Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Front Microbiol ; 12: 734526, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867850

RESUMO

Campylobacter jejuni is a common cause of diarrheal disease worldwide. Human infection typically occurs through the ingestion of contaminated poultry products. We previously demonstrated that an attenuated Escherichia coli live vaccine strain expressing the C. jejuni N-glycan on its surface reduced the Campylobacter load in more than 50% of vaccinated leghorn and broiler birds to undetectable levels (responder birds), whereas the remainder of the animals was still colonized (non-responders). To understand the underlying mechanism, we conducted three vaccination and challenge studies using 135 broiler birds and found a similar responder/non-responder effect. Subsequent genome-wide association studies (GWAS), analyses of bird sex and levels of vaccine-induced IgY responses did not correlate with the responder versus non-responder phenotype. In contrast, antibodies isolated from responder birds displayed a higher Campylobacter-opsonophagocytic activity when compared to antisera from non-responder birds. No differences in the N-glycome of the sera could be detected, although minor changes in IgY glycosylation warrant further investigation. As reported before, the composition of the microbiota, particularly levels of OTU classified as Clostridium spp., Ruminococcaceae and Lachnospiraceae are associated with the response. Transplantation of the cecal microbiota of responder birds into new birds in combination with vaccination resulted in further increases in vaccine-induced antigen-specific IgY responses when compared to birds that did not receive microbiota transplants. Our work suggests that the IgY effector function and microbiota contribute to the efficacy of the E. coli live vaccine, information that could form the basis for the development of improved vaccines targeted at the elimination of C. jejuni from poultry.

2.
ACS Chem Biol ; 16(11): 2690-2701, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34726367

RESUMO

Campylobacter jejuni is a significant cause of human gastroenteritis worldwide, and all strains express an N-glycan that is added to at least 80 different proteins. We characterized 98 C. jejuni isolates from infants from 7 low- and middle-income countries and identified 4 isolates unreactive with our N-glycan-specific antiserum that was raised against the C. jejuni heptasaccharide composed of GalNAc-GalNAc-GalNAc(Glc)-GalNAc-GalNAc-diNAcBac. Mass spectrometric analyses indicated these isolates express a hexasaccharide lacking the glucose branch. Although all 4 strains encode the PglI glucosyltransferase (GlcTF), one aspartate in the DXDD motif was missing, an alteration also present in ∼4% of all available PglI sequences. Deleting this residue from an active PglI resulted in a nonfunctional GlcTF when the protein glycosylation system was reconstituted in E. coli, while replacement with Glu/Ala was not deleterious. Molecular modeling proposed a mechanism for how the DXDD residues and the structure/length beyond the motif influence activity. Mouse vaccination with an E. coli strain expressing the full-length heptasaccharide produced N-glycan-specific antibodies and a corresponding reduction in Campylobacter colonization and weight loss following challenge. However, the antibodies did not recognize the hexasaccharide and were unable to opsonize C. jejuni isolates lacking glucose, suggesting this should be considered when designing N-glycan-based vaccines to prevent campylobacteriosis.


Assuntos
Campylobacter jejuni/metabolismo , Glucose/metabolismo , Polissacarídeos/metabolismo , Sequência de Aminoácidos , Animais , Ácido Aspártico/química , Configuração de Carboidratos , Sequência de Carboidratos , Ensaio de Imunoadsorção Enzimática , Escherichia coli/metabolismo , Glicosilação , Soros Imunes , Camundongos , Fagocitose , Polissacarídeos/química , Alinhamento de Sequência
3.
PLoS One ; 15(12): e0244031, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33378351

RESUMO

Ecotin, first described in Escherichia coli, is a potent inhibitor of a broad range of serine proteases including those typically released by the innate immune system such as neutrophil elastase (NE). Here we describe the identification of ecotin orthologs in various Campylobacter species, including Campylobacter rectus and Campylobacter showae residing in the oral cavity and implicated in the development and progression of periodontal disease in humans. To investigate the function of these ecotins in vitro, the orthologs from C. rectus and C. showae were recombinantly expressed and purified from E. coli. Using CmeA degradation/protection assays, fluorescence resonance energy transfer and NE activity assays, we found that ecotins from C. rectus and C. showae inhibit NE, factor Xa and trypsin, but not the Campylobacter jejuni serine protease HtrA or its ortholog in E. coli, DegP. To further evaluate ecotin function in vivo, an E. coli ecotin-deficient mutant was complemented with the C. rectus and C. showae homologs. Using a neutrophil killing assay, we demonstrate that the low survival rate of the E. coli ecotin-deficient mutant can be rescued upon expression of ecotins from C. rectus and C. showae. In addition, the C. rectus and C. showae ecotins partially compensate for loss of N-glycosylation and increased protease susceptibility in the related pathogen, Campylobacter jejuni, thus implicating a similar role for these proteins in the native host to cope with the protease-rich environment of the oral cavity.


Assuntos
Campylobacter rectus/metabolismo , Campylobacter/metabolismo , Proteínas de Escherichia coli/genética , Proteínas Periplásmicas/genética , Inibidores de Serina Proteinase/metabolismo , Inibidores da Tripsina/metabolismo , Animais , Campylobacter/genética , Campylobacter rectus/genética , Células Cultivadas , Galinhas , Humanos , Neutrófilos/efeitos dos fármacos , Elastase Pancreática/antagonistas & inibidores , Homologia de Sequência , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/farmacologia , Inibidores da Tripsina/farmacologia
4.
Front Microbiol ; 11: 1191, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32625174

RESUMO

Campylobacter fetus is commonly associated with venereal disease and abortions in cattle and sheep, and can also cause intestinal or systemic infections in humans that are immunocompromised, elderly, or exposed to infected livestock. It is also believed that C. fetus infection can result from the consumption or handling of contaminated food products, but C. fetus is rarely detected in food since isolation methods are not suited for its detection and the physiology of the organism makes culturing difficult. In the related species, Campylobacter jejuni, the ability to colonize the host has been linked to N-linked protein glycosylation with quantitative proteomics demonstrating that glycosylation is interconnected with cell physiology. Using label-free quantitative (LFQ) proteomics, we found more than 100 proteins significantly altered in expression in two C. fetus subsp. fetus protein glycosylation (pgl) mutants (pglX and pglJ) compared to the wild-type. Significant increases in the expression of the (NiFe)-hydrogenase HynABC, catalyzing H2-oxidation for energy harvesting, correlated with significantly increased levels of cellular nickel, improved growth in H2 and increased hydrogenase activity, suggesting that N-glycosylation in C. fetus is involved in regulating the HynABC hydrogenase and nickel homeostasis. To further elucidate the function of the C. fetus pgl pathway and its enzymes, heterologous expression in Escherichia coli followed by mutational and functional analyses revealed that PglX and PglY are novel glycosyltransferases involved in extending the C. fetus hexasaccharide beyond the conserved core, while PglJ and PglA have similar activities to their homologs in C. jejuni. In addition, the pgl mutants displayed decreased motility and ethidium bromide efflux and showed an increased sensitivity to antibiotics. This work not only provides insight into the unique protein N-glycosylation pathway of C. fetus, but also expands our knowledge on the influence of protein N-glycosylation on Campylobacter cell physiology.

5.
J Biol Chem ; 295(28): 9513-9530, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32424044

RESUMO

Clostridium perfringens is a leading cause of food-poisoning and causes avian necrotic enteritis, posing a significant problem to both the poultry industry and human health. No effective vaccine against C. perfringens is currently available. Using an antiserum screen of mutants generated from a C. perfringens transposon-mutant library, here we identified an immunoreactive antigen that was lost in a putative glycosyltransferase mutant, suggesting that this antigen is likely a glycoconjugate. Following injection of formalin-fixed whole cells of C. perfringens HN13 (a laboratory strain) and JGS4143 (chicken isolate) intramuscularly into chickens, the HN13-derived antiserum was cross-reactive in immunoblots with all tested 32 field isolates, whereas only 5 of 32 isolates were recognized by JGS4143-derived antiserum. The immunoreactive antigens from both HN13 and JGS4143 were isolated, and structural analysis by MALDI-TOF-MS, GC-MS, and 2D NMR revealed that both were atypical lipoteichoic acids (LTAs) with poly-(ß1→4)-ManNAc backbones substituted with phosphoethanolamine. However, although the ManNAc residues in JGS4143 LTA were phosphoethanolamine-modified, a few of these residues were instead modified with phosphoglycerol in the HN13 LTA. The JGS4143 LTA also had a terminal ribose and ManNAc instead of ManN in the core region, suggesting that these differences may contribute to the broadly cross-reactive response elicited by HN13. In a passive-protection chicken experiment, oral challenge with C. perfringens JGS4143 lead to 22% survival, whereas co-gavage with JGS4143 and α-HN13 antiserum resulted in 89% survival. This serum also induced bacterial killing in opsonophagocytosis assays, suggesting that HN13 LTA is an attractive target for future vaccine-development studies.


Assuntos
Galinhas , Infecções por Clostridium , Clostridium perfringens , Lipopolissacarídeos , Doenças das Aves Domésticas , Ácidos Teicoicos , Animais , Galinhas/imunologia , Galinhas/microbiologia , Infecções por Clostridium/imunologia , Infecções por Clostridium/prevenção & controle , Clostridium perfringens/química , Clostridium perfringens/imunologia , Humanos , Lipopolissacarídeos/química , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/prevenção & controle , Ácidos Teicoicos/química , Ácidos Teicoicos/imunologia , Ácidos Teicoicos/farmacologia
7.
Commun Biol ; 3(1): 2, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31925306

RESUMO

Although the gastrointestinal pathogen Campylobacter jejuni was considered asaccharolytic, >50% of sequenced isolates possess an operon for L-fucose utilization. In C. jejuni NCTC11168, this pathway confers L-fucose chemotaxis and competitive colonization advantages in the piglet diarrhea model, but the catabolic steps remain unknown. Here we solved the putative dehydrogenase structure, resembling FabG of Burkholderia multivorans. The C. jejuni enzyme, FucX, reduces L-fucose and D-arabinose in vitro and both sugars are catabolized by fuc-operon encoded enzymes. This enzyme alone confers chemotaxis to both sugars in a non-carbohydrate-utilizing C. jejuni strain. Although C. jejuni lacks fucosidases, the organism exhibits enhanced growth in vitro when co-cultured with Bacteroides vulgatus, suggesting scavenging may occur. Yet, when excess amino acids are available, C. jejuni prefers them to carbohydrates, indicating a metabolic hierarchy exists. Overall this study increases understanding of nutrient metabolism by this pathogen, and identifies interactions with other gut microbes.


Assuntos
Bacteroides/metabolismo , Campylobacter jejuni/metabolismo , Metabolismo dos Carboidratos , Açúcares/metabolismo , Simbiose , Bacteroides/imunologia , Campylobacter jejuni/imunologia , Quimiotaxia , Fucose/química , Fucose/metabolismo , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Mucinas/metabolismo , Açúcares/química
8.
Glycobiology ; 30(2): 105-119, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31588498

RESUMO

Campylobacter jejuni is a prevalent gastrointestinal pathogen associated with increasing rates of antimicrobial resistance development. It was also the first bacterium demonstrated to possess a general N-linked protein glycosylation pathway capable of modifying > 80 different proteins, including the primary Campylobacter multidrug efflux pump, CmeABC. Here we demonstrate that N-glycosylation is necessary for the function of the efflux pump and may, in part, explain the evolutionary pressure to maintain this protein modification system. Mutants of cmeA in two common wildtype (WT) strains are highly susceptible to erythromycin (EM), ciprofloxacin and bile salts when compared to the isogenic parental strains. Complementation of the cmeA mutants with the native cmeA allele restores the WT phenotype, whereas expression of a cmeA allele with point mutations in both N-glycosylation sites is comparable to the cmeA mutants. Moreover, loss of CmeA glycosylation leads to reduced chicken colonization levels similar to the cmeA knock-out strain, while complementation fully restores colonization. Reconstitution of C. jejuni CmeABC into Escherichia coli together with the C. jejuni N-glycosylation pathway increases the EM minimum inhibitory concentration and decreases ethidium bromide accumulation when compared to cells lacking the pathway. Molecular dynamics simulations reveal that the protein structures of the glycosylated and non-glycosylated CmeA models do not vary from one another, and in vitro studies show no change in CmeA multimerization or peptidoglycan association. Therefore, we conclude that N-glycosylation has a broader influence on CmeABC function most likely playing a role in complex stability.


Assuntos
Proteínas de Bactérias , Campylobacter jejuni , Proteínas de Membrana Transportadoras , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções por Campylobacter/genética , Infecções por Campylobacter/metabolismo , Infecções por Campylobacter/veterinária , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Galinhas , Glicosilação , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/microbiologia
9.
Curr Opin Chem Biol ; 53: 16-24, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31288139

RESUMO

Historically, protein glycosylation was believed to be restricted to eukaryotes, but now is abundantly represented in all three domains of life. The first bacterial N-linked glycosylation system was discovered in the Gram-negative pathogen, Campylobacter jejuni, and subsequently transferred into the heterologous Escherichia coli host beginning a new era of synthetic bacterial glycoengineering. Since then, additional N-glycosylation pathways have been characterized resembling the classical C. jejuni system and unconventional new approaches for N-glycosylation have been uncovered. These include cytoplasmic protein modification, direct glycan transfer to proteins, and use of alternate amino acid acceptors, deepening our understanding of the vast mechanisms bacteria possess for protein modification and providing opportunities to expand the glycoengineering toolbox for designing novel vaccine formulations and protein therapeutics.


Assuntos
Bactérias/genética , Bactérias/metabolismo , Nitrogênio/metabolismo , Biologia Sintética/métodos , Arginina/metabolismo , Engenharia Genética , Glicosilação
10.
Nat Commun ; 10(1): 1390, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30918252

RESUMO

The AB5 toxins cholera toxin (CT) from Vibrio cholerae and heat-labile enterotoxin (LT) from enterotoxigenic Escherichia coli are notorious for their roles in diarrheal disease, but their effect on other intestinal bacteria remains unexplored. Another foodborne pathogen, Campylobacter jejuni, can mimic the GM1 ganglioside receptor of CT and LT. Here we demonstrate that the toxin B-subunits (CTB and LTB) inhibit C. jejuni growth by binding to GM1-mimicking lipooligosaccharides and increasing permeability of the cell membrane. Furthermore, incubation of CTB or LTB with a C. jejuni isolate capable of altering its lipooligosaccharide structure selects for variants lacking the GM1 mimic. Examining the chicken GI tract with immunofluorescence microscopy demonstrates that GM1 reactive structures are abundant on epithelial cells and commensal bacteria, further emphasizing the relevance of this mimicry. Exposure of chickens to CTB or LTB causes shifts in the gut microbial composition, providing evidence for new toxin functions in bacterial gut competition.


Assuntos
Toxinas Bacterianas/farmacologia , Campylobacter jejuni/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Toxina da Cólera/farmacologia , Enterotoxinas/farmacologia , Proteínas de Escherichia coli/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Lipopolissacarídeos/metabolismo , Animais , Bactérias/efeitos dos fármacos , Bactérias/metabolismo , Campylobacter jejuni/metabolismo , Galinhas , Gangliosídeo G(M1)/metabolismo , Glicoconjugados/metabolismo , Mucosa Intestinal/patologia , Microscopia de Fluorescência , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo
11.
Nat Commun ; 10(1): 891, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30792408

RESUMO

Chemical synthesis of conjugate vaccines, consisting of a polysaccharide linked to a protein, can be technically challenging, and in vivo bacterial conjugations (bioconjugations) have emerged as manufacturing alternatives. Bioconjugation relies upon an oligosaccharyltransferase to attach polysaccharides to proteins, but currently employed enzymes are not suitable for the generation of conjugate vaccines when the polysaccharides contain glucose at the reducing end, which is the case for ~75% of Streptococcus pneumoniae capsules. Here, we use an O-linking oligosaccharyltransferase to generate a polyvalent pneumococcal bioconjugate vaccine with polysaccharides containing glucose at their reducing end. In addition, we show that different vaccine carrier proteins can be glycosylated using this system. Pneumococcal bioconjugates are immunogenic, protective and rapidly produced within E. coli using recombinant techniques. These proof-of-principle experiments establish a platform to overcome limitations of other conjugating enzymes enabling the development of bioconjugate vaccines for many important human and animal pathogens.


Assuntos
Escherichia coli/genética , Engenharia Genética/métodos , Vacinas Pneumocócicas/genética , Animais , Cápsulas Bacterianas/genética , Cápsulas Bacterianas/imunologia , Escherichia coli/metabolismo , Glicoproteínas/genética , Glicoproteínas/imunologia , Glicoproteínas/isolamento & purificação , Glicosilação , Humanos , Vacinas Pneumocócicas/isolamento & purificação , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/imunologia , Vacinas Conjugadas/genética , Vacinas Conjugadas/isolamento & purificação , Vacinas Sintéticas/genética , Vacinas Sintéticas/isolamento & purificação
12.
Biotechnol Biofuels ; 11: 259, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30258493

RESUMO

Protein glycosylation pathways have been identified in a variety of bacteria and are best understood in pathogens and commensals in which the glycosylation targets are cell surface proteins, such as S layers, pili, and flagella. In contrast, very little is known about the glycosylation of bacterial enzymes, especially those secreted by cellulolytic bacteria. Caldicellulosiruptor bescii secretes several unique synergistic multifunctional biomass-degrading enzymes, notably cellulase A which is largely responsible for this organism's ability to grow on lignocellulosic biomass without the conventional pretreatment. It was recently discovered that extracellular CelA is heavily glycosylated. In this work, we identified an O-glycosyltransferase in the C. bescii chromosome and targeted it for deletion. The resulting mutant was unable to grow on crystalline cellulose and showed no detectable protein glycosylation. Multifunctional biomass-degrading enzymes in this strain were rapidly degraded. With the genetic tools available in C. bescii, this system represents a unique opportunity to study the role of bacterial enzyme glycosylation as well an investigation of the pathway for protein glycosylation in a non-pathogen.

13.
Microbiology (Reading) ; 164(6): 896-907, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29856309

RESUMO

Phase variation (PV), involving stochastic switches in gene expression, is exploited by the human pathogen Campylobacter jejuni to adapt to different environmental and host niches. Phase-variable genes of C. jejuni modulate expression of multiple surface determinants, and hence may influence host colonization. Population bottlenecks can rapidly remove the diversity generated by PV, and strict single-cell bottlenecks can lead to propagation of PV states with highly divergent phenotypes. Using a combination of high-throughput fragment size analysis and comparison with in vivo and in silico bottleneck models, we have characterized a narrow population bottleneck during the experimental colonization of broiler chickens with C. jejuni strain 81-176. We identified high levels of variation in five PV genes in the inoculum, and subsequently, massively decreased population diversity following colonization. Each bird contained a dominant five-gene phasotype that was present in the inoculum indicative of random sorting through a narrow, non-selective bottleneck during colonization. These results are evidence of the potential for confounding effects of PV on in vivo studies of Campylobacter colonization factors and poultry vaccine studies. Our results are also an argument for population bottlenecks as mediators of stochastic variability in the propensity to survive through the food chain and cause clinical human disease.


Assuntos
Infecções por Campylobacter/veterinária , Campylobacter jejuni/crescimento & desenvolvimento , Campylobacter jejuni/genética , Variação Genética , Doenças das Aves Domésticas/microbiologia , Adaptação Fisiológica , Animais , Vacinas Bacterianas/administração & dosagem , Variação Biológica da População , Infecções por Campylobacter/microbiologia , Campylobacter jejuni/imunologia , Galinhas , Expressão Gênica , Frequência do Gene , Genes Bacterianos/genética , Genoma Bacteriano/genética , Interações Hospedeiro-Patógeno
14.
Glycobiology ; 27(10): 978-989, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28922740

RESUMO

In Campylobacter jejuni, the PglB oligosaccharyltransferase catalyzes the transfer of a heptasaccharide from a lipid donor to asparagine within the D/E-X1-N-X2-S/T sequon (X1,2 ≠ P) or releases this heptasaccharide as free oligosaccharides (fOS). Using available crystal structures and sequence alignments, we identified a DGGK motif near the active site of PglB that is conserved among all Campylobacter species. We demonstrate that amino acid substitutions in the aspartate and lysine residues result in loss of protein glycosylation in the heterologous Escherichia coli system. Similarly, complementation of a C. jejuni pglB knock-out strain with mutated pglB alleles results in reduced levels of N-linked glycoproteins and fOS in the native host. Analysis of the PglB crystal structures from Campylobacter lari and the soluble C-terminal domain from C. jejuni suggests a particularly important structural role for the aspartate residue and the two following glycine residues, as well as a more subtle, less defined role for the lysine residue. Limited proteolysis experiments indicate that conformational changes of wildtype PglB that are induced by the binding of the lipid-linked oligosaccharide are altered by changes in the DGGK motif. Related to these findings, certain Campylobacter species possess two PglB orthologues and we demonstrate that only the orthologue containing the DGGK motif is active. Combining the knowledge gained from the PglB structures and mutagenesis studies, we propose a function for the DGGK motif in affecting the binding of the undecaprenyl-pyrophosphate glycan donor substrate that subsequently influences N-glycan and fOS production.


Assuntos
Proteínas de Bactérias/química , Campylobacter jejuni/enzimologia , Sequência Conservada , Glicosiltransferases/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Glicosiltransferases/metabolismo , Lipopolissacarídeos/química , Simulação de Acoplamento Molecular , Mutação , Polissacarídeos/química , Polissacarídeos/metabolismo , Ligação Proteica
15.
Mol Microbiol ; 101(4): 575-89, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27145048

RESUMO

Campylobacter jejuni and Campylobacter coli are zoonotic pathogens once considered asaccharolytic, but are now known to encode pathways for glucose and fucose uptake/metabolism. For C. jejuni, strains with the fuc locus possess a competitive advantage in animal colonization models. We demonstrate that this locus is present in > 50% of genome-sequenced strains and is prevalent in livestock-associated isolates of both species. To better understand how these campylobacters sense nutrient availability, we examined biofilm formation and chemotaxis to fucose. C. jejuni NCTC11168 forms less biofilms in the presence of fucose, although its fucose permease mutant (fucP) shows no change. In a newly developed chemotaxis assay, both wild-type and the fucP mutant are chemotactic towards fucose. C. jejuni 81-176 naturally lacks the fuc locus and is unable to swim towards fucose. Transfer of the NCTC11168 locus into 81-176 activated fucose uptake and chemotaxis. Fucose chemotaxis also correlated with possession of the pathway for C. jejuni RM1221 (fuc+) and 81116 (fuc-). Systematic mutation of the NCTC11168 locus revealed that Cj0485 is necessary for fucose metabolism and chemotaxis. This study suggests that components for fucose chemotaxis are encoded within the fuc locus, but downstream signals only in fuc + strains, are involved in coordinating fucose availability with biofilm development.


Assuntos
Biofilmes/crescimento & desenvolvimento , Campylobacter jejuni/fisiologia , Fucose/metabolismo , Campylobacter coli/genética , Campylobacter coli/metabolismo , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Metabolismo dos Carboidratos , Quimiotaxia/fisiologia , Fucose/genética , Genótipo
16.
Sci Rep ; 6: 26511, 2016 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-27221144

RESUMO

Campylobacter jejuni is a predominant cause of human gastroenteritis worldwide. Source-attribution studies indicate that chickens are the main reservoir for infection, thus elimination of C. jejuni from poultry would significantly reduce the burden of human disease. We constructed glycoconjugate vaccines combining the conserved C. jejuni N-glycan with a protein carrier, GlycoTag, or fused to the Escherichia coli lipopolysaccharide-core. Vaccination of chickens with the protein-based or E. coli-displayed glycoconjugate showed up to 10-log reduction in C. jejuni colonization and induced N-glycan-specific IgY responses. Moreover, the live E. coli vaccine was cleared prior to C. jejuni challenge and no selection for resistant campylobacter variants was observed. Analyses of the chicken gut communities revealed that the live vaccine did not alter the composition or complexity of the microbiome, thus representing an effective and low-cost strategy to reduce C. jejuni in chickens and its subsequent entry into the food chain.


Assuntos
Vacinas Bacterianas , Infecções por Campylobacter , Campylobacter jejuni , Galinhas , Escherichia coli , Doenças das Aves Domésticas , Animais , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Infecções por Campylobacter/genética , Infecções por Campylobacter/imunologia , Infecções por Campylobacter/prevenção & controle , Infecções por Campylobacter/veterinária , Campylobacter jejuni/genética , Campylobacter jejuni/imunologia , Campylobacter jejuni/patogenicidade , Galinhas/imunologia , Galinhas/microbiologia , Escherichia coli/genética , Escherichia coli/imunologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/prevenção & controle
17.
Sci Rep ; 6: 24931, 2016 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-27103188

RESUMO

The World Health Organization has indicated that we are entering into a post-antibiotic era in which infections that were routinely and successfully treated with antibiotics can now be lethal due to the global dissemination of multidrug resistant strains. Conjugate vaccines are an effective way to create a long-lasting immune response against bacteria. However, these vaccines present many drawbacks such as slow development, high price, and batch-to-batch inconsistencies. Alternate approaches for vaccine development are urgently needed. Here we present a new vaccine consisting of glycoengineered outer membrane vesicles (geOMVs). This platform exploits the fact that the initial steps in the biosynthesis of most bacterial glycans are similar. Therefore, it is possible to easily engineer non-pathogenic Escherichia coli lab strains to produce geOMVs displaying the glycan of the pathogen of interest. In this work we demonstrate the versatility of this platform by showing the efficacy of geOMVs as vaccines against Streptococcus pneumoniae in mice, and against Campylobacter jejuni in chicken. This cost-effective platform could be employed to generate vaccines to prevent infections caused by a wide variety of microbial agents in human and animals.


Assuntos
Campylobacter jejuni/imunologia , Vesículas Extracelulares/imunologia , Vacinas Pneumocócicas/imunologia , Polissacarídeos/imunologia , Streptococcus pneumoniae/imunologia , Animais , Galinhas , Camundongos , Vacinas Pneumocócicas/administração & dosagem , Vacinas Pneumocócicas/isolamento & purificação , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/imunologia , Vacinas Conjugadas/isolamento & purificação
18.
Biopolymers ; 99(10): 772-83, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23749285

RESUMO

All Campylobacter species are capable of N-glycosylating their proteins and releasing the same oligosaccharides into the periplasm as free oligosaccharides (fOS). Previously, analysis of fOS production in Campylobacter required fOS derivatization or large culture volumes and several chromatography steps prior to fOS analysis. In this study, label-free fOS extraction and purification methods were developed and coupled with quantitative analysis techniques. Our method follows three simple steps: (1) fOS extraction from the periplasmic space, (2) fOS purification using silica gel chromatography followed by porous graphitized carbon purification and (3) fOS analysis and accurate quantitation using a combination of thin-layer chromatography, mass spectrometry, NMR, and high performance anion exchange chromatography with pulsed amperometric detection. We applied our techniques to analyze fOS from C. jejuni, C. lari, C. rectus, and C. fetus fetus that produce different fOS structures. We accurately quantified fOS in Campylobacter species that ranged from 7.80 (±0.84) to 49.82 (±0.46) nmoles per gram of wet cell pellet and determined that the C. jejuni fOS comprises 2.5% of the dry cell weight. In addition, a novel di-phosphorylated fOS species was identified in C. lari. This method provides a sensitive and quantitative method to investigate the genesis, biology and breakdown of fOS in the bacterial N-glycosylation systems.


Assuntos
Campylobacter jejuni , Oligossacarídeos , Proteínas de Bactérias/metabolismo , Glicosilação , Espectrometria de Massas , Dados de Sequência Molecular , Oligossacarídeos/química
19.
Infect Immun ; 81(5): 1674-82, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23460522

RESUMO

Campylobacter jejuni is the etiologic agent of human bacterial gastroenteritis worldwide. In contrast, despite heavy colonization, C. jejuni maintains a commensal mode of existence in chickens. The consumption of contaminated chicken products is thought to be the principal mode of C. jejuni transmission to the human population. C. jejuni harbors a system for N-linked protein glycosylation that has been well characterized and modifies more than 60 periplasmic and membrane-bound proteins. However, the precise role of this modification in the biology of C. jejuni remains unexplored. We hypothesized that the N-glycans protect C. jejuni surface proteins from the action of gut proteases. The C. jejuni pglB mutant, deficient in the expression of the oligosaccharyltransferase, exhibited reduced growth in medium supplemented with chicken cecal contents (CCC) compared with that of wild-type (WT) cells. Inactivation of the cecal proteases by heat treatment or with protease inhibitors completely restored bacterial viability and partially rescued bacterial growth. Physiological concentrations of trypsin, but not chymotrypsin, also reduced C. jejuni pglB mutant CFU. Live or dead staining indicated that CCC preferentially influenced C. jejuni growth as opposed to bacterial viability. We identified multiple chicken cecal proteases by mass fingerprinting. The use of protease inhibitors that target specific classes indicated that both metalloproteases and serine proteases were involved in the attenuated growth of the oligosaccharyltransferase mutant. In conclusion, protein N-linked glycosylation of surface proteins may enhance C. jejuni fitness by protecting bacterial proteins from cleavage due to gut proteases.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Campylobacter jejuni/fisiologia , Polissacarídeos Bacterianos/fisiologia , Animais , Infecções por Campylobacter/metabolismo , Infecções por Campylobacter/microbiologia , Ceco/microbiologia , Galinhas/microbiologia , Contagem de Colônia Microbiana , Glicosilação , Hexosiltransferases/deficiência , Hexosiltransferases/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Peptídeo Hidrolases/metabolismo , Doenças das Aves Domésticas/microbiologia
20.
J Biol Chem ; 288(10): 6912-20, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23329827

RESUMO

Protein glycosylation is widespread throughout all three domains of life. Bacterial protein N-glycosylation and its application to engineering recombinant glycoproteins continue to be actively studied. Here, we focus on advances made in the last 2 years, including the characterization of novel bacterial N-glycosylation pathways, examination of pathway enzymes and evolution, biological roles of protein modification in the native host, and exploitation of the N-glycosylation pathways to create novel vaccines and diagnostics.


Assuntos
Proteínas de Bactérias/metabolismo , Glicoproteínas/metabolismo , Periplasma/metabolismo , Transdução de Sinais , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Glicoproteínas/genética , Glicoproteínas/imunologia , Glicosilação , Humanos , Modelos Imunológicos , Mutação/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA