Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Glycobiology ; 33(6): 512-524, 2023 06 21.
Article in English | MEDLINE | ID: mdl-36943318

ABSTRACT

The Actinobacterial species Cellulomonas fimi ATCC484 has long been known to secrete mannose-containing proteins, but a closer examination of glycoproteins associated with the cell has never been reported. Using ConA lectin chromatography and mass spectrometry, we have surveyed the cell-associated glycoproteome from C. fimi and collected detailed information on the glycosylation sites of 19 cell-associated glycoproteins. In addition, we have expressed a previously known C. fimi secreted cellulase, Celf_3184 (formerly CenA), a putative peptide prolyl-isomerase, Celf_2022, and a penicillin-binding protein, Celf_0189, in the mannosylation capable host, Corynebacterium glutamicum. We found that the glycosylation machinery in C. glutamicum was able to use the recombinant C. fimi proteins as substrates and that the glycosylation matched closely that found in the native proteins when expressed in C. fimi. We are pursuing this observation as a prelude to dissecting the biosynthetic machinery and biological consequences of this protein mannosylation.


Subject(s)
Actinobacteria , Actinobacteria/genetics , Glycosylation , Glycoproteins/genetics , Glycoproteins/metabolism , Recombinant Proteins/metabolism , Mannose/metabolism
2.
Biotechnol Bioeng ; 119(9): 2331-2344, 2022 09.
Article in English | MEDLINE | ID: mdl-35508753

ABSTRACT

Alpha-1-antitrypsin (A1AT) is a serine protease inhibitor which blocks the activity of serum proteases including neutrophil elastase to protect the lungs. Its deficiency is known to increase the risk of pulmonary emphysema as well as chronic obstructive pulmonary disease. Currently, the only treatment for patients with A1AT deficiency is weekly injection of plasma-purified A1AT. There is still today no commercial source of therapeutic recombinant A1AT, likely due to significant differences in expression host-specific glycosylation profile and/or high costs associated with the huge therapeutic dose needed. Accordingly, we aimed to produce high levels of recombinant wild-type A1AT, as well as a mutated protein (mutein) version for increased oxidation resistance, with N-glycans analogous to human plasma-derived A1AT. To achieve this, we disrupted two endogenous glycosyltransferase genes controlling core α-1,6-fucosylation (Fut8) and α-2,3-sialylation (ST3Gal4) in CHO cells using CRISPR/Cas9 technology, followed by overexpression of human α-2,6-sialyltransferase (ST6Gal1) using a cumate-inducible expression system. Volumetric A1AT productivity obtained from stable CHO pools was 2.5- to 6.5-fold higher with the cumate-inducible CR5 promoter compared to five strong constitutive promoters. Using the CR5 promoter, glycoengineered stable CHO pools were able to produce over 2.1 and 2.8 g/L of wild-type and mutein forms of A1AT, respectively, with N-glycans analogous to the plasma-derived clinical product Prolastin-C. Supplementation of N-acetylmannosamine to the cell culture media during production increased the overall sialylation of A1AT as well as the proportion of bi-antennary and disialylated A2G2S2 N-glycans. These purified recombinant A1AT proteins showed in vitro inhibitory activity equivalent to Prolastin-C and substitution of methionine residues 351 and 358 with valines rendered A1AT significantly more resistant to oxidation. The recombinant A1AT mutein bearing an improved oxidation resistance described in this study could represent a viable biobetter drug, offering a safe and more stable alternative for augmentation therapy.


Subject(s)
alpha 1-Antitrypsin Deficiency , alpha 1-Antitrypsin , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Polysaccharides , Recombinant Proteins/metabolism , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin/pharmacology , alpha 1-Antitrypsin Deficiency/drug therapy
3.
Proc Natl Acad Sci U S A ; 114(5): E679-E688, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096352

ABSTRACT

The vast majority of proteins are posttranslationally altered, with the addition of covalently linked sugars (glycosylation) being one of the most abundant modifications. However, despite the hydrolysis of protein peptide bonds by peptidases being a process essential to all life on Earth, the fundamental details of how peptidases accommodate posttranslational modifications, including glycosylation, has not been addressed. Through biochemical analyses and X-ray crystallographic structures we show that to hydrolyze their substrates, three structurally related metallopeptidases require the specific recognition of O-linked glycan modifications via carbohydrate-specific subsites immediately adjacent to their peptidase catalytic machinery. The three peptidases showed selectivity for different glycans, revealing protein-specific adaptations to particular glycan modifications, yet always cleaved the peptide bond immediately preceding the glycosylated residue. This insight builds upon the paradigm of how peptidases recognize substrates and provides a molecular understanding of glycoprotein degradation.


Subject(s)
Peptide Hydrolases/metabolism , Polysaccharides/metabolism , Escherichia coli/genetics , Fetuins/metabolism , Glycopeptides/metabolism , Glycosylation , Mucins/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Protein Conformation , Protein Processing, Post-Translational
4.
Bioprocess Biosyst Eng ; 42(5): 711-725, 2019 May.
Article in English | MEDLINE | ID: mdl-30673843

ABSTRACT

Mammalian-inducible expression systems are increasingly available and offer an attractive platform for the production of recombinant proteins. In this work, we have conducted process development for a cumate-inducible GS-CHO cell-line-expressing rituximab. To cope with the limitations encountered in batch when inducing at high cell densities, we have explored the use of fed-batch, sequential medium replacements, and continuous perfusion strategies applied during the pre-induction (growth) phase to enhance process performance in terms of product yield and quality. In shake flask, a fed-batch mode and a complete medium exchange at the time of induction were shown to significantly increase the integral of viable cell concentration and antibody titer compared to batch culture. Further enhancement of product yield was achieved by combining bolus concentrated feed additions with sequential medium replacement, but product galactosylation was reduced compared to fed-batch mode, as a result of the extended culture duration. In bioreactor, combining continuous perfusion of the basal medium with bolus daily feeding during the pre-induction period and harvesting earlier during the production phase is shown to provide a good trade-off between antibody titer and product galactosylation. Overall, our results demonstrate the importance of selecting a suitable operating mode and harvest time when carrying out high-cell-density induction to balance between culture productivity and product quality.


Subject(s)
Batch Cell Culture Techniques , Bioreactors , Rituximab/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Rituximab/isolation & purification
5.
J Biol Chem ; 288(1): 247-54, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23184955

ABSTRACT

Enzymatic addition of GalNAc to isotopically labeled IFNα2a produced in Escherichia coli yielded the O-linked glycoprotein GalNAcα-[(13)C,(15)N]IFNα2a. The three-dimensional structure of GalNAcα-IFNα2a has been determined in solution by NMR spectroscopy at high resolution. Proton-nitrogen heteronuclear Overhauser enhancement measurements revealed that the addition of a single monosaccharide unit at Thr-106 significantly slowed motions of the glycosylation loop on the nanosecond time scale. Subsequent addition of a Gal unit produced Gal(ß1,3)GalNAcα-[(13)C,(15)N]IFNα2a. This extension resulted in a further decrease in the dynamics of this loop. The methodology used here allowed the first such description of the structure and dynamics of an O-glycoprotein and opens the way to the study of this class of proteins.


Subject(s)
Acetylgalactosamine/chemistry , Interferon-alpha/metabolism , Polysaccharides/chemistry , Threonine/chemistry , Acetylgalactosamine/genetics , Computational Biology/methods , Disulfides/chemistry , Escherichia coli/metabolism , Glycoproteins/chemistry , Glycosylation , Humans , Interferon alpha-2 , Interferons/chemistry , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Conformation , Peptides/chemistry , Protein Conformation , Recombinant Proteins/metabolism
6.
Commun Chem ; 6(1): 189, 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37684364

ABSTRACT

Glycosylation is a key quality attribute that must be closely monitored for protein therapeutics. Established assays such as HILIC-Fld of released glycans and LC-MS of glycopeptides work well for glycoproteins with a few glycosylation sites but are less amenable for those with multiple glycosylation sites, resulting in complex datasets that are time consuming to generate and difficult to analyze. As part of efforts to improve preparedness for future pandemics, researchers are currently assessing where time can be saved in the vaccine development and production process. In this context, we evaluated if neutral and acidic monosaccharides analysis via HPAEC-PAD could be used as a rapid and robust alternative to LC-MS and HILIC-Fld for monitoring glycosylation between protein production batches. Using glycoengineered spike proteins we show that the HPAEC-PAD monosaccharide assays could quickly and reproducibly detect both major and minor glycosylation differences between batches. Moreover, the monosaccharide results aligned well with those obtained by HILIC-Fld and LC-MS.

7.
J Biol Chem ; 286(14): 12361-70, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21257763

ABSTRACT

Campylobacter jejuni is well known for synthesizing ganglioside mimics within the glycan component of its lipooligosaccharide (LOS), which have been implicated in triggering Guillain-Barré syndrome. We now confirm that this pathogen is capable of synthesizing a much broader spectrum of host glycolipid/glycoprotein mimics within its LOS. P blood group and paragloboside (lacto-N-neotetraose) antigen mimicry is exhibited by RM1221, a strain isolated from a poultry source. RM1503, a gastroenteritis-associated strain, expresses lacto-N-biose and sialyl-Lewis c units, the latter known as the pancreatic tumor-associated antigen, DU-PAN-2 (or LSTa). C. jejuni GC149, a Guillain-Barré syndrome-associated strain, expresses an unusual sialic acid-containing hybrid oligosaccharide with similarity to both ganglio and Pk antigens and can, through phase variation of its LOS biosynthesis genes, display GT1a or GD3 ganglioside mimics. We show that the sialyltransferase CstII and the galactosyltransferase CgtD are involved in the synthesis of multiple mimic types, with LOS structural diversity achieved through evolving allelic substrate specificity.


Subject(s)
Campylobacter jejuni/metabolism , Gangliosides/metabolism , Lipopolysaccharides/metabolism , Bacterial Proteins/metabolism , Galactosyltransferases/metabolism , Lipopolysaccharides/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Sialyltransferases/metabolism , Substrate Specificity
8.
Org Biomol Chem ; 9(16): 5809-15, 2011 Aug 21.
Article in English | MEDLINE | ID: mdl-21727969

ABSTRACT

Biotinylated analogues of gangliosides GM2, GM1, GD1a and GalNAc-GD1a were synthesized in high yields using glycosyltransferases from Campylobacter jejuni. The presence of a biotin moiety in the aglycone part of these mimics allows for attachment of these materials onto various streptavidin-coated surfaces. Analysis of the interaction of biotin-appended GM1 with the B subunit of Escherichia coli heat-labile enterotoxin performed in a modified ELISA procedure shows the potential of this compound to replace the natural GM1 in toxin detection.


Subject(s)
Biotin/analogs & derivatives , Campylobacter jejuni/enzymology , Gangliosides/chemistry , Gangliosides/metabolism , Glycosyltransferases/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Gangliosides/chemical synthesis
9.
J Biotechnol ; 307: 87-97, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-31697975

ABSTRACT

Alpha-1-antitrypsin (A1AT) is an abundant serum inhibitor of serine proteases. A1AT deficiency is a common genetic disorder which is currently treated with augmentation therapies. These treatments involve weekly injections of patients with purified plasma-derived A1AT. Such therapies can be extremely expensive and rely on plasma donors. Hence, large-scale production of recombinant A1AT (rA1AT) could greatly benefit these patients, as it could decrease the cost of treatments, reduce biosafety concerns and ensure quantitative and qualitative controls of the protein. In this report, we sought to produce α2,6-sialylated rA1AT with our cumate-inducible stable CHO pool expression system. Our different CHO pools could reach volumetric productivities of 1,2 g/L. The human α2,6-sialyltransferase was stably expressed in these cells in order to mimic elevated α2,6-sialylation levels of native A1AT protein. Sialylation of the recombinant protein was stable over the duration of the fed-batch production phase and was higher in a pool where cells were sorted and enriched by FACS based on cell-surface α2,6-sialylation. Addition of ManNAc to the cell culture media during production enhanced both α2,3 and α2,6 A1AT sialylation levels whereas addition of 2F-peracetylfucose potently inhibited fucosylation of the protein. Finally, we demonstrated that rA1AT proteins exhibited human neutrophil elastase inhibitory activities similar to the commercial human plasma-derived A1AT.


Subject(s)
Leukocyte Elastase/antagonists & inhibitors , Sialyltransferases/metabolism , alpha 1-Antitrypsin/metabolism , Animals , Biosimilar Pharmaceuticals/metabolism , CHO Cells , Cricetulus , Humans , Recombinant Proteins , Sialyltransferases/genetics , alpha 1-Antitrypsin/genetics , beta-D-Galactoside alpha 2-6-Sialyltransferase
10.
Curr Res Immunol ; 1: 23-37, 2020 Dec.
Article in English | MEDLINE | ID: mdl-35493857

ABSTRACT

The effector functions of the IgGs are modulated by the N-glycosylation of their Fc region. Particularly, the absence of core fucosylation is known to increase the affinity of IgG1s for the Fcγ receptor IIIa expressed by immune cells, in turn translating in an improvement in the antibody-dependent cellular cytotoxicity. However, the impact of galactosylation and sialylation is still debated in the literature. In this study, we have investigated the influence of high and low levels of core fucosylation, terminal galactosylation and terminal α2,6-sialylation of the Fc N-glycans of trastuzumab on its affinity for the FcγRIIIa. A large panel of antibody glycoforms (i.e., highly α2,6-sialylated or galactosylated IgG1s, with high or low levels of core fucosylation) were generated and characterized, while their interactions with the FcγRs were analysed by a robust surface plasmon resonance-based assay as well as in a cell-based reporter bioassay. Overall, IgG1 glycoforms with reduced fucosylation display a stronger affinity for the FcγRIIIa. In addition, fucosylation, and the presence of terminal galactose and sialic acids are shown to increase the affinity for the FcγRIIIa as compared to the agalactosylated forms. These observations perfectly translate in the response observed in our reporter bioassay.

11.
J Biotechnol ; 298: 45-56, 2019 Jun 10.
Article in English | MEDLINE | ID: mdl-30959136

ABSTRACT

In order to maximize cell growth and productivity for an inducible CHO cell line expressing rituximab, various fed-batch culture strategies were investigated. In each case, the performance was evaluated for cultures induced at moderate and high cell density conditions (4 × 106 and 10 × 106 cells/mL) to assess the impact of the timing of induction. We first demonstrate the importance of starting the feeding process during the growth phase, as this translated into significantly improved integral of viable cells and antibody concentration, when compared to post-induction feeding only. Secondly, we investigated the impact of the feed rate by maintaining different levels of glucose (25, 35 and 50 mM) via a dynamic feeding strategy. The highest antibody concentrations were achieved under a moderate feeding regime for both cell densities at induction, highlighting the risks of under- or over-feeding the cultures. We then evaluated the impact of performing a temperature shift at induction by testing different mild hypothermia conditions. At small-scale, the highest production yields (1.2 g/L) were achieved when the temperature was reduced from 37 to 30 °C during the production phase of a culture induced at high cell density. When the strategy was applied in bioreactor, the better controlled conditions led to even greater product concentrations (1.8 g/L). Furthermore, this production protocol was shown to promote a more galactosylated glycan profile than a bioreactor culture initiated at 34 °C during growth and downshifted to 30 °C during the production phase.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Batch Cell Culture Techniques/methods , Cell Proliferation/genetics , Rituximab/biosynthesis , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , CHO Cells , Cell Survival/genetics , Cricetulus , Glucose/metabolism , Humans , Rituximab/chemistry , Rituximab/genetics
12.
Biotechnol Prog ; 35(1): e2742, 2019 01.
Article in English | MEDLINE | ID: mdl-30414355

ABSTRACT

Inducible mammalian expression systems are becoming increasingly available and are not only useful for the production of cytotoxic/cytostatic products, but also confer the unique ability to uncouple the growth and production phases. In this work, we have specifically investigated how the cell culture state at the time of induction influences the cumate-inducible expression of recombinant rituximab by a GS-CHO cell line. To this end, cells grown in batch and fed-batch cultures were induced at increasing cell densities (1 to 10 × 10 6 cells/mL). In batch, the cell specific productivity and the product yield were found to reduce with increasing cell density at induction. A dynamic feeding strategy using a concentrated nutrient solution applied prior and postinduction allowed to significantly increase the integral of viable cells and led to a 3-fold increase in the volumetric productivity (1.2 g/L). The highest product yields were achieved for intermediate cell densities at induction, as cultures induced during the late exponential phase (10 × 10 6 cells/mL) were associated with a shortened production phase. The final glycosylation patterns remained however similar, irrespective of the cell density at induction. The kinetics of growth and production in a 2 L bioreactor were largely comparable to shake flasks for a similar cell density at induction. The degree of galactosylation was found to decrease over time, but the final glycan distribution at harvest was consistent to that of the shake flasks cultures. Taken together, our results provide useful insights for the rational development of fed-batch cell culture processes involving inducible CHO cells. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 35: e2742, 2019.


Subject(s)
Rituximab/metabolism , Animals , Batch Cell Culture Techniques , Bioreactors , Biotechnology , CHO Cells , Cricetinae , Cricetulus , Glycosylation
13.
Chembiochem ; 9(10): 1632-40, 2008 Jul 02.
Article in English | MEDLINE | ID: mdl-18491328

ABSTRACT

Helicobacter pylori is a highly persistent and common pathogen in humans. It is the causative agent of chronic gastritis and its further stages. HP0826 is the beta-1,4-galactosyltransferase involved in the biosynthesis of the LPS O-chain backbone of H. pylori. Though it was first cloned nearly a decade ago, there are surprisingly limited data about the characteristics of HP0826, especially given its prominent role in H. pylori pathogenicity. We here demonstrate that HP0826 is a highly efficient and promiscuous biocatalyst. We have exploited two novel enzymatic activities for the quantitative synthesis of the thiodisaccharide Gal-beta-S-1,4-GlcNAc-pNP as well as Gal-beta-1,4-Man-pNP. We further show that Neisseria meningitidis beta-1,4-galactosyltransferases LgtB can be used as an equally efficient catalyst in the latter reaction. Thiodisaccharides have been extensively used in structural biology but can also have therapeutic uses. The Gal-beta-1,4-Man linkage is found in the Leishmania species LPG backbone disaccharide repeats and cap, which have been associated with vector binding in Leishmaniasis.


Subject(s)
Helicobacter pylori/metabolism , N-Acetyllactosamine Synthase/metabolism , Thioglycosides/biosynthesis , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Catalysis , Disaccharides/chemical synthesis , Disaccharides/chemistry , Escherichia coli/enzymology , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , N-Acetyllactosamine Synthase/isolation & purification , Thioglycosides/chemistry
14.
J Biotechnol ; 281: 39-47, 2018 Sep 10.
Article in English | MEDLINE | ID: mdl-29886030

ABSTRACT

For pre-clinical evaluation of biotherapeutic candidates, protein production by transient gene expression (TGE) in Chinese Hamster Ovary (CHO) cells offers important advantages, including the capability of rapidly and cost-effectively generating recombinant proteins that are highly similar to those produced in stable CHO clones. We have established a novel CHO clone (CHO-3E7) expressing a form of the Epstein-Barr virus nuclear antigen-1 (EBNA-1) with improved TGE productivity relative to parental CHO cells. Taking advantage of a new transfection-compatible media formulation that permits prolonged, high-density culture, we optimized transfection parameters (cell density, plasmid vector and polyethylenimine concentrations) and post-transfection culture conditions to establish a new, high-performing process for rapid protein production. The growth media is chemically defined, and a single hydrolysate feed is added post-transfection, followed by periodic glucose supplementation. This method gave significantly higher yields than our standard low-cell density, F17-based CHO-3E7 TGE method, averaging several hundred mg/l for a panel of recombinant proteins and antibodies. Purified antibodies produced using the two methods had distinct glycosylation profiles but showed identical target binding kinetics by SPR. Key advantages of this new protein production platform include the cost-effectiveness of the transfection reagent, the commercial availability of the culture media and the ability to perform high-cell-density transfection without media change.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/genetics , Polyethyleneimine , Transfection/methods , Trastuzumab/biosynthesis , Animals , CHO Cells , Cell Count , Cricetulus , Gene Expression
15.
J Clin Invest ; 114(11): 1659-65, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15578098

ABSTRACT

Molecular mimicry of Campylobacter jejuni lipo-oligosaccharides (LOS) with gangliosides in nervous tissue is considered to induce cross-reactive antibodies that lead to Guillain-Barre syndrome (GBS), an acute polyneuropathy. To determine whether specific bacterial genes are crucial for the biosynthesis of ganglioside-like structures and the induction of anti-ganglioside antibodies, we characterized the C. jejuni LOS biosynthesis gene locus in GBS-associated and control strains. We demonstrated that specific types of the LOS biosynthesis gene locus are associated with GBS and with the expression of ganglioside-mimicking structures. Campylobacter knockout mutants of 2 potential GBS marker genes, both involved in LOS sialylation, expressed truncated LOS structures without sialic acid, showed reduced reactivity with GBS patient serum, and failed to induce an anti-ganglioside antibody response in mice. We demonstrate, for the first time, to our knowledge, that specific bacterial genes are crucial for the induction of anti-ganglioside antibodies.


Subject(s)
Autoantibodies/biosynthesis , Campylobacter jejuni/genetics , Gangliosides/immunology , Guillain-Barre Syndrome/immunology , Lipopolysaccharides , Molecular Mimicry , Animals , Biomarkers , Campylobacter jejuni/chemistry , Carbohydrate Conformation , Cross Reactions , Gangliosides/chemistry , Genes, Bacterial , Humans , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/chemistry , Mice , Molecular Sequence Data , Mutation , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/immunology
16.
PLoS One ; 11(3): e0151186, 2016.
Article in English | MEDLINE | ID: mdl-26950732

ABSTRACT

The bacteria in the genus Cellulomonas are known for their ability to degrade plant cell wall biomass. Cellulomonas fimi ATCC 484 and C. flavigena ATCC 482 have been the subject of much research into secreted cellulases and hemicellulases. Recently the genome sequences of both C. fimi ATCC 484 and C. flavigena ATCC 482 were published, and a genome comparison has revealed their full spectrum of possible carbohydrate-active enzymes (CAZymes). Using mass spectrometry, we have compared the proteins secreted by C. fimi and C. flavigena during growth on the soluble cellulose substrate, carboxymethylcellulose (CMC), as well as a soluble xylan fraction. Many known C. fimi CAZymes were detected, which validated our analysis, as were a number of new CAZymes and other proteins that, though identified in the genome, have not previously been observed in the secretome of either organism. Our data also shows that many of these are co-expressed on growth of either CMC or xylan. This analysis provides a new perspective on Cellulomonas enzymes and provides many new CAZyme targets for characterization.


Subject(s)
Cellulomonas/metabolism , Proteomics , Carboxymethylcellulose Sodium/metabolism , Cellulomonas/enzymology , Species Specificity , Xylans/metabolism
17.
Biochemistry ; 46(50): 14704-14, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18034462

ABSTRACT

The lipooligosaccharides (LOS) of Campylobacter jejuni is an important virulence factor. Its core oligosaccharide component is frequently sialylated and bears a close resemblance with host gangliosides. The display of ganglioside mimics by this bacterium is believed to trigger the onset of the autoimmune condition Guillain-Barré syndrome (GBS) in some individuals. Considerable effort has been directed toward the structural characterization of the glycan component of the LOS of C. jejuni strains isolated from GBS patients. Capillary electrophoresis-mass spectrometry (CE-MS) has been a particularly useful analytical technique applied toward this task. Conventional analysis of bacterial LOS by CE-MS has generally involved the prior removal of O-acyl lipid chains, which is necessary for the effective solubilization and separation of the heterogeneous ensemble of LOS species. Unfortunately, O-deacylation causes the undesired removal of important glycan-associated O-linked modifications, such as O-acetate and O-linked amino acids. In this report, we describe a CE-MS technique developed for the rapid analysis of fully intact LOS from C. jejuni. Using this method, we report the structural characterization of the glycan from 10 GBS-associated strains and two enteritis strains, using material isolated from as little as one colony. The application of this technique has enabled us to unambiguously identify LOS-bound O-acetylated sialic acid in a number of strains and has revealed for the first time that C. jejuni frequently modifies its core with O-linked glycine. Our studies demonstrate that MS-based structural analysis of bacterial LOS can be optimized to the level where only a single-colony quantity of material is required and time-consuming chemical treatments can be avoided.


Subject(s)
Campylobacter jejuni/metabolism , Glycine/chemistry , Lipopolysaccharides/chemistry , Mass Spectrometry/methods , N-Acetylneuraminic Acid/chemistry , Acetylation , Carbohydrate Sequence , Electrophoresis, Capillary , Glycine/metabolism , Lipopolysaccharides/metabolism , Molecular Sequence Data , N-Acetylneuraminic Acid/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
18.
Infect Immun ; 75(3): 1245-54, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17261613

ABSTRACT

Molecular mimicry between lipooligosaccharides (LOS) of Campylobacter jejuni and gangliosides in peripheral nerves plays a crucial role in the pathogenesis of C. jejuni-related Guillain-Barré syndrome (GBS). We have analyzed the LOS outer core structures of 26 C. jejuni strains associated with GBS and its variant, Miller Fisher syndrome (MFS), by capillary electrophoresis coupled with electrospray ionization mass spectrometry. Sixteen out of 22 (73%) GBS-associated and all 4 (100%) MFS-associated strains expressed LOS with ganglioside mimics. GM1a was the most prevalent ganglioside mimic in GBS-associated strains (10/22, 45%), and in eight of these strains, GM1a was found in combination with GD1a mimics. All seven strains isolated from patients with ophthalmoplegia (GBS or MFS) expressed disialylated (GD3 or GD1c) mimics. Three out of 22 GBS-associated strains (14%) did not express sialylated ganglioside mimics because their LOS locus lacked the genes necessary for sialylation. Three other strains (14%) did not express ganglioside mimics because of frameshift mutations in either the cstII sialyltransferase gene or the cgtB galactosyltransferase gene. It is not possible to determine if these mutations were already present during C. jejuni infection. This is the first report in which mass spectrometry combined with DNA sequence data were used to infer the LOS outer core structures of a large number of neuropathy-associated C. jejuni strains. We conclude that molecular mimicry between gangliosides and C. jejuni LOS is the presumable pathogenic mechanism in most cases of C. jejuni-related GBS. However, our findings suggest that in some cases, other mechanisms may play a role. Further examination of the disease etiology in these patients is mandatory.


Subject(s)
Campylobacter jejuni/chemistry , Guillain-Barre Syndrome/metabolism , Guillain-Barre Syndrome/microbiology , Lipopolysaccharides/chemistry , Miller Fisher Syndrome/metabolism , Miller Fisher Syndrome/microbiology , Amino Acid Sequence , Campylobacter jejuni/genetics , Campylobacter jejuni/metabolism , Carbohydrate Sequence , Guillain-Barre Syndrome/enzymology , Humans , Lipopolysaccharides/metabolism , Miller Fisher Syndrome/enzymology , Molecular Mimicry , Molecular Sequence Data , Sialyltransferases/chemistry , Sialyltransferases/genetics
19.
Electrophoresis ; 26(17): 3360-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16097024

ABSTRACT

Lipooligosaccharide (LOS) is the major component of the external membrane of Campylobacter jejuni. LOS contains a hydrophobic moiety, lipid A, and a hydrophilic moiety, oligosaccharide. Due to the unique mimicry of human ganglioside structures and potential involvement in the induction of the autoimmune polyneuropathies, Guillain-Barré and Miller Fisher syndromes, the structural characterization of C. jejuni LOS has received much attention. We have been using capillary zone electrophoresis-mass spectrometry to analyze O-deacylated LOS from C. jejuni. In an attempt to optimize the separation conditions, the effect of methanol on the separation of LOS was investigated. It was found that methanol resulted in stronger adsorption of LOS onto the wall of fused-silica capillary. In this paper, we applied this adsorption to perform electrophoresis-assisted open-tubular liquid chromatography electrospray mass spectrometry for the analysis of O-deacylated LOS mixtures from C. jejuni. The analytical potential of the proposed strategy for the analysis of O-deacylated LOS glycoforms from five bacterial colonies is demonstrated. Online tandem mass spectrometry is shown to provide a powerful tool for characterization of variations in the hexosamine backbone, phosphorylation of the lipid A, and sialic acid sequence information.


Subject(s)
Campylobacter jejuni/chemistry , Electrophoresis, Capillary/methods , Lipopolysaccharides/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Carbohydrate Sequence
20.
Mol Microbiol ; 55(1): 90-103, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15612919

ABSTRACT

We recently demonstrated that Campylobacter jejuni produces a capsular polysaccharide (CPS) that is the major antigenic component of the classical Penner serotyping system distinguishing Campylobacter into >60 groups. Although the wide variety of C. jejuni serotypes are suggestive of structural differences in CPS, the genetic mechanisms of such differences are unknown. In this study we sequenced biosynthetic cps regions, ranging in size from 15 to 34 kb, from selected C. jejuni strains of HS:1, HS:19, HS:23, HS:36, HS:23/36 and HS:41 serotypes. Comparison of the determined cps sequences of the HS:1, HS:19 and HS:41 strains with the sequenced strain, NCTC11168 (HS:2), provides evidence for multiple mechanisms of structural variation including exchange of capsular genes and entire clusters by horizontal transfer, gene duplication, deletion, fusion and contingency gene variation. In contrast, the HS:23, HS:36 and HS:23/36 cps sequences were highly conserved. We report the first detailed structural analysis of 81-176 (HS:23/36) and G1 (HS:1) and refine the previous structural interpretations of the HS:19, HS:23, HS:36 and HS:41 serostrains. For the first time, we demonstrate the commonality and function of a second heptose biosynthetic pathway for Campylobacter CPS independent of the pathway for lipooligosaccharide (LOS) biosynthesis and identify a novel heptosyltransferase utilized by this alternate pathway. Furthermore, we show the retention of two functional heptose isomerases in Campylobacter and the sharing of a phosphatase for both LOS and CPS heptose biosynthesis.


Subject(s)
Bacterial Capsules/genetics , Campylobacter jejuni/genetics , Genetic Variation , Bacterial Capsules/chemistry , Bacterial Capsules/metabolism , Campylobacter jejuni/metabolism , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/physiology , Carbohydrate Sequence , DNA, Bacterial , Gene Duplication , Gene Transfer, Horizontal , Genes, Bacterial , Glycosyltransferases/genetics , Glycosyltransferases/physiology , Heptoses/chemistry , Heptoses/genetics , Lipopolysaccharides/biosynthesis , Molecular Sequence Data , Multigene Family , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , Recombination, Genetic , Sequence Analysis, DNA , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL