Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 644
Filter
1.
BMC Microbiol ; 24(1): 156, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724913

ABSTRACT

BACKGROUND: To establish a method to induce Campylobacter jejuni colonization in the intestines of C57BL/6 mice through antibiotic-induced microbiome depletion. RESULTS: Fifty-four female C57BL/6 mice were divided into the normal, control, and experimental groups. The experimental group was administered intragastric cefoperazone sodium and sulbactam sodium (50 mg/mL) for 2 days; then, the experimental and control mice were intragastrically administered 200 µL C. jejuni, which was repeated once more after 2 days. Animal feces were collected, and the HipO gene of C. jejuni was detected using TaqMan qPCR from day 1 to day 14 after modeling completion. Immunofluorescence was used to detect intestinal C. jejuni colonization on day 14, and pathological changes were observed using hematoxylin and eosin staining. Additionally, 16S rDNA analyses of the intestinal contents were conducted on day 14. In the experimental group, C. jejuni was detected in the feces from days 1 to 14 on TaqMan qPCR, and immunofluorescence-labeled C. jejuni were visibly discernable in the intestinal lumen. The intestinal mucosa was generally intact and showed no significant inflammatory-cell infiltration. Diversity analysis of the colonic microbiota showed significant inter-group differences. In the experimental group, the composition of the colonic microbiota differed from that in the other 2 groups at the phylum level, and was characterized by a higher proportion of Bacteroidetes and a lower proportion of Firmicutes. CONCLUSIONS: Microbiome depletion induced by cefoperazone sodium and sulbactam sodium could promote long-term colonization of C. jejuni in the intestines of mice.


Subject(s)
Anti-Bacterial Agents , Campylobacter Infections , Campylobacter jejuni , Cefoperazone , Feces , Gastrointestinal Microbiome , Mice, Inbred C57BL , RNA, Ribosomal, 16S , Sulbactam , Animals , Campylobacter jejuni/drug effects , Campylobacter jejuni/growth & development , Female , Anti-Bacterial Agents/pharmacology , Cefoperazone/pharmacology , Feces/microbiology , Campylobacter Infections/microbiology , Mice , Gastrointestinal Microbiome/drug effects , Sulbactam/pharmacology , RNA, Ribosomal, 16S/genetics , Intestines/microbiology , Colon/microbiology , Colon/pathology , Disease Models, Animal , Intestinal Mucosa/microbiology , Intestinal Mucosa/drug effects , DNA, Bacterial/genetics , DNA, Ribosomal/genetics
2.
Arch Microbiol ; 206(6): 260, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38744718

ABSTRACT

Campylobacter jejuni is known to enter a viable but non-culturable (VBNC) state when exposed to environmental stresses. Microarray and quantitative real-time polymerase chain reaction (qPCR) analyses were performed to elucidate the genes related to the induction of the VBNC state. The C. jejuni NCTC11168 strain was cultured under low-temperature or high-osmotic stress conditions to induce the VBNC state. mRNA expression in the VBNC state was investigated using microarray analysis, and the gene encoding peptidoglycan-associated lipoprotein, Pal, was selected as the internal control gene using qPCR analysis and software. The three genes showing particularly large increases in mRNA expression, cj1500, cj1254, and cj1040, were involved in respiration, DNA repair, and transporters, respectively. However, formate dehydrogenase encoded by cj1500 showed decreased activity in the VBNC state. Taken together, C. jejuni actively changed its mRNA expression during induction of the VBNC state, and protein activities did not always match the mRNA expression levels.


Subject(s)
Bacterial Proteins , Campylobacter jejuni , Gene Expression Regulation, Bacterial , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microbial Viability , Osmotic Pressure , Stress, Physiological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Gene Expression Profiling
3.
Am J Trop Med Hyg ; 106(2): 513-522, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34844207

ABSTRACT

Livestock can provide benefits to low-income households, yet may expose children to zoonotic enteropathogens that cause illness and negative long-term health outcomes. The aim of this cross-sectional study was to determine whether livestock-related risk factors, including animal ownership, exposure to animal feces, and consumption of animal-source foods, were associated with bacterial zoonotic enteropathogen infections in children 6-59 months old in Greater Accra, Ghana. Stool samples from 259 children and 156 household chickens were analyzed for atypical enteropathogenic Escherichia coli (aEPEC), Campylobacter jejuni/coli (C. jejuni/coli), Salmonella, and Shiga toxin-producing Escherichia coli (STEC) using quantitative polymerase chain reaction (qPCR). aEPEC, C. jejuni/coli, STEC, and Salmonella were detected in 45.6%, 11.6%, 4.3%, and 0.8% of children's stool samples, respectively. In adjusted logistic regression models, household ownership of goats or sheep was associated with STEC detection in children (odds ratio [95% confidence interval]: 4.30 [1.32, 14.08]), as were positive detection of STEC in chicken feces (7.85 [2.54, 24.30]) and frequent consumption of fresh cow's milk (3.03 [1.75, 5.24]). No livestock-related risk factors were associated with aEPEC or C. jejuni/coli infection in children. Our findings suggest that ruminant ownership in southern Ghana may expose children to STEC through household fecal contamination and foodborne routes. The lack of association between livestock risk factors and the more commonly detected pathogens, aEPEC and C. jejuni/coli, warrants further research, particularly to help explain how animal-keeping and sanitation practices affect transmission of fecal pathogens that were highly prevalent in chicken feces.


Subject(s)
Campylobacter Infections/epidemiology , Escherichia coli Infections/epidemiology , Livestock/microbiology , Ruminants/microbiology , Salmonella Infections/epidemiology , Animals , Campylobacter Infections/diagnosis , Campylobacter Infections/microbiology , Campylobacter jejuni/growth & development , Campylobacter jejuni/pathogenicity , Cattle , Chickens/microbiology , Child, Preschool , Cross-Sectional Studies , Enteropathogenic Escherichia coli/growth & development , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/diagnosis , Escherichia coli Infections/microbiology , Feces/microbiology , Ghana , Goats , Humans , Infant , Logistic Models , Milk/microbiology , Salmonella/growth & development , Salmonella/pathogenicity , Salmonella Infections/diagnosis , Salmonella Infections/microbiology , Sheep , Shiga-Toxigenic Escherichia coli/growth & development , Shiga-Toxigenic Escherichia coli/pathogenicity
4.
Viruses ; 13(8)2021 07 22.
Article in English | MEDLINE | ID: mdl-34452294

ABSTRACT

Campylobacteriosis is the most commonly reported gastrointestinal disease in humans. Campybacter jejuni is the main cause of the infection, and bacterial colonization in broiler chickens is widespread and difficult to prevent, leading to high risk of occurrence in broiler meat. Phage therapy represents an alternative strategy to control Campylobacter in poultry. The aim of this work was to assess the efficacy of two field-isolated bacteriophages against experimental infections with an anti-microbial resistant (AMR) Campylobacter jejuni strain. A two-step phage application was tested according to a specific combination between chickens' rearing time and specific multiplicities of infections (MOIs), in order to reduce the Campylobacter load in the animals at slaughtering and to limit the development of phage-resistant mutants. In particular, 75 broilers were divided into three groups (A, B and C), and phages were administered to animals of groups B and C at day 38 (Φ 16-izsam) and 39 (Φ 7-izsam) at MOI 0.1 (group B) and 1 (group C). All broilers were euthanized at day 40, and Campylobacter jejuni was enumerated in cecal contents. Reductions in Campylobacter counts were statistically significant in both group B (1 log10 colony forming units (cfu)/gram (gr)) and group C (2 log10 cfu/gr), compared to the control group. Our findings provide evidence about the ability of phage therapy to reduce the Campylobacter load in poultry before slaughtering, also associated with anti-microbial resistance pattern.


Subject(s)
Campylobacter Infections/veterinary , Campylobacter jejuni/growth & development , Chickens/microbiology , Phage Therapy , Poultry Diseases/therapy , Animals , Bacterial Load , Bacteriophages/physiology , Campylobacter Infections/microbiology , Campylobacter Infections/therapy , Cecum/microbiology , Poultry Diseases/microbiology
5.
Food Microbiol ; 100: 103853, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34416958

ABSTRACT

The combined effects of ethylenediaminetetraacetic acid (EDTA) and bacteriophage (phage) treatment of foodborne pathogens were investigated. Although viable counts for Campylobacter jejuni decreased by 1.5 log after incubation for 8 h in the presence of phage PC10, re-growth was observed thereafter. The combination of phage PC10 and 1 mM EDTA significantly inhibited the re-growth of C. jejuni. The viable counts for C. jejuni decreased by 2.6 log (P < 0.05) compared with that of the initial count after 24 h. Moreover, EDTA at 0.67 or 1.3 mM, combined with the specific lytic phages, also effectively inhibited the re-growth of phage-resistant cells of Campylobacter coli, Salmonella enterica serovar Enteritidis, and Salmonella enterica serovar Typhimurium. In addition, the combined effects of lytic phages and EDTA were investigated on the viability of Campylobacter in BHI broth at low temperatures followed by the optimum growth temperature. The re-growth of C. coli was significantly inhibited by the coexistence of 1.3 mM EDTA, and the viable counts of surviving bacteria was about the same as the initial viable count after the incubation. This is the first study demonstrating the combined use of lytic phages and EDTA is effective in inhibiting the re-growth of phage-resistant bacteria in Gram-negative bacteria.


Subject(s)
Bacteriophages/physiology , Campylobacter coli/growth & development , Campylobacter jejuni/growth & development , Edetic Acid/pharmacology , Salmonella enteritidis/growth & development , Salmonella typhimurium/growth & development , Campylobacter coli/drug effects , Campylobacter coli/virology , Campylobacter jejuni/drug effects , Campylobacter jejuni/virology , Microbial Viability , Salmonella enteritidis/drug effects , Salmonella enteritidis/virology , Salmonella typhimurium/drug effects , Salmonella typhimurium/virology
6.
Appl Environ Microbiol ; 87(19): e0109921, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34319799

ABSTRACT

New approaches for the control of Campylobacter jejuni biofilms in the food industry are being studied intensively. Natural products are promising alternative antimicrobial substances to control biofilm production, with particular emphasis on plant extracts. Dried flowers of Lavandula angustifolia were used to produce essential oil (LEO), an ethanol extract (LEF), and an ethanol extract of Lavandula postdistillation waste material (LEW). The chemical compositions determined for these Lavandula preparations included seven major compounds that were selected for further testing. These were tested against C. jejuni for biofilm degradation and removal. Next-generation sequencing was used to study the molecular mechanisms underlying LEO actions against C. jejuni adhesion and motility. Analysis of LEO revealed 1,8-cineol, linalool, and linalyl acetate as the main components. For LEF and LEW, the main components were phenolic acid glycosides, with flavonoids rarely present. The MICs of the Lavandula preparations and pure compounds against C. jejuni ranged from 0.2 mg/ml to 1 mg/ml. LEO showed the strongest biofilm degradation. The reduction of C. jejuni adhesion was ≥1 log10 CFU/ml, which satisfies European Food Safety Authority recommendations. Lavandula preparations reduced C. jejuni motility by almost 50%, which consequently can impact biofilm formation. These data are in line with the transcriptome analysis of C. jejuni, which indicated that LEO downregulated genes important for biofilm formation. LEW also showed good antibacterial and antibiofilm effects, particularly against adhesion and motility mechanisms. This defines an innovative approach using alternative strategies and novel targets to combat bacterial biofilm formation and, hence, the potential to develop new effective agents with biofilm-degrading activities. IMPORTANCE The Lavandula preparations used in this study are found to be effective against C. jejuni, a common foodborne pathogen. They show antibiofilm properties at subinhibitory concentrations in terms of promoting biofilm degradation and inhibiting cell adhesion and motility, which are involved in the initial steps of biofilm formation. These results are confirmed by transcriptome analysis, which highlights the effect of Lavandula essential oil on C. jejuni biofilm properties. We show that the waste material from the hydrodistillation of Lavandula has particular antibiofilm effects, suggesting that it has potential for reuse for industrial purposes. This study highlights the need for efforts directed toward such innovative approaches and alternative strategies against biofilm formation and maintenance by developing new naturally derived agents with antibiofilm activities.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Campylobacter jejuni/drug effects , Lavandula , Oils, Volatile/pharmacology , Plant Extracts/pharmacology , Plant Oils/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Adhesion/drug effects , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Campylobacter jejuni/physiology , Flavonoids/analysis , Flavonoids/pharmacology , Flowers , Gene Expression Regulation, Bacterial/drug effects , Oils, Volatile/chemistry , Phytochemicals/analysis , Phytochemicals/pharmacology , Plant Extracts/chemistry , Plant Oils/chemistry , Waste Products
7.
Sci Rep ; 11(1): 13837, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34226590

ABSTRACT

In retail meat products, Campylobacter jejuni, C. coli, and Staphylococcus aureus have been reported in high prevalence. The polymicrobial interaction between Campylobacter and other bacteria could enhance Campylobacter survival during the adverse conditions encountered during retail meat processing and storage. This study was designed to investigate the potential role of S. aureus from retail meats in enhancing the survival of Campylobacter exposed to low temperature, aerobic conditions, and biofilm formation. Results indicated that viable S. aureus cells and filter-sterilized cell-free media obtained from S. aureus prolonged the survival of Campylobacter at low temperature and during aerobic conditions. Biofilm formation of Campylobacter strains was significantly enhanced in the presence of viable S. aureus cells, but the results were inconclusive when extracts from cell-free media were used. In conclusion, the presence of S. aureus cells enhances survivability of Campylobacter strains in adverse conditions such as low temperature and aerobic conditions. Further investigations are warranted to understand the interaction between Campylobacter and S. aureus, and effective intervention strategies are needed to reduce the incidence of both foodborne pathogens in retail meat products.


Subject(s)
Biofilms/growth & development , Campylobacter jejuni/genetics , Meat/microbiology , Staphylococcus aureus/genetics , Campylobacter coli/genetics , Campylobacter coli/growth & development , Campylobacter coli/pathogenicity , Campylobacter jejuni/growth & development , Campylobacter jejuni/pathogenicity , Coinfection/genetics , Coinfection/microbiology , Food Microbiology , Humans , Meat Products/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity
8.
BMC Microbiol ; 21(1): 169, 2021 06 05.
Article in English | MEDLINE | ID: mdl-34090326

ABSTRACT

BACKGROUND: Campylobacter jejuni is the major micro-bacillary pathogen responsible for human coloenteritis. Lactic acid bacteria (LAB) have been shown to protect against Campylobacter infection. However, LAB with a good ability to inhibit the growth of C. jejuni in vitro are less effective in animals and animal models, and have the disadvantages of high cost, a long cycle, cumbersome operation and insignificant immune response indicators. Caenorhabditis elegans is increasingly used to screen probiotics for their anti-pathogenic properties. However, no research on the use of C. elegans to screen for probiotic candidates antagonistic to C. jejuni has been conducted to date. RESULTS: This study established a lifespan model of C. elegans, enabling the preselection of LAB to counter C. jejuni infection. A potential protective mechanism of LAB was identified. Some distinct LAB species offered a high level of protection to C. elegans against C. jejuni. The LAB strains with a high protection rate reduced the load of C. jejuni in C. elegans. The transcription of antibacterial peptide genes, MAPK and Daf-16 signalling pathway-related genes was elevated using the LAB isolates with a high protection rate. The reliability of the lifespan model of C. elegans was verified using mice and chickens infected with C. jejuni. CONCLUSIONS: The results showed that different LAB had different abilities to protect C. elegans against C. jejuni. C. elegans provides a reliable model for researchers to screen for LAB that are antagonistic to C. jejuni on a large scale.


Subject(s)
Caenorhabditis elegans/drug effects , Caenorhabditis elegans/immunology , Campylobacter Infections/drug therapy , Campylobacter jejuni/drug effects , Disease Models, Animal , Lactobacillales/physiology , Probiotics/administration & dosage , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Campylobacter Infections/genetics , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Campylobacter jejuni/growth & development , Chickens/genetics , Chickens/immunology , Chickens/microbiology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Mice/genetics , Mice/immunology , Mice/microbiology , Mice, Inbred C57BL , Nematoda/genetics , Nematoda/immunology , Nematoda/microbiology
9.
PLoS One ; 16(2): e0247305, 2021.
Article in English | MEDLINE | ID: mdl-33621246

ABSTRACT

The Campylobacter jejuni capsule type HS1 complex is one of the most common serotypes identified worldwide, and consists of strains typing as HS1, HS1/44, HS44 and HS1/8. The capsule structure of the HS1 type strain was shown previously to be composed of teichoic-acid like glycerol-galactosyl phosphate repeats [4-)-α-D-Galp-(1-2)-Gro-(1-P-] with non-stoichiometric fructose branches at the C2 and C3 of Gal and non-stoichiometric methyl phosphoramidate (MeOPN) modifications on the C3 of the fructose. Here, we demonstrate that the capsule of an HS1/44 strain is identical to that of the type strain of HS1, and the capsule of HS1/8 is also identical to HS1, except for an additional site of MeOPN modification at C6 of Gal. The DNA sequence of the capsule locus of an HS44 strain included an insertion of 10 genes, and the strain expressed two capsules, one identical to the HS1 type strain, but with no fructose branches, and another composed of heptoses and MeOPN. We also characterize a HS1 capsule biosynthesis gene, HS1.08, as a fructose transferase responsible for the attachment of the ß-D-fructofuranoses residues at C2 and C3 of the Gal unit. In summary, the common component of all members of the HS1 complex is the teichoic-acid like backbone that is likely responsible for the observed sero-cross reactivity.


Subject(s)
Campylobacter jejuni/growth & development , Polysaccharides, Bacterial/genetics , Sequence Analysis, DNA/methods , Bacterial Capsules/genetics , Campylobacter jejuni/classification , Campylobacter jejuni/genetics , Carbohydrate Sequence , Multigene Family , Mutation , Serogroup
10.
Biomolecules ; 11(2)2021 02 05.
Article in English | MEDLINE | ID: mdl-33562547

ABSTRACT

Green synthesized metal oxide nanoparticles (NPs) have prominent applications in antimicrobial packaging systems. Here we have attempted for the fabrication of chitosan-based nanocomposite film containing Urtica dioica leaf extract derived copper oxide (CuO) and zinc oxide (ZnO) NPs for shelf-life extension of the packaged guava fruits. Electron microscopy and spectroscopy analysis of the CuO and ZnO NPs exhibited nano-scale size, spherical morphologies, and negative ζ-potential values. The NPs possessed appreciable antioxidant and antimicrobial activity (AMA) in order of CuO NPs > ZnO NPs >nettle extract. Therefore, this work establishes for the first time the successful synthesis of CuO NPs and compares its antimicrobial and antioxidant properties with ZnO NPs. On incorporation in chitosan, the polymer nanocomposite films were developed by solvent casting technique. The developed films were transparent, had low antioxidant but substantial AMA. The NP supplementation improved the film characteristics as evident from the decrease in moisture content, water holding capacity, and solubility of the films. The nanocomposite films improved the quality attributes and shelf life of guava fruits by one week on packaging and storage compared to unpackaged control fruits. Therefore, this study demonstrates the higher antimicrobial potential of the nettle leaf extract derived CuO/ZnO NPs for development of antimicrobial nanocomposite films as a promising packaging solution for enhancing the shelf life of various perishable fruits.


Subject(s)
Anti-Bacterial Agents/chemistry , Antioxidants/chemistry , Food Preservation/methods , Metal Nanoparticles/chemistry , Nanocomposites/chemistry , Urtica dioica/chemistry , Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Campylobacter jejuni/drug effects , Campylobacter jejuni/growth & development , Copper/chemistry , Enterobacter cloacae/drug effects , Enterobacter cloacae/growth & development , Food Packaging/methods , Food Storage , Fruit/chemistry , Fruit/microbiology , Humans , Membranes, Artificial , Microbial Sensitivity Tests , Plant Extracts , Plant Leaves/chemistry , Psidium , Salmonella typhi/drug effects , Salmonella typhi/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Zinc Oxide/chemistry
11.
Lett Appl Microbiol ; 72(1): 36-40, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32964486

ABSTRACT

Campylobacter spp. is the leading cause of bacterial gastroenteritis worldwide and poultry are the primary reservoir. The aim of this study was to investigate the survival and/or growth of Campylobacter jejuni NCTC 11168 in broiler digestate prepared from commercial starter, grower and finisher feed formulations. Bolton broth and digestates were prepared, inoculated with C. jejuni NCTC 11168 (approximately 3 log10 CFU per ml) and incubated under microaerobic conditions at 42°C for 24 h. Samples were taken at t = 0 (immediately after inoculation) and every 3 h thereafter, serially diluted and plated onto mCCDA. Campylobacter jejuni grew as expected in Bolton broth (control) reaching the early stationary phase after approximately 15 h. In contrast, although bacterial concentrations were maintained for at least 9 h, none of the feed digestates supported the growth of C. jejuni, which were not detected after 15 h. It is suggested that the nutrients available in the feed digestates are not enough to support C. jejuni growth and that additional factors may be at play in the avian gastrointestinal tract.


Subject(s)
Animal Feed/microbiology , Campylobacter Infections/microbiology , Campylobacter jejuni/growth & development , Chickens/microbiology , Disease Reservoirs/microbiology , Gastroenteritis/microbiology , Poultry Diseases/microbiology , Animals , Campylobacter Infections/epidemiology , Gastroenteritis/epidemiology , Humans
12.
FEMS Microbiol Lett ; 367(20)2020 11 05.
Article in English | MEDLINE | ID: mdl-33098301

ABSTRACT

Campylobacter jejuni is a major cause of food-borne human bacterial gastroenteritis but animal models for C. jejuni mediated disease remain limited because C. jejuni poorly colonizes immunocompetent, conventionally-reared (Conv-R) mice. Thus, a reliable rodent model (i.e. persistent colonization) is desirable in order to evaluate C. jejuni-mediated gastrointestinal disease and mechanisms of pathogenicity. As the nature and complexity of the microbiota likely impacts colonization resistance for C. jejuni, Conv-R and gnotobiotic C3H/HeN mice were used to evaluate the persistence of C. jejuni colonization and development of disease. A total of four C. jejuni isolates readily and persistently colonized ASF mice and induced mild mucosal inflammation in the proximal colon, but C. jejuni did not stably colonize nor induce lesions in Conv-R mice. This suggests that the pathogenesis of C. jejuni is influenced by the microbiota, and that ASF mice offer a reproducible model to study the influence of the microbiota on the ability of C. jejuni to colonize the gut and to mediate gastroenteritis.


Subject(s)
Campylobacter jejuni/growth & development , Colitis/microbiology , Microbial Interactions/physiology , Microbiota/physiology , Animals , Campylobacter Infections/microbiology , Germ-Free Life , Mice , Mice, Inbred C3H
13.
Cell Microbiol ; 22(12): e13252, 2020 12.
Article in English | MEDLINE | ID: mdl-32827216

ABSTRACT

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.


Subject(s)
Bacteroides fragilis/enzymology , Campylobacter jejuni/growth & development , Campylobacter jejuni/pathogenicity , Fucose/metabolism , Mucins/metabolism , alpha-L-Fucosidase/metabolism , Caco-2 Cells , Campylobacter jejuni/genetics , Humans , Microbial Interactions/physiology , Virulence , alpha-L-Fucosidase/biosynthesis
14.
Microbiology (Reading) ; 166(9): 849-853, 2020 09.
Article in English | MEDLINE | ID: mdl-32697188

ABSTRACT

Campylobacter jejuni is a major bacterial foodborne-pathogen. Ciprofloxacin is an important antibiotic for the treatment of C. jejuni, albeit high rates of fluoroquinolone resistance have limited its usefulness. Persister-cells are transiently antibiotic-tolerant fractions of bacterial populations and their occurrence has been associated with recalcitrant and persistent bacterial infections. Here, time-kill assays with ciprofloxacin (200×MIC, 25 µg ml-1) were performed in C. jejuni strains 81-176 and RM1221 and persister-cells were found. The frequency of survivors after 8 h of ciprofloxacin exposure was approx. 10-3 for both strains, while after 22 h the frequency was between 10-5-10-7, depending on the strain and growth-phase. Interestingly, the stationary-phase cultures did not display more persister-cells compared to exponential-phase cultures, in contrast to what has been observed in other bacterial species. Persister-cells after ampicillin exposure (100×MIC, 200 µg ml-1) were not detected, implying that persister-cell formation in C. jejuni is antibiotic-specific. In attempts to identify the mechanism of ciprofloxacin persister-cell formation, stringent or SOS responses were not found to play major roles. Overall, this study reports ciprofloxacin persister-cells in C. jejuni and challenges the notion of persister-cells as plainly dormant non-growing cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Campylobacter jejuni/drug effects , Campylobacter jejuni/physiology , Ciprofloxacin/pharmacology , Ampicillin/pharmacology , Bacterial Load/drug effects , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , DNA Damage , Drug Resistance, Bacterial , Drug Tolerance , Microbial Sensitivity Tests , SOS Response, Genetics
15.
J Mol Biol ; 432(19): 5244-5258, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32710984

ABSTRACT

In response to changes in their environment bacteria need to change both their protein and phospholipid repertoire to match environmental requirements, but the dynamics of bacterial phospholipid composition under different growth conditions is still largely unknown. In the present study, we investigated the phospholipidome of the bacterial pathogen Campylobacter jejuni. Transcription profiling on logarithmic and stationary phase grown cells of the microaerophilic human pathogen C. jejuni using RNA-seq revealed differential expression of putative phospholipid biosynthesis genes. By applying high-performance liquid chromatography tandem-mass spectrometry, we identified 203 phospholipid species representing the first determination of the phospholipidome of this pathogen. We identified nine different phospholipid classes carrying between one and three acyl chains. Phospholipidome analysis on bacteria of different ages (0-5 days) showed rapid changes in the ratio of phospholipids containing ethanolamine, or glycerol as phospholipid head group and in the number of cyclopropane bond containing fatty acids. Oxygen concentration influenced the percentage of lysophospholipids, and cyclo-propane bonds containing acyl chains. We show that large amounts of the phospholipids are lysophospholipids (30-45%), which mutant studies reveal are needed for normal C. jejuni motility at low oxygen conditions. C. jejuni possesses an unusual phospholipidome that is highly dynamic in response to environmental changes.


Subject(s)
Campylobacter Infections/microbiology , Campylobacter jejuni/metabolism , Oxygen/metabolism , Phospholipids/metabolism , Biosynthetic Pathways , Campylobacter jejuni/chemistry , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Gene Expression Regulation, Bacterial , Genes, Bacterial , Humans , Lipidomics , Lysophospholipids/analysis , Lysophospholipids/genetics , Lysophospholipids/metabolism , Metabolome , Phospholipids/analysis , Phospholipids/genetics , Transcriptome
16.
J Agric Food Chem ; 68(31): 8455-8461, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32663006

ABSTRACT

Herein, a high-affinity single-stranded DNA aptamer (59 nt) against Campylobacter jejuni, defined as CJA1, was obtained using the whole-bacterium-based systemic evolution of ligands by exponential enrichment procedure. CJA1 was analyzed with a stable secondary structure and low dissociation constant (Kd) value of 1.37 ± 0.28 nM. The potential use of CJA1 was exemplified by the construction of a hetero-sandwich platform, in which C. jejuni was bound with a biotin-tagged CJA1 to perform a colorimetric reaction that is associated with visible color changes and detectable optical responses. Dependent upon this sensing platform, C. jejuni can be detected from 1.7 × 101 to 1.7 × 106 colony-forming units (CFU)/mL. The limit of detection (LOD) is obtained as 10 CFU/mL in PBS. The specificity study showed that the sensing platform is easy to distinguish C. jejuni from other common pathogens. Moreover, the C. jejuni-contaminated milk samples can also be accurately probed (LOD = 13 CFU/mL) without sacrificing its assay abilities, indicating the promising prospect of CJA1 in the fields of biosensing and diagnostics.


Subject(s)
Campylobacter jejuni/isolation & purification , Colorimetry/methods , Food Microbiology/methods , SELEX Aptamer Technique/methods , Animals , Aptamers, Nucleotide/genetics , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Cattle , Food Contamination/analysis , Limit of Detection , Milk/microbiology
17.
Mol Cell Proteomics ; 19(8): 1263-1280, 2020 08.
Article in English | MEDLINE | ID: mdl-32376616

ABSTRACT

Campylobacter jejuni is a major cause of food-borne gastroenteritis. Proteomics by label-based two-dimensional liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) identified proteins associated with growth in 0.1% sodium deoxycholate (DOC, a component of gut bile salts), and system-wide validation was performed by data-independent acquisition (DIA-SWATH-MS). LC-MS/MS quantified 1326 proteins (∼82% of the predicted C. jejuni proteome), of which 1104 were validated in additional biological replicates by DIA-SWATH-MS. DOC resulted in a profound proteome shift with 512 proteins showing significantly altered abundance. Induced proteins were associated with flagellar motility and antibiotic resistance; and these correlated with increased DOC motility and resistance to polymyxin B and ciprofloxacin. DOC also increased human Caco-2 cell adherence and invasion. Abundances of proteins involved in nutrient transport were altered by DOC and aligned with intracellular changes to their respective carbon sources. DOC increased intracellular levels of sulfur-containing amino acids (cysteine and methionine) and the dipeptide cystine (Cys-Cys), which also correlated with reduced resistance to oxidative stress. A DOC induced transport protein was Cj0025c, which has sequence similarity to bacterial Cys-Cys transporters. Deletion of cj0025c (Δcj0025c) resulted in proteome changes consistent with sulfur starvation, as well as attenuated invasion, reduced motility, atypical morphology, increased antimicrobial susceptibility and poor biofilm formation. Targeted metabolomics showed Δcj0025c could use known C. jejuni amino and organic acid substrates commensurate with wild-type. Medium Cys-Cys levels however, were maintained in Δcj0025c relative to wild-type. A toxic Cys-Cys mimic (selenocystine) inhibited wild-type growth, but not Δcj0025c Provision of an alternate sulfur source (2 mm thiosulfate) restored Δcj0025c motility. Our data confirm that Cj0025c is a Cys-Cys transporter that we have named TcyP consistent with the nomenclature of homologous proteins in other species.


Subject(s)
Bacterial Proteins/metabolism , Campylobacter Infections/metabolism , Campylobacter Infections/microbiology , Campylobacter jejuni/growth & development , Carrier Proteins/metabolism , Cystine/metabolism , Deoxycholic Acid/pharmacology , Proteomics , Campylobacter jejuni/drug effects , Campylobacter jejuni/pathogenicity , Carbon/pharmacology , Humans , Oxidative Stress/drug effects , Phenotype , Proteome/metabolism , Sulfur/deficiency , Virulence/drug effects
18.
J Vis Exp ; (157)2020 03 10.
Article in English | MEDLINE | ID: mdl-32225160

ABSTRACT

A culture from human stool for diagnosis of Campylobacter-based intestinal illness takes several days, a wait that taxes the fortitude of the physician and the patient. A culture is also prone to false negative results from random loss of viability during specimen handling, overgrowth of other fecal flora, and poor growth of several pathogenic Campylobacter species on traditional media. These problems can confound clinical decisions on patient treatment and have limited the field from answering fundamental questions on Campylobacter growth and infections. We describe a procedure that estimates the lower limit of bacterial numbers that can be detected by a culture and a method for quantifying survival of C. jejuni in media used for transport of this fragile organism. Knowing this information, it becomes possible to set clinically relevant detection thresholds for diagnostic tests and address unstudied issues of whether non-symptomatic colonization is prevalent, if co-infection with other enteric pathogens is common, or if bacterial load correlates with symptoms or serious sequelae. The study also included testing of 1,552 prospectively collected patient diarrheal fecal specimens that were initially classified by conventional culture and further tested by a new enzyme immunoassay. Positive and discrepant specimens were then screened by four molecular methods to assign true-positive or true-negative status. The 5 non-culture methods showed complete agreement on all 48 positive and discrepant specimens, while the culture mis-identified 14 (28%). The specimens that were incorrectly identified by culture included 13 false negative and 1 false positive sample. This basic protocol can be used with multiple Campylobacter spp. and will allow the numbers of Campylobacter bacteria that produce symptoms of gastroenteritis in humans to be determined and for prevalence rates to be updated.


Subject(s)
Campylobacter jejuni/cytology , Culture Media , Culture Techniques/methods , Feces/microbiology , Limit of Detection , Microbial Viability , Transportation , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Colony Count, Microbial , Genes, Bacterial , Humans
19.
Sci Rep ; 10(1): 3686, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111893

ABSTRACT

Campylobacter is the major bacterial agent of human gastroenteritis worldwide and represents a crucial global public health burden. Species differentiation of C. jejuni and C. coli and phylogenetic analysis is challenged by inter-species horizontal gene transfer. Routine real-time PCR on more than 4000 C. jejuni and C. coli field strains identified isolates with ambiguous PCR results for species differentiation, in particular, from the isolation source eggs. K-mer analysis of whole genome sequencing data indicated the presence of C. coli hybrid strains with huge amounts of C. jejuni introgression. Recombination events were distributed over the whole chromosome. MLST typing was impaired, since C. jejuni sequences were also found in six of the seven housekeeping genes. cgMLST suggested that the strains were phylogenetically unrelated. Intriguingly, the strains shared a stress response set of C. jejuni variant genes, with proposed roles in oxidative, osmotic and general stress defence, chromosome maintenance and repair, membrane transport, cell wall and capsular biosynthesis and chemotaxis. The results have practical impact on routine typing and on the understanding of the functional adaption to harsh environments, enabling successful spreading and persistence of Campylobacter.


Subject(s)
Campylobacter Infections/genetics , Campylobacter coli/genetics , Campylobacter jejuni/growth & development , Gastroenteritis/genetics , Genetic Variation , Genome, Bacterial , Recombination, Genetic , Animals , Campylobacter Infections/diagnosis , Campylobacter Infections/microbiology , Campylobacter coli/pathogenicity , Campylobacter jejuni/pathogenicity , Gastroenteritis/diagnosis , Gastroenteritis/microbiology , Humans , Whole Genome Sequencing
20.
BMC Vet Res ; 16(1): 34, 2020 Feb 03.
Article in English | MEDLINE | ID: mdl-32013961

ABSTRACT

BACKGROUND: Campylobacter spp. are a major cause of bacterial food-borne diarrhoeal disease. This mainly arises through contamination of meat products during processing. For infection, Campylobacter spp. must adhere to epithelial cells of the mucus layer, survive conditions of the gastrointestinal tract, and colonise the intestine of the host. Addition of probiotic bacteria might promote competitive adhesion to epithelial cells, consequently reducing Campylobacter jejuni colonisation. Effect of Lactobacillus spp. (PCS20, PCS22, PCS25, LGG, PCK9) on C. jejuni adhesion, invasion and translocation in pig (PSI cl.1) and chicken (B1OXI) small-intestine cell lines, as well as pig enterocytes (CLAB) was investigated. RESULTS: Overall, in competitive adhesion assays with PSI cl.1 and CLAB cell monolayers, the addition of Lactobacillus spp. reduced C. jejuni adherence to the cell surface, and negatively affected the C. jejuni invasion. Interestingly, Lactobacillus spp. significantly impaired C. jejuni adhesion in three-dimensional functional PSI cl.1 and B1OXI cell models. Also, C. jejuni did not translocate across PSI cl.1 and B1OXI cell monolayers when co-incubated with probiotics. Among selected probiotics, Lactobacillus rhamnosus LGG was the strain that reduced adhesion efficacy of C. jejuni most significantly under co-culture conditions. CONCLUSION: The addition of Lactobacillus spp. to feed additives in livestock nutrition might be an effective novel strategy that targets Campylobacter adhesion to epithelial cells, and thus prevents colonisation, reduces the transmission, and finally lowers the incidence of human campylobacteriosis.


Subject(s)
Bacterial Adhesion , Bacterial Translocation , Campylobacter Infections/microbiology , Campylobacter jejuni/physiology , Lactobacillus/physiology , Animals , Campylobacter Infections/physiopathology , Campylobacter jejuni/growth & development , Cell Line , Chickens , Epithelial Cells/microbiology , Probiotics , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...