Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Microb Pathog ; 193: 106752, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38880315

RESUMO

Arcobacter butzleri is a foodborne pathogen that mainly causes enteritis in humans, but the number of cases of bacteraemia has increased in recent years. However, there is still limited knowledge on the pathogenic mechanisms of this bacterium. To investigate how A. butzleri causes disease, single knockout mutants were constructed in the cadF, ABU_RS00335, ciaB, and flaAB genes, which might be involved in adhesion and invasion properties. These mutants and the isogenic wild-type (WT) were then tested for their ability to adhere and invade human Caco-2 and HT29-MTX cells. The adhesion and invasion of A. butzleri RM4018 strain was also visualized by a Leica CTR 6500 confocal microscope. The adhesion and invasion abilities of mutants lacking the invasion antigen CiaB or a functional flagellum were lower than those of the WTs. However, the extent of the decrease varied depending on the strain and/or cell line. Mutants lacking the fibronectin (FN)-binding protein CadF consistently exhibited reduced abilities, while the inactivation of the other studied FN-binding protein, ABU_RS00335, led to a reduction in only one of the two strains tested. Therefore, the ciaB and flaAB genes appear to be important for A. butzleri adhesion and invasion properties, while cadF appears to be indispensable.

2.
Glycobiology ; 32(1): 11-22, 2022 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-34939094

RESUMO

All bacteria display surface-exposed glycans that can play an important role in their interaction with the host and in select cases mimic the glycans found on host cells, an event called molecular or glycan mimicry. In this review, we highlight the key bacteria that display human glycan mimicry and provide an overview of the involved glycan structures. We also discuss the general trends and outstanding questions associated with human glycan mimicry by bacteria. Finally, we provide an overview of several techniques that have emerged from the discipline of chemical glycobiology, which can aid in the study of the composition, variability, interaction and functional role of these mimicking glycans.


Assuntos
Mimetismo Molecular , Polissacarídeos , Bactérias , Glicômica , Humanos , Polissacarídeos/química , Polissacarídeos Bacterianos
3.
Chembiochem ; 23(19): e202200340, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-35877976

RESUMO

The interactions between bacteria and their host often rely on recognition processes that involve host or bacterial glycans. Glycoengineering techniques make it possible to modify and study the glycans on the host's eukaryotic cells, but only a few are available for the study of bacterial glycans. Here, we have adapted selective exoenzymatic labeling (SEEL), a chemical reporter strategy, to label the lipooligosaccharides of the bacterial pathogen Neisseria gonorrhoeae, using the recombinant glycosyltransferase ST6Gal1, and three synthetic CMP-sialic acid derivatives. We show that SEEL treatment does not affect cell viability and can introduce an α2,6-linked sialic acid with a reporter group on the lipooligosaccharides by Western blot, flow cytometry and fluorescent microscopy. This new bacterial glycoengineering technique allows for the precise modification, here with α2,6-sialoside derivatives, and direct detection of specific surface glycans on live bacteria, which will aid in further unravelling the precise biological functions of bacterial glycans.


Assuntos
Ácido N-Acetilneuramínico do Monofosfato de Citidina , Neisseria gonorrhoeae , Ácido N-Acetilneuramínico do Monofosfato de Citidina/metabolismo , Glicosiltransferases/metabolismo , Lipopolissacarídeos , Ácido N-Acetilneuramínico , Polissacarídeos Bacterianos/metabolismo , Ácidos Siálicos/metabolismo
4.
Cell Microbiol ; 22(12): e13252, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32827216

RESUMO

The enteropathogenic bacterium, Campylobacter jejuni, was considered to be non-saccharolytic, but recently it emerged that l-fucose plays a central role in C. jejuni virulence. Half of C. jejuni clinical isolates possess an operon for l-fucose utilisation. In the intestinal tract, l-fucose is abundantly available in mucin O-linked glycan structures, but C. jejuni lacks a fucosidase enzyme essential to release the l-fucose. We set out to determine how C. jejuni can gain access to these intestinal l-fucosides. Growth of the fuc + C. jejuni strains, 129,108 and NCTC 11168, increased in the presence of l-fucose while fucose permease knockout strains did not benefit from additional l-fucose. With fucosidase assays and an activity-based probe, we confirmed that Bacteriodes fragilis, an abundant member of the intestinal microbiota, secretes active fucosidases. In the presence of mucins, C. jejuni was dependent on B. fragilis fucosidase activity for increased growth. Campylobacter jejuni invaded Caco-2 intestinal cells that express complex O-linked glycan structures that contain l-fucose. In infection experiments, C. jejuni was more invasive in the presence of B. fragilis and this increase is due to fucosidase activity. We conclude that C. jejuni fuc + strains are dependent on exogenous fucosidases for increased growth and invasion.


Assuntos
Bacteroides fragilis/enzimologia , Campylobacter jejuni/crescimento & desenvolvimento , Campylobacter jejuni/patogenicidade , Fucose/metabolismo , Mucinas/metabolismo , alfa-L-Fucosidase/metabolismo , Células CACO-2 , Campylobacter jejuni/genética , Humanos , Interações Microbianas/fisiologia , Virulência , alfa-L-Fucosidase/biossíntese
5.
Angew Chem Int Ed Engl ; 60(47): 24811-24816, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34519150

RESUMO

Campylobacter jejuni is the major human food-borne pathogen. Its bipolar flagella are heavily O-glycosylated with microbial sialic acids and essential for its motility and pathogenicity. However, both the glycosylation of flagella and the exact contribution of legionaminic acid (Leg) to flagellar activity is poorly understood. Herein, we report the development of a metabolic labeling method for Leg glycosylation on bacterial flagella with probes based on azide-modified Leg precursors. The hereby azido-Leg labeled flagellin could be detected by Western blot analysis and imaged on intact bacteria. Using the probes on C. jejuni and its isogenic maf4 mutant we also further substantiated the identification of Maf4 as a putative Leg glycosyltransferase. Further evidence was provided by UPLC-MS detection of labeled CMP-Leg and an in silico model of Maf4. This method and the developed probes will facilitate the study of Leg glycosylation and the functional role of this modification in C. jejuni motility and invasiveness.


Assuntos
Campylobacter jejuni/metabolismo , Flagelina/metabolismo , Ácidos Siálicos/metabolismo , Transferases/metabolismo , Campylobacter jejuni/química , Configuração de Carboidratos , Flagelina/química , Glicosilação , Humanos , Ácidos Siálicos/análise , Transferases/química
6.
Mol Microbiol ; 105(4): 637-651, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28586527

RESUMO

The generation of a membrane potential (Δψ), the major constituent of the proton motive force (pmf), is crucial for ATP synthesis, transport of nutrients and flagellar rotation. Campylobacter jejuni harbors a branched electron transport chain, enabling respiration with different electron donors and acceptors. Here, we demonstrate that a relatively high Δψ is only generated in the presence of either formate as electron donor or oxygen as electron acceptor, in combination with an acceptor/donor respectively. We show the necessity of the pmf for motility and growth of C. jejuni. ATP generation is not only accomplished by oxidative phosphorylation via the pmf, but also by substrate-level phosphorylation via the enzyme AckA. In response to a low oxygen tension, C. jejuni increases the transcription and activity of the donor complexes formate dehydrogenase (FdhABC) and hydrogenase (HydABCD) as well as the transcription of the alternative respiratory acceptor complexes. Our findings suggest that in the gut of warm-blooded animals, C. jejuni depends on at least formate or hydrogen as donor (in the anaerobic lumen) or oxygen as acceptor (near the epithelial cells) to generate a pmf that sustains efficient motility and growth for colonization and pathogenesis.


Assuntos
Campylobacter jejuni/metabolismo , Força Próton-Motriz/fisiologia , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , Formiatos/metabolismo , Hidrogênio , Potenciais da Membrana , Oxirredução , Oxigênio , Fosforilação
7.
Environ Microbiol ; 20(4): 1374-1388, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29318721

RESUMO

Bacteria have evolved different mechanisms to catabolize carbon sources from nutrient mixtures. They first consume their preferred carbon source, before others are used. Regulatory mechanisms adapt the metabolism accordingly to maximize growth and to outcompete other organisms. The human pathogen Campylobacter jejuni is an asaccharolytic Gram-negative bacterium that catabolizes amino acids and organic acids for growth. It prefers serine and aspartate as carbon sources, however it lacks all regulators known to be involved in regulating carbon source utilization in other organisms. In which manner C. jejuni adapts its metabolism towards the presence or absence of preferred carbon sources is unknown. In this study, we show with transcriptomic analysis and enzyme assays how C. jejuni adapts its metabolism in response to its preferred carbon sources. In the presence of serine as well as lactate and pyruvate C. jejuni inhibits the utilization of other carbon sources, by repressing the expression of a number of central metabolic enzymes. The regulatory proteins RacR, Cj1000 and CsrA play a role in the regulation of these metabolic enzymes. This metabolism dependent transcriptional repression correlates with an accumulation of intracellular succinate. Hence, we propose a demand-based catabolite repression mechanism in C. jejuni, depended on intracellular succinate levels.


Assuntos
Campylobacter jejuni/metabolismo , Repressão Catabólica/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Ácido Succínico/metabolismo , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/genética , Carbono/metabolismo , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/genética , Humanos , Ácido Láctico/metabolismo , Ácido Pirúvico/metabolismo , Serina/metabolismo , Fatores de Transcrição/metabolismo
8.
Mol Microbiol ; 102(2): 207-220, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27353476

RESUMO

Bacterial flagella assembly is tightly regulated to ensure a timely and sequential production of the various flagellum constituents. In the pathogen Campylobacter jejuni the hierarchy in flagella biosynthesis is largely determined at the transcriptional level through the activity of the alternative sigma factors sigma54 and sigma28 . Here, we report that C. jejuni flagellin levels are also controlled at the post-transcriptional level via the thus far poorly-characterized flagellar assembly factor FliW. Analysis of flagellin synthesis in C. jejuni 81116 and a ΔfliW knock-out mutant showed reduced flagellin protein levels in the mutant strain while ectopic expression of FliW resulted in enhanced levels. Real-time RT-PCR revealed relatively minor changes in flaA and flaB mRNA levels for the recombinant and parent strain consistent with post-transcriptional regulation. Purified FliW was found to bind to FlaA and FlaB flagellin as well as to the global post-transcriptional regulator CsrA. Inactivation of CsrA resulted in increased levels of flagellin translation. An in vitro translation assay confirmed the regulatory role of CsrA in flagellin biosynthesis. We propose that competitive reciprocal binding of FliW to flagellins and the RNA binding protein CsrA serves as a feedback mechanism to control the number of cytosolic flagellin copies at the protein level.


Assuntos
Campylobacter jejuni/metabolismo , Flagelina/metabolismo , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/genética , Retroalimentação Fisiológica , Flagelos/metabolismo , Flagelina/biossíntese , Regulação Bacteriana da Expressão Gênica/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Fator sigma/metabolismo
9.
Environ Microbiol ; 17(4): 1049-64, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24707969

RESUMO

The natural environment of the human pathogen Campylobacter jejuni is the gastrointestinal tract of warm-blooded animals. In the gut, the availability of oxygen is limited; therefore, less efficient electron acceptors such as nitrate or fumarate are used by C. jejuni. The molecular mechanisms that regulate the activity of the highly branched respiratory chain of C. jejuni are still a mystery mainly because C. jejuni lacks homologues of transcription factors known to regulate energy metabolism in other bacteria. Here we demonstrate that dependent on the available electron acceptors the two-component system RacRS controls the production of fumarate from aspartate, as well as its transport and reduction to succinate. Transcription profiling, DNAse protection and functional assays showed that phosphorylated RacR binds to and represses at least five promoter elements located in front of genes involved in the uptake and synthesis of fumarate. The RacRS system is active in the presence of nitrate and trimethyl-amine-N-oxide under oxygen-limited conditions when fumarate is less preferred as an alternative electron acceptor. In the inactive state, RacRS allows utilization of fumarate for respiration. The unique C. jejuni RacRS regulatory system illustrates the disparate evolution of Campylobacter and aids the survival of this pathogen.


Assuntos
Proteínas de Bactérias/metabolismo , Campylobacter jejuni/metabolismo , Metabolismo Energético/fisiologia , Fumaratos/metabolismo , Trato Gastrointestinal/microbiologia , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Animais , Proteínas de Bactérias/genética , Sequência de Bases , Transporte Biológico/genética , Ciclo do Ácido Cítrico/genética , Transporte de Elétrons/fisiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Dados de Sequência Molecular , Nitratos/metabolismo , Oxigênio/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Ácido Succínico/metabolismo , Transativadores/genética
10.
PLoS Pathog ; 9(5): e1003393, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23737749

RESUMO

The pathogen Campylobacter jejuni is the principal cause of bacterial food-borne infections. The mechanism(s) that contribute to bacterial survival and disease are still poorly understood. In other bacterial species, type VI secretion systems (T6SS) are increasingly recognized to contribute to bacterial pathogenesis by toxic effects on host cells or competing bacterial species. Here we report the presence of a functional Type VI secretion system in C. jejuni. Proteome and genetic analyses revealed that C. jejuni strain 108 contains a 17-kb T6SS gene cluster consisting of 13 T6SS-conserved genes, including the T6SS hallmark genes hcp and vgrG. The cluster lacks an ortholog of the ClpV ATPase considered important for T6SS function. The sequence and organization of the C. jejuni T6SS genes resemble those of the T6SS located on the HHGI1 pathogenicity island of Helicobacter hepaticus. The C. jejuni T6SS is integrated into the earlier acquired Campylobacter integrated element CJIE3 and is present in about 10% of C. jejuni isolates including several isolates derived from patients with the rare clinical feature of C. jejuni bacteremia. Targeted mutagenesis of C. jejuni T6SS genes revealed T6SS-dependent secretion of the Hcp needle protein into the culture supernatant. Infection assays provided evidence that the C. jejuni T6SS confers contact-dependent cytotoxicity towards red blood cells but not macrophages. This trait was observed only in a capsule-deficient bacterial phenotype. The unique C. jejuni T6SS phenotype of capsule-sensitive contact-mediated hemolysis represents a novel evolutionary pathway of T6SS in bacteria and expands the repertoire of virulence properties associated with T6SS.


Assuntos
Cápsulas Bacterianas , Proteínas de Bactérias , Sistemas de Secreção Bacterianos/genética , Campylobacter jejuni , Citotoxinas , Polissacarídeos Bacterianos , Animais , Cápsulas Bacterianas/genética , Cápsulas Bacterianas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Linhagem Celular , Citotoxinas/genética , Citotoxinas/metabolismo , Eritrócitos/metabolismo , Eritrócitos/microbiologia , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Família Multigênica , Polissacarídeos Bacterianos/genética , Polissacarídeos Bacterianos/metabolismo
11.
Antibiotics (Basel) ; 12(3)2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36978293

RESUMO

Campylobacter (C.) spp. are the most important foodborne, bacterial, and zoonotic pathogens worldwide. Resistance monitoring of foodborne bacterial pathogens is an important tool to control antimicrobial resistance as a part of the "One Health" approach. The detection and functionality of new resistance genes are of paramount importance in applying more effective screening methods based on whole genome sequencing (WGS). Most tetracycline-resistant C. spp. isolates harbor tet(O), a gene that encodes a ribosomal protection protein. Here we describe tet(O)_3, which has been identified in two food isolates of C. jejuni and is very similar to the tet(O) gene in Streptococcus pneumoniae, having a truncated promoter sequence. This gene confers resistance to tetracycline below 1 mg/L, which is the epidemiological cut-off value. We have analyzed the entire genome of these two isolates, together with a C. jejuni isolate found to have high-level resistance to tetracycline. In contrast to the highly resistant isolate, the promoter of tet(O)_3 is highly responsive to tetracycline, as observed by reverse transcription polymerase chain reaction (RT-PCR). In addition, the two isolates possess a CRISPR repeat, fluoroquinolone resistance due to the gyrA point mutation C257T, a ß-lactamase resistance gene blaOXA-184, a multidrug efflux pump CmeABC and its repressor CmeR, but no plasmid. Low-level antibiotic resistant C. jejuni might therefore have an advantage for surviving in non-host environments.

12.
Adv Microb Physiol ; 82: 129-154, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36948653

RESUMO

Lysophospholipids (LPLs) are lipid-derived metabolic intermediates in the cell membrane. The biological functions of LPLs are distinct from their corresponding phospholipids. In eukaryotic cells LPLs are important bioactive signaling molecules that regulate many important biological processes, but in bacteria the function of LPLs is still not fully defined. Bacterial LPLs are usually present in cells in very small amounts, but can strongly increase under certain environmental conditions. In addition to their basic function as precursors in membrane lipid metabolism, the formation of distinct LPLs contributes to the proliferation of bacteria under harsh circumstances or may act as signaling molecules in bacterial pathogenesis. This review provides an overview of the current knowledge of the biological functions of bacterial LPLs including lysoPE, lysoPA, lysoPC, lysoPG, lysoPS and lysoPI in bacterial adaptation, survival, and host-microbe interactions.


Assuntos
Fenômenos Biológicos , Lisofosfolipídeos , Lisofosfolipídeos/metabolismo , Transdução de Sinais , Metabolismo dos Lipídeos , Bactérias/metabolismo
13.
Gut Microbes ; 15(2): 2262592, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37768138

RESUMO

Enteric bacteria need to adapt to endure the antibacterial activities of bile salts in the gut. Phospholipase A (PldA) is a key enzyme in the maintenance of bacterial membrane homeostasis. Bacteria respond to stress by modulating their membrane composition. Campylobacter jejuni is the most common cause of human worldwide. However, the mechanism by which C. jejuni adapts and survives in the gut environment is not fully understood. In this study, we investigated the roles of PldA, bile salt sodium deoxycholate (DOC), and oxygen availability in C. jejuni biology, mimicking an in vivo situation. Growth curves were used to determine the adaptation of C. jejuni to bile salts. RNA-seq and functional assays were employed to investigate the PldA-dependent and DOC-induced changes in gene expression that influence bacterial physiology. Survival studies were performed to address oxidative stress defense in C. jejuni. Here, we discovered that PldA of C. jejuni is required for optimal growth in the presence of bile salt DOC. Under high oxygen conditions, DOC is toxic to C. jejuni, but under low oxygen conditions, as is present in the lumen of the gut, C. jejuni benefits from DOC. C. jejuni PldA seems to enable the use of iron needed for optimal growth in the presence of DOC but makes the bacterium more vulnerable to oxidative stress. In conclusion, DOC stimulates C. jejuni growth under low oxygen conditions and alters colony morphology in a PldA-dependent manner. C. jejuni benefits from DOC by upregulating iron metabolism in a PldA-dependent manner.


Assuntos
Campylobacter jejuni , Microbioma Gastrointestinal , Humanos , Ácidos e Sais Biliares/farmacologia , Ácido Desoxicólico/farmacologia , Ferro , Oxigênio
14.
Toxins (Basel) ; 15(10)2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37888617

RESUMO

Clostridioides difficile infection is expected to become the most common healthcare-associated infection worldwide. C. difficile-induced pathogenicity is significantly attributed to its enterotoxin, TcdA, which primarily targets Rho-GTPases involved in regulating cytoskeletal and tight junction (TJ) dynamics, thus leading to cytoskeleton breakdown and ultimately increased intestinal permeability. This study investigated whether two non-digestible oligosaccharides (NDOs), alginate (AOS) and chitosan (COS) oligosaccharides, possess antipathogenic and barrier-protective properties against C. difficile bacteria and TcdA toxin, respectively. Both NDOs significantly reduced C. difficile growth, while cell cytotoxicity assays demonstrated that neither COS nor AOS significantly attenuated the TcdA-induced cell death 24 h post-exposure. The challenge of Caco-2 monolayers with increasing TcdA concentrations increased paracellular permeability, as measured by TEER and LY flux assays. In this experimental setup, COS completely abolished, and AOS mitigated, the deleterious effects of TcdA on the monolayer's integrity. These events were not accompanied by alterations in ZO-1 and occludin protein levels; however, immunofluorescence microscopy revealed that both AOS and COS prevented the TcdA-induced occludin mislocalization. Finally, both NDOs accelerated TJ reassembly upon a calcium-switch assay. Overall, this study established the antipathogenic and barrier-protective capacity of AOS and COS against C. difficile and its toxin, TcdA, while revealing their ability to promote TJ reassembly in Caco-2 cells.


Assuntos
Toxinas Bacterianas , Quitosana , Clostridioides difficile , Humanos , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Células CACO-2 , Quitosana/farmacologia , Clostridioides/metabolismo , Alginatos/farmacologia , Ocludina , Enterotoxinas/toxicidade , Enterotoxinas/metabolismo , Oligossacarídeos/farmacologia , Oligossacarídeos/metabolismo
15.
Gut Microbes ; 14(1): 2091371, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35797141

RESUMO

Lysophospholipids (LPLs) are crucial for regulating epithelial integrity and homeostasis in eukaryotes, however the effects of LPLs produced by bacteria on host cells is largely unknown. The membrane of the human bacterial pathogen Campylobacter jejuni is rich in LPLs. Although C. jejuni possesses several virulence factors, it lacks traditional virulence factors like type III secretion systems, present in most enteropathogens. Here, we provide evidence that membrane lipids lysophosphatidylethanolamines (lysoPEs) of C. jejuni are able to lyse erythrocytes and are toxic for HeLa and Caco-2 cells. Lactate dehydrogenase (LDH) release assays and confocal microscopy revealed that lysoPE permeabilizes the cells. LysoPE toxicity was partially rescued by oxidative stress inhibitors, indicating that intracellular reactive oxygen species may contribute to the cell damage. Our results show that especially the short-chain lysoPEs (C:14) which is abundantly present in the C. jejuni membrane may be considered as a novel virulence factor.


Assuntos
Infecções por Campylobacter , Campylobacter jejuni , Microbioma Gastrointestinal , Células CACO-2 , Infecções por Campylobacter/microbiologia , Membrana Celular/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Fatores de Virulência/metabolismo
16.
J Biol Chem ; 285(21): 15828-36, 2010 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-20351099

RESUMO

Modification of the lipid A moiety of bacterial lipopolysaccharide influences cell wall properties, endotoxic activity, and bacterial resistance to antimicrobial peptides. Known modifications are variation in the number or length of acyl chains and/or attached phosphoryl groups. Here we identified two genes (gnnA and gnnB) in the major foodborne pathogen Campylobacter jejuni that enable the synthesis of a GlcN3N precursor UDP 2-acetamido-3-amino-2,3-dideoxy-alpha-D-glucopyranose (UDP-GlcNAc3N) in the lipid A backbone. Mass spectrometry of purified lipooligosaccharide verified that the gene products facilitate the formation of a 2,3-diamino-2,3-dideoxy-D-glucose (GlcN3N) disaccharide lipid A backbone when compared with the beta-1'-6-linked D-glucosamine (GlcN) disaccharide observed in Escherichia coli lipid A. Functional assays showed that inactivation of the gnnA or gnnB gene enhanced the TLR4-MD2-mediated NF-kappaB activation. The mutants also displayed increased susceptibility to killing by the antimicrobial peptides polymyxin B, colistin and the chicken cathelicidin-1. The gnnA and gnnB genes are organized in one operon with hemH, encoding a ferrochelatase catalyzing the last step in heme biosynthesis. These results indicate that lipid A modification resulting in amide-linked acyl chains in the lipid A is an effective mechanism to evade activation of the innate host defense and killing by antimicrobial peptides.


Assuntos
Anti-Infecciosos/farmacologia , Campylobacter jejuni/metabolismo , Farmacorresistência Bacteriana/efeitos dos fármacos , Lipídeo A/metabolismo , Receptor 4 Toll-Like/metabolismo , Açúcares de Uridina Difosfato/metabolismo , Animais , Campylobacter jejuni/genética , Campylobacter jejuni/imunologia , Campylobacter jejuni/patogenicidade , Configuração de Carboidratos , Galinhas , Farmacorresistência Bacteriana/genética , Células HeLa , Humanos , Lipídeo A/genética , Lipídeo A/imunologia , Antígeno 96 de Linfócito/genética , Antígeno 96 de Linfócito/imunologia , Antígeno 96 de Linfócito/metabolismo , NF-kappa B/genética , NF-kappa B/imunologia , NF-kappa B/metabolismo , Óperon/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Açúcares de Uridina Difosfato/genética
17.
Mol Microbiol ; 75(6): 1577-91, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20199595

RESUMO

Regulation of the biosynthesis of the flagellar filament in bacteria containing multiple flagellin genes is not well understood. The major food-borne pathogen Campylobacter jejuni possesses on both poles a flagellum that consists of two different flagellin subunits, FlaA and FlaB. Here we identify the protein Cj1464 as a regulator of C. jejuni flagellin biosynthesis. The protein shares characteristics of the FlgM family of anti-sigma factor proteins: it represses transcription of sigma(28)-dependent genes, forms a complex with sigma factor FliA, and is secreted through the flagellar filament. However, unlike other FlgM proteins, the interaction of C. jejuni FlgM with FliA is regulated by temperature and the protein does not inhibit FliA activity during the formation of the hook-basal body complex (HBB). Instead, C. jejuni FlgM limits the length of the flagellar filament by suppressing the synthesis of both the sigma(28)- and the sigma(54)-dependent flagellins. The main function of the C. jejuni FlgM therefore is not to silence sigma(28)-dependent genes until the HBB is completed, but to prevent unlimited elongation of the flagellum, which otherwise leads to reduced bacterial motility.


Assuntos
Proteínas de Bactérias/metabolismo , Campylobacter jejuni/fisiologia , Flagelos/fisiologia , Regulação Bacteriana da Expressão Gênica , Fator sigma/metabolismo , Temperatura , Proteínas de Bactérias/genética , Campylobacter jejuni/metabolismo , Campylobacter jejuni/efeitos da radiação , Campylobacter jejuni/ultraestrutura , Flagelos/metabolismo , Flagelos/efeitos da radiação , Flagelos/ultraestrutura , Flagelina/metabolismo , Deleção de Genes , Locomoção , Substâncias Macromoleculares/metabolismo , Microscopia Eletrônica de Transmissão
18.
Metabolites ; 11(5)2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33947169

RESUMO

The versatile compound n-butanol is one of the most promising biofuels for use in existing internal combustion engines, contributing to a smooth transition towards a clean energy society. Furthermore, n-butanol is a valuable resource to produce more complex molecules such as bioplastics. Microbial production of n-butanol from waste materials is hampered by the biotoxicity of n-butanol as it interferes with the proper functioning of lipid membranes. In this study we perform a large-scale investigation of the complete lipid-related enzyme machinery and its response to exposure to a sublethal concentration of n-butanol. We profiled, in triplicate, the growth characteristics and phospholipidomes of 116 different genetic constructs of E. coli, both in the presence and absence of 0.5% n-butanol (v/v). This led to the identification of 230 lipid species and subsequently to the reconstruction of the network of metabolites, enzymes and lipid properties driving the homeostasis of the E. coli lipidome. We were able to identify key lipids and biochemical pathways leading to altered n-butanol tolerance. The data led to new conceptual insights into the bacterial lipid metabolism which are discussed.

19.
J Bacteriol ; 192(11): 2729-36, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20348251

RESUMO

Two-component systems are widespread prokaryotic signal transduction devices which allow the regulation of cellular functions in response to changing environmental conditions. The two-component system DccRS (Cj1223c-Cj1222c) of Campylobacter jejuni is important for the colonization of chickens. Here, we dissect the DccRS system in more detail and provide evidence that the sensor DccS selectively phosphorylates the cognate effector, DccR. Microarray expression profiling, real-time reverse transcription-PCR (RT-PCR), electrophoretic mobility shift assay, and primer extension analyses revealed that the DccRS regulon of strain 81116 consists of five promoter elements, all containing the consensus direct repeat sequence WTTCAC-N6-TTCACW covering the putative -35 promoter regions. One of these promoters is located in front of an operon encoding a putative macrolide efflux pump while the others are in front of genes coding for putative periplasmic or membrane proteins. The DccRS-regulated genes in C. jejuni strain 81116 are needed to enhance early in vivo growth of C. jejuni in 7-day-old chickens. The DccRS system is activated in the late stationary bacterial growth phase, probably by released metabolic products. Whole-genome mRNA profiling and real-time RT-PCR analysis under these conditions demonstrated that the system has no influence on the transcription of genes outside the DccRS regulon.


Assuntos
Campylobacter jejuni/crescimento & desenvolvimento , Campylobacter jejuni/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulon/fisiologia , Animais , Proteínas de Bactérias/genética , Campylobacter jejuni/genética , Galinhas , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Bacteriana da Expressão Gênica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Regulon/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Curr Top Microbiol Immunol ; 337: 197-229, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19812984

RESUMO

Campylobacter jejuni is the principal bacterial foodborne pathogen. A major challenge still is to identify the virulence strategies exploited by C. jejuni. Recent genomics, proteomics, and metabolomics approaches indicate that C. jejuni displays extensive inter- and intrastrain variation. The diverse behavior enables bacterial adaptation to different environmental conditions and directs interactions with the gut mucosa. Here, we report recent progress in understanding the molecular mechanisms and functional consequences of the phenotype diversity. The results suggest that C. jejuni actively penetrates the intestinal mucus layer, secretes proteins mainly via its flagellar apparatus, is engulfed by intestinal cells, and can disrupt the integrity of the epithelial lining. C. jejuni stimulates the proinflammatory pathway and the production of a large repertoire of cytokines, chemokines, and innate effector molecules. Novel experimental infection models suggest that the activation of the innate immune response is important for the development of intestinal pathology.


Assuntos
Infecções por Campylobacter/microbiologia , Campylobacter jejuni/patogenicidade , Infecções por Campylobacter/imunologia , Campylobacter jejuni/fisiologia , Humanos , Imunidade Inata , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA