosaccharide. In the midst of semi-rough-form LPS glycoforms, H. pylori 26695 and SS1 also expressed in the O-chain region a difucosylated antigen, alpha-L-Fucp(1-3)-alpha-L-Fucp(1-4)-beta-D-GlcpNAc, and the cancer-cell-related type-1 or type-2 linear B-blood-group antigen, alpha-D-Galp(1-3)-beta-D-Galp(1-3 or 4)-beta-D-GlcpNAc. The LPS of H. pylori strain P466 carried the cancer-associated type-2 sialyl Lex blood-group antigen, and the LPS from strain UA915 expressed a type-1 Leb blood-group unit. These findings should aid investigations that focus on identifying and characterizing genes responsible for LPS biosynthesis in genomic strains 26695 and J99, and in understanding the role of H. pylori LPS in animal model studies. The LPSs from the H. pylori strains studied to date were grouped into specific glycotype families.


Subject(s)
Helicobacter pylori/chemistry , Lewis X Antigen/chemistry , Lipopolysaccharides/chemistry , Lipopolysaccharides/classification , Animals , Carbohydrate Sequence , Helicobacter pylori/classification , Helicobacter pylori/genetics , Helicobacter pylori/immunology , Lewis Blood Group Antigens/chemistry , Lipopolysaccharides/analysis , Magnetic Resonance Spectroscopy , Mice/microbiology , Molecular Sequence Data , Oligosaccharides/chemistry , Species Specificity
17.
J Biol Chem ; 274(49): 35129-38, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10574995

ABSTRACT

During O antigen lipopolysaccharide (LPS) synthesis in bacteria, transmembrane migration of undecaprenylpyrophosphate (Und-P-P)-bound O antigen subunits occurs before their polymerization and ligation to the rest of the LPS molecule. Despite the general nature of the translocation process, putative O-antigen translocases display a low level of amino acid sequence similarity. In this work, we investigated whether complete O antigen subunits are required for translocation. We demonstrate that a single sugar, GlcNAc, can be incorporated to LPS of Escherichia coli K-12. This incorporation required the functions of two O antigen synthesis genes, wecA (UDP-GlcNAc:Und-P GlcNAc-1-P transferase) and wzx (O-antigen translocase). Complementation experiments with putative O-antigen translocases from E. coli O7 and Salmonella enterica indicated that translocation of O antigen subunits is independent of the chemical structure of the saccharide moiety. Furthermore, complementation with putative translocases involved in synthesis of exopolysaccharides demonstrated that these proteins could not participate in O antigen assembly. Our data indicate that recognition of a complete Und-P-P-bound O antigen subunit is not required for translocation and suggest a model for O antigen synthesis involving recognition of Und-P-P-linked sugars by a putative complex made of Wzx translocase and other proteins involved in the processing of O antigen.


Subject(s)
Bacterial Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , O Antigens/biosynthesis , O Antigens/metabolism , Agglutination , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Hexosyltransferases/metabolism , Lipid A/metabolism , Membrane Proteins , Mutagenesis , Oligosaccharides/metabolism , Recombinant Proteins/metabolism , Spectrometry, Mass, Fast Atom Bombardment
18.
Infect Immun ; 67(10): 5361-6, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10496917

ABSTRACT

The lipopolysaccharide (LPS) of Helicobacter pylori expresses the Lewis x (Lex) and/or Ley antigen. We have shown previously that H. pylori LPS displays phase variation whereby an Lex-positive strain yields variants with different LPS serotypes, for example, Lex plus Ley or nonfucosylated polylactosamine. H. pylori has two alpha3-fucosyltransferase genes that both contain poly(C) tracts. We now demonstrate that these tracts can shorten or lengthen randomly, which results in reversible frameshifting and inactivation of the gene products. We provide genetic and serological evidence that this mechanism causes H. pylori LPS phase variation and demonstrate that the on or off status of alpha3-fucosyltransferase genes determines the LPS serotypes of phase variants and clinical isolates. The role of the alpha3-fucosyltransferase gene products in determining the LPS serotype was confirmed by structural-chemical analysis of alpha3-fucosyltransferase knockout mutants. The data also show that the two alpha3-fucosyltransferase genes code for enzymes with different fine specificities, and we propose the names futA and futB to designate the orthologs of the H. pylori 26695 alpha3-fucosyltransferase genes HP0379 and HP0651, respectively. The data also show that the alpha3-fucosylation precedes alpha2-fucosylation [corrected], an order of events opposite to that which prevails in mammals. Finally, the data provide an understanding at the molecular level of the mechanisms underlying LPS diversity in H. pylori, which may play an important role in adaptation to the host.


Subject(s)
Fucosyltransferases/genetics , Helicobacter pylori/pathogenicity , Lipopolysaccharides/chemistry , Poly C/chemistry , Fucosyltransferases/physiology , Lewis Blood Group Antigens/analysis , Lewis X Antigen/analysis , Mutation
19.
J Bacteriol ; 181(3): 772-80, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9922239

ABSTRACT

Escherichia coli O8:K40 coexpresses two distinct lipopolysaccharide (LPS) structures on its surface. The O8 polysaccharide is a mannose homopolymer with a trisaccharide repeat unit and is synthesized by an ABC-2 transport-dependent pathway. The K40LPS backbone structure is composed of a trisaccharide repeating unit of N-acetylglucosamine (GlcNAc) and glucuronic acid (GlcA) and has an uncommon substitution, an L-serine moiety attached to glucuronic acid. The gene cluster responsible for synthesis of the K40 polysaccharide has previously been cloned and sequenced and was found to contain six open reading frames (ORFs) (P. A. Amor and C. Whitfield, Mol. Microbiol. 26:145-161, 1997). Here, we demonstrate that insertional inactivation of orf1 results in the accumulation of a semirough (SR)-K40LPS form which retains reactivity with specific polyclonal serum in Western immunoblots. Structural and compositional analysis of the SR-K40LPS reveals that it comprises a single K40 repeat unit attached to lipid A core. The lack of polymerization of the K40 polysaccharide indicates that orf1 encodes the K40 polymerase (Wzy) and that assembly of the K40 polysaccharide occurs via a Wzy-dependent pathway (in contrast to that of the O8 polysaccharide). Inactivation of orf3 also results in the accumulation of an SR-LPS form which fails to react with specific polyclonal K40 serum in Western immunoblots. Methylation linkage analysis and fast atom bombardment-mass spectrometry of this SR-LPS reveals that the biological repeat unit of the K40 polysaccharide is GlcNAc-GlcA-GlcNAc. Additionally, this structure lacks the L-serine substitution of GlcA. These results show that (i) orf3 encodes the enzyme responsible for the addition of the L-serine residue to the K40 backbone and (ii) substitution of individual K40 repeats with L-serine is essential for their recognition and polymerization into the K40 polysaccharide by Wzy.


Subject(s)
Antigens, Bacterial , Antigens, Surface/biosynthesis , Escherichia coli Proteins/biosynthesis , Escherichia coli/metabolism , Glycosyltransferases/biosynthesis , Lipopolysaccharides/biosynthesis , Serine/metabolism , Acetylglucosamine/analysis , Antigens, Surface/chemistry , Antigens, Surface/genetics , Carbohydrate Conformation , Carbohydrate Sequence , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Glucuronates/analysis , Glucuronic Acid , Glycosyltransferases/chemistry , Glycosyltransferases/genetics , Lipopolysaccharides/chemistry , Molecular Sequence Data , Mutagenesis, Insertional , Open Reading Frames , Polymerase Chain Reaction , Restriction Mapping , Trisaccharides/chemistry
20.
J Biol Chem ; 273(41): 26310-6, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9756860

ABSTRACT

The waaY, waaQ, and waaP genes are located in the central operon of the waa (formerly rfa) locus on the chromosome of Escherichia coli. This locus contains genes whose products are involved in the assembly of the core region of the lipopolysaccharide molecule. In the R1 core prototype strain, E. coli F470, there are nine genes in this operon, and all but waaY, waaQ, and waaP have been assigned function. In this study, the waaY, waaQ, and waaP genes were independently mutated by insertion of a non-polar antibiotic resistance cassette, and the structures of the resulting mutant core oligosaccharides were determined by chemical analyses and phosphorus-nuclear magnetic resonance spectroscopy. All three of these mutations were shown to affect the modification of the heptose region of the core, a region whose structure is critical to outer membrane stability. Mutation of waaY resulted in a core oligosaccharide devoid of phosphate on HepII. Mutation of waaQ resulted in loss of the branch HepIII residue on HepII and impeded the activity of WaaY. Mutation of waaP resulted in loss of phosphoryl substituents on HepI and obviated WaaQ and WaaY activity. Only mutation of waaP resulted in hypersensitivity to novobiocin and sodium dodecyl sulfate, a characteristic of deep-rough mutations.


Subject(s)
Escherichia coli/metabolism , Genes, Bacterial , Lipopolysaccharides/metabolism , Base Sequence , Carbohydrate Conformation , Carbohydrate Sequence , Cell Membrane/metabolism , DNA Primers , Escherichia coli/genetics , Lipopolysaccharides/chemistry , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Mutagenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...