Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Microb Cell Fact ; 22(1): 159, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596672

RESUMEN

Conjugate vaccines produced either by chemical or biologically conjugation have been demonstrated to be safe and efficacious in protection against several deadly bacterial diseases. However, conjugate vaccine assembly and production have several shortcomings which hinders their wider availability. Here, we developed a tool, Mobile-element Assisted Glycoconjugation by Insertion on Chromosome, MAGIC, a novel biotechnological platform that overcomes the limitations of the current conjugate vaccine design method(s). As a model, we focused our design on a leading bioconjugation method using N-oligosaccharyltransferase (OTase), PglB. The installation of MAGIC led to at least twofold increase in glycoconjugate yield via MAGIC when compared to conventional N-OTase based bioconjugation method(s). Then, we improved MAGIC to (a) allow rapid installation of glycoengineering component(s), (b) omit the usage of antibiotics, (c) reduce the dependence on protein induction agents. Furthermore, we show the modularity of the MAGIC platform in performing glycoengineering in bacterial species that are less genetically tractable than the commonly used Escherichia coli. The MAGIC system promises a rapid, robust and versatile method to develop vaccines against serious bacterial pathogens. We anticipate the utility of the MAGIC platform could enhance vaccines production due to its compatibility with virtually any bioconjugation method, thus expanding vaccine biopreparedness toolbox.


Asunto(s)
Antibacterianos , Biotecnología , Vacunas Conjugadas , Escherichia coli/genética , Desarrollo de Vacunas
2.
Microb Cell Fact ; 21(1): 66, 2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35449016

RESUMEN

BACKGROUND: Glycoengineering, in the biotechnology workhorse bacterium, Escherichia coli, is a rapidly evolving field, particularly for the production of glycoconjugate vaccine candidates (bioconjugation). Efficient production of glycoconjugates requires the coordinated expression within the bacterial cell of three components: a carrier protein, a glycan antigen and a coupling enzyme, in a timely fashion. Thus, the choice of a suitable E. coli host cell is of paramount importance. Microbial chassis engineering has long been used to improve yields of chemicals and biopolymers, but its application to vaccine production is sparse. RESULTS: In this study we have engineered a family of 11 E. coli strains by the removal and/or addition of components rationally selected for enhanced expression of Streptococcus pneumoniae capsular polysaccharides with the scope of increasing yield of pneumococcal conjugate vaccines. Importantly, all strains express a detoxified version of endotoxin, a concerning contaminant of therapeutics produced in bacterial cells. The genomic background of each strain was altered using CRISPR in an iterative fashion to generate strains without antibiotic markers or scar sequences. CONCLUSIONS: Amongst the 11 modified strains generated in this study, E. coli Falcon, Peregrine and Sparrowhawk all showed increased production of S. pneumoniae serotype 4 capsule. Eagle (a strain without enterobacterial common antigen, containing a GalNAc epimerase and PglB expressed from the chromosome) and Sparrowhawk (a strain without enterobacterial common antigen, O-antigen ligase and chain length determinant, containing a GalNAc epimerase and chain length regulators from Streptococcus pneumoniae) respectively produced an AcrA-SP4 conjugate with 4 × and 14 × more glycan than that produced in the base strain, W3110. Beyond their application to the production of pneumococcal vaccine candidates, the bank of 11 new strains will be an invaluable resource for the glycoengineering community.


Asunto(s)
Escherichia coli , Glicoconjugados , Vacunas Bacterianas/genética , Escherichia coli/metabolismo , Glicoconjugados/metabolismo , Polisacáridos/metabolismo , Polisacáridos Bacterianos/metabolismo , Racemasas y Epimerasas/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Vacunas Conjugadas
3.
Microb Cell Fact ; 21(1): 6, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34986868

RESUMEN

BACKGROUND: Campylobacter is an animal and zoonotic pathogen of global importance, and a pressing need exists for effective vaccines, including those that make use of conserved polysaccharide antigens. To this end, we adapted Protein Glycan Coupling Technology (PGCT) to develop a versatile Escherichia coli strain capable of generating multiple glycoconjugate vaccine candidates against Campylobacter jejuni. RESULTS: We generated a glycoengineering E. coli strain containing the conserved C. jejuni heptasaccharide coding region integrated in its chromosome as a model glycan. This methodology confers three advantages: (i) reduction of plasmids and antibiotic markers used for PGCT, (ii) swift generation of many glycan-protein combinations and consequent rapid identification of the most antigenic proteins or peptides, and (iii) increased genetic stability of the polysaccharide coding-region. In this study, by using the model glycan expressing strain, we were able to test proteins from C. jejuni, Pseudomonas aeruginosa (both Gram-negative), and Clostridium perfringens (Gram-positive) as acceptors. Using this pgl integrant E. coli strain, four glycoconjugates were readily generated. Two glycoconjugates, where both protein and glycan are from C. jejuni (double-hit vaccines), and two glycoconjugates, where the glycan antigen is conjugated to a detoxified toxin from a different pathogen (single-hit vaccines). Because the downstream application of Live Attenuated Vaccine Strains (LAVS) against C. jejuni is to be used in poultry, which have a higher body temperature of 42 °C, we investigated the effect of temperature on protein expression and glycosylation in the E. coli pgl integrant strain. CONCLUSIONS: We determined that glycosylation is temperature dependent and that for the combination of heptasaccharide and carriers used in this study, the level of PglB available for glycosylation is a step limiting factor in the glycosylation reaction. We also demonstrated that temperature affects the ability of PglB to glycosylate its substrates in an in vitro glycosylation assay independent of its transcriptional level.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cromosomas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Glicoconjugados/metabolismo , Temperatura , Proteínas Bacterianas/genética , Vacunas Bacterianas , Campylobacter jejuni/genética , Campylobacter jejuni/inmunología , Glicosilación , Proteínas de la Membrana/genética , Ingeniería Metabólica/métodos , Polisacáridos Bacterianos/genética
4.
Microb Cell Fact ; 20(1): 193, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34600535

RESUMEN

BACKGROUND: Poultry is the world's most popular animal-based food and global production has tripled in the past 20 years alone. Low-cost vaccines that can be combined to protect poultry against multiple infections are a current global imperative. Glycoconjugate vaccines, which consist of an immunogenic protein covalently coupled to glycan antigens of the targeted pathogen, have a proven track record in human vaccinology, but have yet to be used for livestock due to prohibitively high manufacturing costs. To overcome this, we use Protein Glycan Coupling Technology (PGCT), which enables the production of glycoconjugates in bacterial cells at considerably reduced costs, to generate a candidate glycan-based live vaccine intended to simultaneously protect against Campylobacter jejuni, avian pathogenic Escherichia coli (APEC) and Clostridium perfringens. Campylobacter is the most common cause of food poisoning, whereas colibacillosis and necrotic enteritis are widespread and devastating infectious diseases in poultry. RESULTS: We demonstrate the functional transfer of C. jejuni protein glycosylation (pgl) locus into the genome of APEC χ7122 serotype O78:H9. The integration caused mild attenuation of the χ7122 strain following oral inoculation of chickens without impairing its ability to colonise the respiratory tract. We exploit the χ7122 pgl integrant as bacterial vectors delivering a glycoprotein decorated with the C. jejuni heptasaccharide glycan antigen. To this end we engineered χ7122 pgl to express glycosylated NetB toxoid from C. perfringens and tested its ability to reduce caecal colonisation of chickens by C. jejuni and protect against intra-air sac challenge with the homologous APEC strain. CONCLUSIONS: We generated a candidate glycan-based multivalent live vaccine with the potential to induce protection against key avian and zoonotic pathogens (C. jejuni, APEC, C. perfringens). The live vaccine failed to significantly reduce Campylobacter colonisation under the conditions tested but was protective against homologous APEC challenge. Nevertheless, we present a strategy towards the production of low-cost "live-attenuated multivalent vaccine factories" with the ability to express glycoconjugates in poultry.


Asunto(s)
Infecciones por Campylobacter/prevención & control , Infecciones por Clostridium/prevención & control , Infecciones por Escherichia coli/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Desarrollo de Vacunas/métodos , Animales , Campylobacter jejuni/inmunología , Pollos , Clostridium perfringens/inmunología , Escherichia coli/inmunología , Vacunas Atenuadas/inmunología , Vacunas Combinadas/inmunología
5.
J Bacteriol ; 202(9)2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32015149

RESUMEN

In Escherichia coli, citrate-mediated iron transport is a key nonheme pathway for the acquisition of iron. Binding of ferric citrate to the outer membrane protein FecA induces a signal cascade that ultimately activates the cytoplasmic sigma factor FecI, resulting in transcription of the fecABCDE ferric citrate transport genes. Central to this process is signal transduction mediated by the inner membrane protein FecR. FecR spans the inner membrane through a single transmembrane helix, which is flanked by cytoplasm- and periplasm-orientated moieties at the N and C termini. The transmembrane helix of FecR resembles a twin-arginine signal sequence, and the substitution of the paired arginine residues of the consensus motif decouples the FecR-FecI signal cascade, rendering the cells unable to activate transcription of the fec operon when grown on ferric citrate. Furthermore, the fusion of beta-lactamase C-terminal to the FecR transmembrane helix results in translocation of the C-terminal domain that is dependent on the twin-arginine translocation (Tat) system. Our findings demonstrate that FecR belongs to a select group of bitopic inner membrane proteins that contain an internal twin-arginine signal sequence.IMPORTANCE Iron is essential for nearly all living organisms due to its role in metabolic processes and as a cofactor for many enzymes. The FecRI signal transduction pathway regulates citrate-mediated iron import in many Gram-negative bacteria, including Escherichia coli The interactions of FecR with the outer membrane protein FecA and cytoplasmic anti-sigma factor FecI have been extensively studied. However, the mechanism by which FecR inserts into the membrane has not previously been reported. In this study, we demonstrate that the targeting of FecR to the cytoplasmic membrane is dependent on the Tat system. As such, FecR represents a new class of bitopic Tat-dependent membrane proteins with an internal twin-arginine signal sequence.


Asunto(s)
Sistemas de Secreción Bacterianos/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Compuestos Férricos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Factor sigma/metabolismo , Sistemas de Secreción Bacterianos/genética , Membrana Celular/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Proteínas de Transporte de Membrana/genética , Transporte de Proteínas , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factor sigma/genética
6.
BMC Vet Res ; 15(1): 6, 2019 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-30606265

RESUMEN

BACKGROUND: Actinobacillus pleuropneumoniae is the causative agent of porcine pleuropneumonia and represents a major burden to the livestock industry. Virulence can largely be attributed to the secretion of a series of haemolytic toxins, which are highly immunogenic. A. pleuropneumoniae also encodes a cytoplasmic N-glycosylation system, which involves the modification of high molecular weight adhesins with glucose residues. Central to this process is the soluble N-glycosyl transferase, ngt, which is encoded in an operon with a subsequent glycosyl transferase, agt. Plasmid-borne recombinant expression of these genes in E. coli results in the production of a glucose polymer on peptides containing the appropriate acceptor sequon, NX(S/T). However to date, there is little evidence to suggest that such a glucose polymer is formed on its target peptides in A. pleuropneumoniae. Both the toxins and glycosylation system represent potential targets for the basis of a vaccine against A. pleuropneumoniae infection. RESULTS: In this study, we developed cytoplasmic glycoengineering to construct glycoconjugate vaccine candidates composed of soluble toxin fragments modified by glucose. We transferred ngt and agt to the chromosome of Escherichia coli in order to generate a native-like operon for glycoengineering. A single chromosomal copy of ngt and agt resulted in the glucosylation of toxin fragments by a short glycan, rather than a polymer. CONCLUSIONS: A vaccine candidate that combines toxin fragment with a conserved glycan offers a novel approach to generating epitopes important for both colonisation and disease progression.


Asunto(s)
Infecciones por Actinobacillus/veterinaria , Actinobacillus pleuropneumoniae/inmunología , Toxinas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Actinobacillus/inmunología , Infecciones por Actinobacillus/prevención & control , Animales , Escherichia coli/genética , Ingeniería Genética/métodos , Ingeniería Genética/veterinaria , Glicoconjugados/genética , Glicoconjugados/inmunología , Microorganismos Modificados Genéticamente/genética , Pleuroneumonía/inmunología , Pleuroneumonía/prevención & control , Pleuroneumonía/veterinaria , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/prevención & control , Vacunas Conjugadas/inmunología
7.
Bioorg Med Chem ; 26(11): 2973-2983, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29602676

RESUMEN

Lipooligosaccharide (LOS) structures in the outer core of Gram-negative mucosal pathogens such as Neisseria meningitidis and Haemophilus influenzae contain characteristic glycoepitopes that contribute significantly to bacterial virulence. An important example is the digalactoside epitope generated by the retaining α-1,4-galactosyltransferase LgtC. These digalactosides camouflage the pathogen from the host immune system and increase its serum resistance. Small molecular inhibitors of LgtC are therefore sought after as chemical tools to study bacterial virulence, and as potential candidates for anti-virulence drug discovery. We have recently discovered a new class of non-substrate-like inhibitors of LgtC. The new inhibitors act via a covalent mode of action, targeting a non-catalytic cysteine residue in the LgtC active site. Here, we describe, for the first time, structure-activity relationships for this new class of glycosyltransferase inhibitors. We have carried out a detailed analysis of the inhibition kinetics to establish the relative contribution of the non-covalent binding and the covalent inactivation steps for overall inhibitory activity. Selected inhibitors were also evaluated against a serum-resistant strain of Haemophilus influenzae, but did not enhance the killing effect of human serum.


Asunto(s)
Inhibidores Enzimáticos/química , Glicosiltransferasas/antagonistas & inhibidores , Neisseria meningitidis/enzimología , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Glicosiltransferasas/metabolismo , Haemophilus influenzae/efectos de los fármacos , Cinética , Neisseria meningitidis/efectos de los fármacos , Pirazoles/síntesis química , Pirazoles/química , Pirazoles/farmacología , Relación Estructura-Actividad
8.
Glycobiology ; 26(4): 398-409, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26610891

RESUMEN

Bacterial N-linking oligosaccharyl transferases (OTase enzymes) transfer lipid-linked glycans to selected proteins in the periplasm and were first described in the intestinal pathogen Campylobacter jejuni, a member of the ε-proteobacteria-subdivision of bacteria. More recently, orthologues from other ε-proteobacterial Campylobacter and Helicobacter species and a δ-proteobacterium, Desulfovibrio desulfuricans, have been described, suggesting that these two subdivisions of bacteria may be a source of further N-linked protein glycosylation systems. Whole-genome sequencing of both ε- and δ-proteobacteria from deep-sea vent habitats, a rich source of species from these subdivisions, revealed putative ORFs encoding OTase enzymes and associated adjacent glycosyltransferases similar to the C. jejuni N-linked glycosylation locus. We expressed putative OTase ORFs from the deep-sea vent species Nitratiruptor tergarcus, Sulfurovum lithotrophicum and Deferribacter desulfuricans in Escherichia coli and showed that they were able to functionally complement the C. jejuni OTase, CjPglB. The enzymes were shown to possess relaxed glycan specificity, transferring diverse glycan structures and demonstrated different glycosylation sequon specificities. Additionally, a permissive D. desulfuricans acceptor protein was identified, and we provide evidence that the N-linked glycan synthesized by N. tergarcus and S. lithotrophicum contains an acetylated sugar at the reducing end. This work demonstrates that deep-sea vent bacteria encode functional N-glycosylation machineries and are a potential source of biotechnologically important OTase enzymes.


Asunto(s)
Hexosiltransferasas/genética , Proteínas de la Membrana/genética , Polisacáridos/metabolismo , Proteobacteria/genética , Escherichia coli/genética , Genoma Bacteriano , Glicosilación , Hexosiltransferasas/biosíntesis , Hexosiltransferasas/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/metabolismo , Océanos y Mares , Polisacáridos/biosíntesis , Proteobacteria/enzimología , Especificidad por Sustrato
9.
Infect Immun ; 84(3): 701-10, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26712202

RESUMEN

Burkholderia pseudomallei, the causative agent of melioidosis, has complex and poorly understood extracellular and intracellular lifestyles. We used transposon-directed insertion site sequencing (TraDIS) to retrospectively analyze a transposon library that had previously been screened through a BALB/c mouse model to identify genes important for growth and survival in vivo. This allowed us to identify the insertion sites and phenotypes of negatively selected mutants that were previously overlooked due to technical constraints. All 23 unique genes identified in the original screen were confirmed by TraDIS, and an additional 105 mutants with various degrees of attenuation in vivo were identified. Five of the newly identified genes were chosen for further characterization, and clean, unmarked bpsl2248, tex, rpiR, bpsl1728, and bpss1528 deletion mutants were constructed from the wild-type strain K96243. Each of these mutants was tested in vitro and in vivo to confirm their attenuated phenotypes and investigate the nature of the attenuation. Our results confirm that we have identified new genes important to in vivo virulence with roles in different stages of B. pseudomallei pathogenesis, including extracellular and intracellular survival. Of particular interest, deletion of the transcription accessory protein Tex was shown to be highly attenuating, and the tex mutant was capable of providing protective immunity against challenge with wild-type B. pseudomallei, suggesting that the genes identified in our TraDIS screen have the potential to be investigated as live vaccine candidates.


Asunto(s)
Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/crecimiento & desarrollo , Burkholderia pseudomallei/metabolismo , Melioidosis/microbiología , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Burkholderia pseudomallei/genética , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Viabilidad Microbiana , Factores de Virulencia/genética
10.
Microbiology (Reading) ; 161(Pt 2): 387-400, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25414045

RESUMEN

Actinobacillus pleuropneumoniae is responsible for swine pleuropneumonia, a respiratory disease that causes significant global economic loss. Its virulence depends on many factors, such as capsular polysaccharides, RTX toxins and iron-acquisition systems. Analysis of virulence may require easy-to-use models that approximate mammalian infection and avoid ethical issues. Here, we investigate the potential use of the wax moth Galleria mellonella as an informative model for A. pleuropneumoniae infection. Genotypically distinct A. pleuropneumoniae clinical isolates were able to kill larvae at 37 °C but had different LD50 values, ranging from 10(4) to 10(7) c.f.u. per larva. The most virulent isolate (1022) was able to persist and replicate within the insect, while the least virulent (780) was rapidly cleared. We observed a decrease in haemocyte concentration, aggregation and DNA damage post-infection with isolate 1022. Melanization points around bacterial cells were observed in the fat body and pericardial tissues of infected G. mellonella, indicating vigorous cell and humoral immune responses close to the larval dorsal vessel. As found in pigs, an A. pleuropneumoniae hfq mutant was significantly attenuated for infection in the G. mellonella model. Additionally, the model could be used to assess the effectiveness of several antimicrobial agents against A. pleuropneumoniae in vivo. G. mellonella is a suitable inexpensive alternative infection model that can be used to study the virulence of A. pleuropneumoniae, as well as assess the effectiveness of antimicrobial agents against this pathogen.


Asunto(s)
Infecciones por Actinobacillus/microbiología , Actinobacillus pleuropneumoniae/fisiología , Modelos Animales de Enfermedad , Mariposas Nocturnas/microbiología , Actinobacillus pleuropneumoniae/genética , Actinobacillus pleuropneumoniae/patogenicidad , Animales , Humanos , Larva/microbiología , Virulencia
11.
Mol Microbiol ; 89(5): 811-5, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23844892

RESUMEN

Protein glycosylation and capsular polysaccharide formation are increasingly recognized as playing central roles in the survival and virulence of bacterial pathogens. In this issue of Molecular Microbiology, structural analysis in Acinetobacter baumannii 17978 revealed that a pentasaccharide that decorates glycoproteins is formed of the same building blocks used for capsule biosynthesis demonstrating split roles for this glycan. Disruption of PglC, the initiating glycosyltransferase responsible for attachment of the first sugar to undecaprenylphosphate abolished glycoprotein production and capsule biosynthesis. Both pathways are demonstrated to be important in biofilm formation and pathogenesis, and disabling their synthesis should provide a useful route for antimicrobial design. Shared polysaccharide usage reduces the genetic and metabolic burden in a bacterial cell and is an emerging theme among bacterial pathogens that need to be energy efficient for their streamlined lifestyle.


Asunto(s)
Acinetobacter baumannii/metabolismo , Cápsulas Bacterianas/metabolismo , Redes y Vías Metabólicas/genética , Procesamiento Proteico-Postraduccional , Animales
12.
Microb Biotechnol ; 17(6): e14480, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38858807

RESUMEN

The application of bacterial oligosaccharyltransferases (OSTs) such as the Campylobacter jejuni PglB for glycoengineering has attracted considerable interest in glycoengineering and glycoconjugate vaccine development. However, PglB has limited specificity for glycans that can be transferred to candidate proteins, which along with other factors is dependent on the reducing end sugar of glycans. In this study, we developed a cell-free glycosylation assay that offers the speed and simplicity of a 'yes' or 'no' determination. Using the assay, we tested the activity of eleven PglBs from Campylobacter species and more distantly related bacteria. The following assorted glycans with diverse reducing end sugars were tested for transfer, including Streptococcus pneumoniae capsule serotype 4, Salmonella enterica serovar Typhimurium O antigen (B1), Francisella tularensis O antigen, Escherichia coli O9 antigen and Campylobacter jejuni heptasaccharide. Interestingly, while PglBs from the same genus showed high activity, whereas divergent PglBs differed in their transfer of glycans to an acceptor protein. Notably for glycoengineering purposes, Campylobacter hepaticus and Campylobacter subantarcticus PglBs showed high glycosylation efficiency, with C. hepaticus PglB potentially being useful for glycoconjugate vaccine production. This study demonstrates the versatility of the cell-free assay in rapidly assessing an OST to couple glycan/carrier protein combinations and lays the foundation for future screening of PglBs by linking amino acid similarity to glycosyltransferase activity.


Asunto(s)
Hexosiltransferasas , Proteínas de la Membrana , Hexosiltransferasas/genética , Hexosiltransferasas/metabolismo , Hexosiltransferasas/química , Glicosilación , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Campylobacter/genética , Campylobacter/enzimología , Campylobacter/metabolismo , Polisacáridos/metabolismo , Sistema Libre de Células , Campylobacter jejuni/enzimología , Campylobacter jejuni/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Glicoconjugados/metabolismo
13.
Vaccines (Basel) ; 12(4)2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38675777

RESUMEN

Campylobacter is a major cause of acute gastroenteritis in humans, and infections can be followed by inflammatory neuropathies and other sequelae. Handling or consumption of poultry meat is the primary risk factor for human campylobacteriosis, and C. jejuni remains highly prevalent in retail chicken in many countries. Control of Campylobacter in the avian reservoir is expected to limit the incidence of human disease. Toward this aim, we evaluated a glycoconjugate vaccine comprising the fibronectin-binding adhesin FlpA conjugated to up to ten moieties of the conserved N-linked heptasaccharide glycan of C. jejuni or with FlpA alone. The glycan dose significantly exceeded previous trials using FlpA with two N-glycan moieties. Vaccinated birds were challenged with C. jejuni orally or by exposure to seeder-birds colonised by C. jejuni to mimic natural transmission. No protection against caecal colonisation was observed with FlpA or the FlpA glycoconjugate vaccine. FlpA-specific antibody responses were significantly induced in vaccinated birds at the point of challenge relative to mock-vaccinated birds. A slight but significant antibody response to the N-glycan was detected after vaccination with FlpA-10×GT and challenge. As other laboratories have reported protection against Campylobacter with FlpA and glycoconjugate vaccines in chickens, our data indicate that vaccine-mediated immunity may be sensitive to host- or study-specific variables.

14.
Antimicrob Agents Chemother ; 57(3): 1513-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23263008

RESUMEN

Emerging resistance to current antibiotics raises the need for new microbial drug targets. We show that targeting branched-chain amino acid (BCAA) biosynthesis using sulfonylurea herbicides, which inhibit the BCAA biosynthetic enzyme acetohydroxyacid synthase (AHAS), can exert bacteriostatic effects on several pathogenic bacteria, including Burkholderia pseudomallei, Pseudomonas aeruginosa, and Acinetobacter baumannii. Our results suggest that targeting biosynthetic enzymes like AHAS, which are lacking in humans, could represent a promising antimicrobial drug strategy.


Asunto(s)
Acetolactato Sintasa/antagonistas & inhibidores , Acinetobacter baumannii/efectos de los fármacos , Proteínas Bacterianas/antagonistas & inhibidores , Burkholderia pseudomallei/efectos de los fármacos , Herbicidas/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Compuestos de Sulfonilurea/farmacología , Acetolactato Sintasa/metabolismo , Acinetobacter baumannii/enzimología , Acinetobacter baumannii/crecimiento & desarrollo , Aminoácidos de Cadena Ramificada/antagonistas & inhibidores , Aminoácidos de Cadena Ramificada/biosíntesis , Animales , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/enzimología , Burkholderia pseudomallei/crecimiento & desarrollo , Femenino , Melioidosis/tratamiento farmacológico , Melioidosis/microbiología , Melioidosis/mortalidad , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/mortalidad , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/crecimiento & desarrollo , Análisis de Supervivencia
15.
Infect Immun ; 80(11): 3921-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22927051

RESUMEN

Burkholderia pseudomallei is the causative pathogen of melioidosis, of which a major predisposing factor is diabetes mellitus. Polymorphonuclear neutrophils (PMNs) kill microbes extracellularly by the release of neutrophil extracellular traps (NETs). PMNs play a key role in the control of melioidosis, but the involvement of NETs in killing of B. pseudomallei remains obscure. Here, we showed that bactericidal NETs were released from human PMNs in response to B. pseudomallei in a dose- and time-dependent manner. B. pseudomallei-induced NET formation required NADPH oxidase activation but not phosphatidylinositol-3 kinase, mitogen-activated protein kinases, or Src family kinase signaling pathways. B. pseudomallei mutants defective in the virulence-associated Bsa type III protein secretion system (T3SS) or capsular polysaccharide I (CPS-I) induced elevated levels of NETs. NET induction by such mutants was associated with increased bacterial killing, phagocytosis, and oxidative burst by PMNs. Taken together the data imply that T3SS and the capsule may play a role in evading the induction of NETs. Importantly, PMNs from diabetic subjects released NETs at a lower level than PMNs from healthy subjects. Modulation of NET formation may therefore be associated with the pathogenesis and control of melioidosis.


Asunto(s)
Burkholderia pseudomallei/inmunología , Melioidosis/inmunología , Neutrófilos/inmunología , Estallido Respiratorio/inmunología , Anticuerpos Antibacterianos/análisis , Proteínas Bacterianas , Burkholderia pseudomallei/patogenicidad , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Viabilidad Microbiana , Fagocitosis , Virulencia , Factores de Virulencia
16.
Infect Immun ; 80(3): 1209-21, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22252864

RESUMEN

Burkholderia pseudomallei is the causative agent of melioidosis, a disease endemic to regions of Southeast Asia and Northern Australia. Both humans and a range of other animal species are susceptible to melioidosis, and the production of a group 3 polysaccharide capsule in B. pseudomallei is essential for virulence. B. pseudomallei capsular polysaccharide (CPS) I comprises unbranched manno-heptopyranose residues and is encoded by a 34.5-kb locus on chromosome 1. Despite the importance of this locus, the role of all of the genes within this region is unclear. We inactivated 18 of these genes and analyzed their phenotype using Western blotting and immunofluorescence staining. Furthermore, by combining this approach with bioinformatic analysis, we were able to develop a model for CPS I biosynthesis and export. We report that inactivating gmhA, wcbJ, and wcbN in B. pseudomallei K96243 retains the immunogenic integrity of the polysaccharide despite causing attenuation in the BALB/c murine infection model. Mice immunized with the B. pseudomallei K96243 mutants lacking a functional copy of either gmhA or wcbJ were afforded significant levels of protection against a wild-type B. pseudomallei K96243 challenge.


Asunto(s)
Vías Biosintéticas/genética , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/metabolismo , Polisacáridos Bacterianos/genética , Polisacáridos Bacterianos/metabolismo , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Western Blotting , Burkholderia pseudomallei/inmunología , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Sitios Genéticos , Melioidosis/microbiología , Melioidosis/patología , Ratones , Ratones Endogámicos BALB C , Familia de Multigenes , Polisacáridos Bacterianos/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo
17.
Front Vet Sci ; 8: 686088, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34150901

RESUMEN

The recent increase in new technologies to analyze host-pathogen interaction has fostered a race to develop new methodologies to assess these not only on the cellular level, but also on the tissue level. Due to mouse-other mammal differences, there is a desperate need to develop relevant tissue models that can more closely recapitulate the host tissue during disease and repair. Whereas organoids and organs-on-a-chip technologies have their benefits, they still cannot provide the cellular and structural complexity of the host tissue. Here, precision cut tissue slices (PCTS) may provide invaluable models for complex ex-vivo generated tissues to assess host-pathogen interaction as well as potential vaccine responses in a "whole organ" manner. In this mini review, we discuss the current literature regarding PCTS in veterinary species and advocate that PCTS represent remarkable tools to further close the gap between target identification, subsequent translation of results into clinical studies, and thus opening avenues for future precision medicine approaches.

18.
Vaccine ; 39(51): 7413-7420, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34799141

RESUMEN

Campylobacter jejuni is the leading bacterial cause of human gastroenteritis worldwide and handling or consumption of contaminated poultry meat is the key source of infection. Glycoconjugate vaccines containing the C. jejuni N-glycan have been reported to be partially protective in chickens. However, our previous studies with subunit vaccines comprising the C. jejuni FlpA or SodB proteins with up to two or three C. jejuni N-glycans, respectively, failed to elicit significant protection. In this study, protein glycan coupling technology was used to add up to ten C. jejuni N-glycans onto a detoxified form of Pseudomonas aeruginosa exotoxin A (ExoA). The glycoprotein, G-ExoA, was evaluated for efficacy against intestinal colonisation of White Leghorn chickens by C. jejuni strains M1 and 11168H relative to unglycosylated ExoA. Chickens were challenged with the minimum dose required for reliable colonisation, which was 102 colony-forming units (CFU) for strain M1 and and 104 CFU for strain 11168H. Vaccine-specific serum IgY was detected in chickens vaccinated with both ExoA and G-ExoA. However, no reduction in caecal colonisation by C. jejuni was observed. While the glycan dose achieved with G-ExoA was higher than FlpA- or SodB-based glycoconjugates that were previously evaluated, it was lower than that of glycoconjugates where protection against C. jejuni has been reported, indicating that protection may be highly sensitive to the amount of glycan presented and/or study-specific variables.


Asunto(s)
Infecciones por Campylobacter , Campylobacter jejuni , Enfermedades de las Aves de Corral , Animales , Infecciones por Campylobacter/prevención & control , Infecciones por Campylobacter/veterinaria , Pollos , Glicoconjugados , Humanos , Polisacáridos , Enfermedades de las Aves de Corral/prevención & control , Vacunas de Subunidad
19.
BMC Microbiol ; 10: 171, 2010 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-20540813

RESUMEN

BACKGROUND: Burkholderia pseudomallei is the causative agent of melioidosis where the highest reported incidence world wide is in the Northeast of Thailand, where saline soil and water are prevalent. Moreover, recent reports indicate a potential pathogenic role for B. pseudomallei in cystic fibrosis lung disease, where an increased sodium chloride (NaCl) concentration in airway surface liquid has been proposed. These observations raise the possibility that high salinity may represent a favorable niche for B. pseudomallei. We therefore investigated the global transcriptional response of B. pseudomallei to increased salinity using microarray analysis. RESULTS: Transcriptome analysis of B. pseudomallei under salt stress revealed several genes significantly up-regulated in the presence of 320 mM NaCl including genes associated with the bsa-derived Type III secretion system (T3SS). Microarray data were verified by reverse transcriptase-polymerase chain reactions (RT-PCR). Western blot analysis confirmed the increased expression and secretion of the invasion-associated type III secreted proteins BipD and BopE in B. pseudomallei cultures at 170 and 320 mM NaCl relative to salt-free medium. Furthermore, salt-treated B. pseudomallei exhibited greater invasion efficiency into the lung epithelial cell line A549 in a manner partly dependent on a functional Bsa system. CONCLUSIONS: B. pseudomallei responds to salt stress by modulating the transcription of a relatively small set of genes, among which is the bsa locus associated with invasion and virulence. Expression and secretion of Bsa-secreted proteins was elevated in the presence of exogenous salt and the invasion efficiency was enhanced. Our data indicate that salinity has the potential to influence the virulence of B. pseudomallei.


Asunto(s)
Burkholderia pseudomallei/efectos de los fármacos , Burkholderia pseudomallei/metabolismo , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Transcripción Genética , Burkholderia pseudomallei/genética , Medios de Cultivo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cloruro de Sodio , Estrés Fisiológico
20.
mBio ; 11(6)2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203757

RESUMEN

The substantial rise in multidrug-resistant bacterial infections is a current global imperative. Cumulative efforts to characterize antimicrobial resistance in bacteria has demonstrated the spread of six families of multidrug efflux pumps, of which resistance-nodulation-cell division (RND) is the major mechanism of multidrug resistance in Gram-negative bacteria. RND is composed of a tripartite protein assembly and confers resistance to a range of unrelated compounds. In the major enteric pathogen Campylobacter jejuni, the three protein components of RND are posttranslationally modified with N-linked glycans. The direct role of N-linked glycans in C. jejuni and other bacteria has long been elusive. Here, we present the first detailed account of the role of N-linked glycans and the link between N-glycosylation and antimicrobial resistance in C. jejuni We demonstrate the multifunctional role of N-linked glycans in enhancing protein thermostability, stabilizing protein complexes and the promotion of protein-protein interaction, thus mediating antimicrobial resistance via enhancing multidrug efflux pump activity. This affirms that glycosylation is critical for multidrug efflux pump assembly. We present a generalized strategy that could be used to investigate general glycosylation system in Campylobacter genus and a potential target to develop antimicrobials against multidrug-resistant pathogens.IMPORTANCE Nearly all bacterial species have at least a single glycosylation system, but the direct effects of these posttranslational protein modifications are unresolved. Glycoproteome-wide analysis of several bacterial pathogens has revealed general glycan modifications of virulence factors and protein assemblies. Using Campylobacter jejuni as a model organism, we have studied the role of general N-linked glycans in the multidrug efflux pump commonly found in Gram-negative bacteria. We show, for the first time, the direct link between N-linked glycans and multidrug efflux pump activity. At the protein level, we demonstrate that N-linked glycans play a role in enhancing protein thermostability and mediating the assembly of the multidrug efflux pump to promote antimicrobial resistance, highlighting the importance of this posttranslational modification in bacterial physiology. Similar roles for glycans are expected to be found in other Gram-negative pathogens that possess general protein glycosylation systems.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Antibacterianos/farmacología , Infecciones por Campylobacter/microbiología , Campylobacter/fisiología , Farmacorresistencia Bacteriana Múltiple , Procesamiento Proteico-Postraduccional , Proteoma , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Campylobacter/efectos de los fármacos , Campylobacter/genética , Campylobacter jejuni/efectos de los fármacos , Campylobacter jejuni/genética , Campylobacter jejuni/fisiología , Glicosilación , Factores de Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA