Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 630
Filter
1.
Nat Commun ; 13(1): 976, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35190534

ABSTRACT

The MORDOR trial in Niger, Malawi, and Tanzania found that biannual mass distribution of azithromycin to children younger than 5 years led to a 13.5% reduction in all-cause mortality (NCT02048007). To help elucidate the mechanism for mortality reduction, we report IgG responses to 11 malaria, bacterial, and protozoan pathogens using a multiplex bead assay in pre-specified substudy of 30 communities in the rural Niger placebo-controlled trial over a three-year period (n = 5642 blood specimens, n = 3814 children ages 1-59 months). Mass azithromycin reduces Campylobacter spp. force of infection by 29% (hazard ratio = 0.71, 95% CI: 0.56, 0.89; P = 0.004) but serological measures show no significant differences between groups for other pathogens against a backdrop of high transmission. Results align with a recent microbiome study in the communities. Given significant sequelae of Campylobacter infection among preschool aged children, our results support an important mechanism through which biannual mass distribution of azithromycin likely reduces mortality in Niger.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Azithromycin/administration & dosage , Child Mortality , Immunoglobulin G/blood , Mass Drug Administration , Campylobacter Infections/blood , Campylobacter Infections/immunology , Campylobacter Infections/mortality , Campylobacter Infections/prevention & control , Child , Child, Preschool , Cryptosporidiosis/blood , Cryptosporidiosis/immunology , Cryptosporidiosis/mortality , Cryptosporidiosis/parasitology , Drug Resistance, Bacterial , Escherichia coli Infections/blood , Escherichia coli Infections/immunology , Escherichia coli Infections/mortality , Escherichia coli Infections/prevention & control , Follow-Up Studies , Giardiasis/blood , Giardiasis/immunology , Giardiasis/mortality , Giardiasis/parasitology , Humans , Immunoglobulin G/immunology , Infant , Malaria/blood , Malaria/immunology , Malaria/mortality , Malaria/parasitology , Niger/epidemiology , Rural Population/statistics & numerical data , Salmonella Infections/blood , Salmonella Infections/immunology , Salmonella Infections/mortality , Salmonella Infections/prevention & control
2.
PLoS Pathog ; 17(8): e1009787, 2021 08.
Article in English | MEDLINE | ID: mdl-34339468

ABSTRACT

The Gram-negative bacterium Campylobacter jejuni is a major cause of foodborne disease in humans. After infection, C. jejuni rapidly colonizes the mucus layer of the small and large intestine and induces a potent pro-inflammatory response characterized by the production of a large repertoire of cytokines, chemokines, and innate effector molecules, resulting in (bloody) diarrhea. The virulence mechanisms by which C. jejuni causes this intestinal response are still largely unknown. Here we show that C. jejuni releases a potent pro-inflammatory compound into its environment, which activates an NF-κB-mediated pro-inflammatory response including the induction of CXCL8, CXCL2, TNFAIP2 and PTGS2. This response was dependent on a functional ALPK1 receptor and independent of Toll-like Receptor and Nod-like Receptor signaling. Chemical characterization, inactivation of the heptose-biosynthesis pathway by the deletion of the hldE gene and in vitro engineering identified the released factor as the LOS-intermediate ADP-heptose and/or related heptose phosphates. During C. jejuni infection of intestinal cells, the ALPK1-NF-κB axis was potently activated by released heptose metabolites without the need for a type III or type IV injection machinery. Our results classify ADP-heptose and/or related heptose phosphates as a major virulence factor of C. jejuni that may play an important role during Campylobacter infection in humans.


Subject(s)
Campylobacter Infections/immunology , Campylobacter jejuni/immunology , Epithelial Cells/immunology , Inflammation/immunology , Intestines/immunology , NF-kappa B/metabolism , Protein Kinases/metabolism , Campylobacter Infections/metabolism , Campylobacter Infections/microbiology , Cytokines , Epithelial Cells/metabolism , Epithelial Cells/microbiology , HeLa Cells , Humans , Immunity, Innate/immunology , Inflammation/metabolism , Inflammation/microbiology , Intestines/microbiology , NF-kappa B/genetics , Protein Kinases/genetics , Signal Transduction , Virulence , Virulence Factors/metabolism
3.
Am J Trop Med Hyg ; 105(3): 822-827, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34255738

ABSTRACT

Multiplex bead assays (MBAs) for serologic testing have become more prevalent in public health surveys, but few studies have assessed their test performance. As part of a trachoma study conducted in a rural part of Ethiopia in 2016, dried blood spots (DBS) were collected from a random sample of 393 children aged 0 to 9 years, with at least two separate 6-mm DBS collected on a filter card. Samples eluted from DBS were processed using an MBA on the Luminex platform for antibodies against 13 antigens of nine infectious organisms: Chlamydia trachomatis, Vibrio cholera, enterotoxigenic Escherichia coli, Cryptosporidium parvum, Entamoeba histolytica, Camplyobacter jejuni, Salmonella typhimurium Group B, Salmonella enteritidis Group D, and Giardia lamblia. Two separate DBS from each child were processed. The first DBS was run a single time, with the MBA set to read 100 beads per well. The second DBS was run twice, first at 100 beads per well and then at 50 beads per well. Results were expressed as the median fluorescence intensity minus background (MFI-BG), and classified as seropositive or seronegative according to external standards. Agreement between the three runs was high, with intraclass correlation coefficients of > 0.85 for the two Salmonella antibody responses and > 0.95 for the other 11 antibody responses. Agreement was also high for the dichotomous seropositivity indicators, with Cohen's kappa statistics exceeding 0.87 for each antibody assay. These results suggest that serologic testing on the Luminex platform had strong test performance characteristics for analyzing antibodies using DBS.


Subject(s)
Dried Blood Spot Testing/methods , Serologic Tests/methods , Campylobacter Infections/diagnosis , Campylobacter Infections/epidemiology , Campylobacter Infections/immunology , Campylobacter jejuni/immunology , Child , Child, Preschool , Chlamydia trachomatis/immunology , Cholera/diagnosis , Cholera/epidemiology , Cholera/immunology , Cryptosporidiosis/diagnosis , Cryptosporidiosis/epidemiology , Cryptosporidiosis/immunology , Cryptosporidium parvum/immunology , Entamoeba histolytica/immunology , Entamoebiasis/diagnosis , Entamoebiasis/epidemiology , Entamoebiasis/immunology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/diagnosis , Escherichia coli Infections/epidemiology , Escherichia coli Infections/immunology , Ethiopia/epidemiology , Female , Giardia lamblia/immunology , Giardiasis/diagnosis , Giardiasis/epidemiology , Giardiasis/immunology , Humans , Infant , Infant, Newborn , Male , Salmonella Infections/diagnosis , Salmonella Infections/epidemiology , Salmonella Infections/immunology , Salmonella enteritidis/immunology , Salmonella typhimurium/immunology , Sensitivity and Specificity , Seroepidemiologic Studies , Trachoma/diagnosis , Trachoma/epidemiology , Trachoma/immunology , Vibrio cholerae/immunology
4.
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
5.
PLoS One ; 16(5): e0251039, 2021.
Article in English | MEDLINE | ID: mdl-33951106

ABSTRACT

INTRODUCTION: While Campylobacter jejuni is a leading foodborne bacterial pathogen worldwide, it poses a particular risk to susceptible populations in low- and middle-income countries (LMICs). A capsule-conjugate vaccine approach has been proposed as a potential solution, but little information exists on circulating C. jejuni capsule types in LMICs. The capsule is the major serodeterminant of the Penner typing scheme, which is based on serum recognition of Campylobacter heat-stable antigens. We conducted a systematic review and meta-analysis to estimate the distribution of Penner serotypes associated with C. jejuni enteritis in LMICs. Vaccine coverage assessments for hypothetical regional and global C. jejuni vaccines were also estimated. METHODS: A systematic review of the literature published from 1980 to 2019 was performed using PubMed, Scopus, and Web of Science databases. Articles were assessed for eligibility and data were abstracted. Pooled C. jejuni serotype prevalence in LMICs was estimated by region and globally using random-effects models. RESULTS: A total of 36 studies were included, capturing 4,434 isolates from LMICs. Fifteen serotypes were present in a sufficient number of studies to be included in analyses. Among these, HS4c was the most common serotype globally (12.6%), though leading capsule types varied among regions. HS2, HS3c, HS4c, HS5/31, HS8/17, and HS10 were all among the 10 most common region-specific serotypes. CONCLUSIONS: The results of this review suggest that an octavalent vaccine could provide up to 66.9% coverage of typable strains worldwide, and 56.8-69.0% regionally. This review also highlights the paucity of available data on capsules in LMICs; more testing is needed to inform vaccine development efforts.


Subject(s)
Campylobacter Infections/immunology , Campylobacter jejuni/immunology , Bacterial Typing Techniques/methods , Campylobacter Infections/microbiology , Developing Countries , Humans , Prevalence , Serogroup , Serotyping/methods
6.
Infect Immun ; 89(8): e0011621, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34031129

ABSTRACT

Campylobacter spp. are the leading cause of bacterium-derived gastroenteritis worldwide, impacting 96 million individuals annually. Unlike other bacterial pathogens of the gastrointestinal tract, Campylobacter spp. lack many of the classical virulence factors that are often associated with the ability to induce disease in humans, including an array of canonical secretion systems and toxins. Consequently, the clinical manifestations of human campylobacteriosis and its resulting gastrointestinal pathology are believed to be primarily due to the host immune response toward the bacterium. Further, while gastrointestinal infection is usually self-limiting, numerous postinfectious disorders can occur, including the development of Guillain-Barré syndrome, reactive arthritis, and irritable bowel syndrome. Because gastrointestinal disease likely results from the host immune response, the development of these postinfectious disorders may be due to dysregulation or misdirection of the same inflammatory response. As a result, it is becoming increasingly important to the Campylobacter field, and human health, that the cellular immune responses toward Campylobacter be better understood, including which immunological events are critical to the development of disease and the postinfectious disorders mentioned above. In this review, we collectively cover the cellular immune responses across susceptible hosts to Campylobacter jejuni infection, along with the tissue pathology and postinfectious disorders which may develop.


Subject(s)
Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Campylobacter/immunology , Disease Susceptibility/immunology , Host-Pathogen Interactions/immunology , Immunity, Cellular , Animals , Campylobacter Infections/complications , Gastrointestinal Diseases/complications , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Humans
7.
Pol J Microbiol ; 70(1): 99-106, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33815531

ABSTRACT

Campylobacter fetus subsp. fetus is the causal agent of sporadic abortion in bovines and infertility that produces economic losses in livestock. In many infectious diseases, the immune response has an important role in limiting the invasion and proliferation of bacterial pathogens. Innate immune sensing of microorganisms is mediated by pattern-recognition receptors (PRRs) that identify pathogen-associated molecular patterns (PAMPs) and induces the secretion of several proinflammatory cytokines, like IL-1ß, TNF-α, and IL-8. In this study, the expression of IL-1ß, TNF-α, IL-8, and IFN-γ in bovine endometrial epithelial cells infected with C. fetus and Salmonella Typhimurium (a bacterial invasion control) was analyzed. The results showed that expression levels of IL-1ß and IL-8 were high at the beginning of the infection and decreased throughout the intracellular period. Unlike in this same assay, the expression levels of IFN-γ increased through time and reached the highest peak at 4 hours post infection. In cells infected with S. Typhimurium, the results showed that IL8 expression levels were highly induced by infection but not IFN-γ. In cells infected with S. Typhimurium or C. fetus subsp. fetus, the results showed that TNF-α expression did not show any change during infection. A cytoskeleton inhibition assay was performed to determine if cytokine expression was modified by C. fetus subsp. fetus intracellular invasion. IL-1ß and IL-8 expression were downregulated when an intracellular invasion was avoided. The results obtained in this study suggest that bovine endometrial epithelial cells could recognize C. fetus subsp. fetus resulting in early proinflammatory response.


Subject(s)
Campylobacter Infections/veterinary , Campylobacter/physiology , Cattle Diseases/immunology , Endometrium/immunology , Epithelial Cells/immunology , Animals , Campylobacter/genetics , Campylobacter Infections/genetics , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Cattle , Cattle Diseases/genetics , Cattle Diseases/microbiology , Cytokines/genetics , Cytokines/immunology , Endometrium/microbiology , Epithelial Cells/microbiology , Female , Host-Pathogen Interactions
8.
Gut Microbes ; 13(1): 1-16, 2021.
Article in English | MEDLINE | ID: mdl-33382951

ABSTRACT

Campylobacter jejuni is among the leading causes of bacterial foodborne illness. Poultry is the major reservoir and source of human campylobacteriosis. Currently, there is no effective and practical method to decrease C. jejuni colonization in chickens or to reduce human infections. Additionally, antibiotic-resistant infections pose a serious public health concern; therefore, antibiotic-alternative approaches are needed to reduce transmission of C. jejuni including resistant bacteria from chickens to humans. Here, we evaluated the effect of E. coli Nissle 1917 (EcN) on innate responses of polarized HT-29 cells and consequently on C. jejuni 81176 infections in HT-29 cells. Pre-treatment of HT-29 cells with EcN for 4 h had a significant effect on the invasion of different C. jejuni strains (2 h post-infection) (P < .05) and no intracellular C. jejuni (24 h post-infection) were recovered. To further understand how EcN mediates its impact on C. jejuni's survival inside the cells, we used Human Antibacterial RT2 ProfilerTM PCR arrays to profile gene expression in HT-29 cells after treatment with EcN with or without C. jejuni 81-176 infection. Our results suggest that pre-treatment of the HT-29 cells with EcN induced the anti-inflammatory cytokines and activated the anti-apoptotic Akt signaling which likely to protect the cells against the proinflammatory and apoptosis responses induced by C. jejuni. EcN also positively affected the expression of genes involved in cellular maintenance, growth, development, and proliferation. Further, EcN modulated the expression of genes involved in protective innate immunity, such as TLRs, ERK1/2, p38 MAPK, Ap1, JNK, IL1B, IL17A, and NF-κB signaling.


Subject(s)
Campylobacter jejuni/drug effects , Escherichia coli , Immunomodulation , Probiotics/pharmacology , Apoptosis/drug effects , Bacterial Adhesion/drug effects , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Cell Proliferation/drug effects , Cytokines/metabolism , Gene Regulatory Networks/drug effects , HT29 Cells , Humans , Immunity, Innate/drug effects , Microbial Viability/drug effects
9.
Mucosal Immunol ; 14(3): 703-716, 2021 05.
Article in English | MEDLINE | ID: mdl-33214656

ABSTRACT

Innate lymphoid cells (ILCs) are a heterogeneous family of immune regulators that protect against mucosal pathogens but can also promote intestinal pathology. Although the plasticity between ILCs populations has been described, the role of mucosal pathogens in inducing ILC conversion leading to intestinal pathology remains unclear. Here we demonstrate that IFNγ-producing ILCs are responsible for promoting intestinal pathology in a mouse model of enterocolitis caused by Campylobacter jejuni, a common human enteric pathogen. Phenotypic analysis revealed a distinct population of IFNγ-producing NK1.1-T-bet+ILCs that accumulated in the intestine of C. jejuni-infected mice. Adoptive transfer experiments demonstrated their capacity to promote intestinal pathology. Inactivation of T-bet in NKp46+ ILCs ameliorated disease. Transcriptome analysis and cell-fate mapping experiments revealed that IFNγ-producing NK1.1-ILCs correspond to ILC1 profile and develop from RORγt+ progenitors. Collectively, we identified a distinct population of NK1.1-ex-ILC3s that promotes intestinal pathology through IFNγ production in response to C. jejuni infection.


Subject(s)
Campylobacter Infections/immunology , Campylobacter jejuni/physiology , Colitis/immunology , Intestines/immunology , Lymphocytes/immunology , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Humans , Immunity, Innate , Interferon-gamma/metabolism , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Th1 Cells/immunology
10.
BMJ Case Rep ; 13(12)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33370970

ABSTRACT

Campylobacter species are known to cause enteritis. However, over the past 40-50 years, there have been reports of varying presentations, such as cellulitis, spondylodiscitis and bacteraemia. Of the Campylobacter species, Campylobacter jejuni is the most common culprit for causing bacteraemia, however, Campylobacter coli bacteraemia is becoming more prevalent. Here, we discuss an unusual case of C. coli bacteraemia in a patient with decompensated liver cirrhosis.


Subject(s)
Bacteremia/microbiology , Campylobacter Infections/microbiology , Campylobacter coli/isolation & purification , Colitis/microbiology , Liver Cirrhosis/complications , Anti-Bacterial Agents/therapeutic use , Antigens, Bacterial/isolation & purification , Bacteremia/diagnosis , Bacteremia/immunology , Campylobacter Infections/diagnosis , Campylobacter Infections/immunology , Campylobacter Infections/therapy , Campylobacter coli/immunology , Colitis/diagnosis , Colitis/immunology , Colitis/therapy , Drug Therapy, Combination , Electrolytes/administration & dosage , Feces/microbiology , Fluid Therapy/methods , Humans , Liver Cirrhosis/immunology , Male , Middle Aged
11.
Poult Sci ; 99(11): 5407-5414, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33142457

ABSTRACT

In recent years, several studies emphasize the deleterious effects of Campylobacter jejuni on the chicken intestine. In this context, it was shown that C. jejuni, contrary to the general belief, has a negative influence on the gut barrier in chickens. More precisely, we demonstrated that C. jejuni affects gut physiology characterized by changes in ion transport and transepithelial ion conductance, but the underlying mechanism is yet to be investigated. In the actual study, to determine epithelial paracellular permeability, the mucosal to serosal flux of 14C-mannitol in the small and large intestine was measured applying Ussing chamber. A total of seventy-five 1-day-old Ross 308 broiler chickens were housed in floor pens on wood shavings with feed and water provided ad libitum. Birds were randomly allocated to 3 different groups (n = 25 with 5 replicates/group) and infected at 14 d of age with a high (108 colony forming units [CFU]) or a low (104 CFU) dose of C. jejuni and a third group kept as noninfected control. Infection with the low dose of C. jejuni resulted in delayed cecal colonization but equalized at 21 d postinfection, independent of the dose. Invasion of liver and spleen with C. jejuni was only noticed in birds infected with 108 (CFU). Body weight (BW) and body weight gain of all birds infected with C. jejuni were lower than in the control group and varied with the dose of infection, confirming a negative correlation between the infection dose and birds BW. Mannitol flux in jejunum and cecum was significantly (P < 0.05) higher in all C. jejuni infected birds compared with control birds. Likewise, significant differences in mannitol flux of both jejunum and cecum were detected depending on the infection dose of C. jejuni. The correlation analyses revealed a positive relationship between Campylobacter dose and mannitol flux of both jejunum and cecum. Altogether, the actual results emphasize that the adverse effect of C. jejuni on gut permeability arises in a dose-dependent manner.


Subject(s)
Campylobacter Infections , Campylobacter jejuni , Poultry Diseases , Animals , Campylobacter Infections/immunology , Campylobacter Infections/veterinary , Cell Membrane Permeability/immunology , Chickens , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Poultry Diseases/immunology , Poultry Diseases/microbiology
12.
Gut Microbes ; 12(1): 1-25, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32887530

ABSTRACT

The symptoms of infectious diarrheal disease are mediated by a combination of a pathogen's virulence factors and the host immune system. Campylobacter jejuni is the leading bacterial cause of diarrhea worldwide due to its near-ubiquitous zoonotic association with poultry. One of the outstanding questions is to what extent the bacteria are responsible for the diarrheal symptoms via intestinal cell necrosis versus immune cell initiated tissue damage. To determine the stepwise process of inflammation that leads to diarrhea, we used a piglet ligated intestinal loop model to study the intestinal response to C. jejuni. Pigs were chosen due to the anatomical similarity between the porcine and the human intestine. We found that the abundance of neutrophil related proteins increased in the intestinal lumen during C. jejuni infection, including proteins related to neutrophil migration (neutrophil elastase and MMP9), actin reorganization (Arp2/3), and antimicrobial proteins (lipocalin-2, myeloperoxidase, S100A8, and S100A9). The appearance of neutrophil proteins also corresponded with increases of the inflammatory cytokines IL-8 and TNF-α. Compared to infection with the C. jejuni wild-type strain, infection with the noninvasive C. jejuni ∆ciaD mutant resulted in a blunted inflammatory response, with less inflammatory cytokines and neutrophil markers. These findings indicate that intestinal inflammation is driven by C. jejuni virulence and that neutrophils are the predominant cell type responding to C. jejuni infection. We propose that this model can be used as a platform to study the early immune events during infection with intestinal pathogens.


Subject(s)
Campylobacter Infections/immunology , Campylobacter jejuni/immunology , Cytokines/immunology , Intestine, Small/immunology , Intestine, Small/microbiology , Neutrophils/immunology , Animals , Campylobacter Infections/microbiology , Campylobacter jejuni/genetics , Campylobacter jejuni/metabolism , Campylobacter jejuni/pathogenicity , Cell Line , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Gastrointestinal Microbiome , Inflammation/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Small/pathology , Macrophages/immunology , Proteome/analysis , Swine , Swine, Miniature , Transcriptome , Virulence/genetics , Virulence Factors/metabolism
13.
Front Immunol ; 11: 1011, 2020.
Article in English | MEDLINE | ID: mdl-32582158

ABSTRACT

Campylobacter infection is one of the most common causes of bacterial gastroenteritis worldwide and a major global health threat due to the rapid development of antibiotic resistance. Currently, there are no vaccines approved to prevent campylobacteriosis, and rehydration is the main form of therapy. Secretory immunoglobulin A (SIgA) is the main antibody class found in mucous secretions, including human milk, and serves as the first line of defense for the gastrointestinal epithelium against enteric pathogens. In this study, we describe the prophylactic activity of orally delivered recombinant SIgA generated from two human monoclonal antibodies (CAA1 and CCG4) isolated for their reactivity against the flagellar-capping protein FliD, which is essential for bacteria motility and highly conserved across Campylobacter species associated with severe enteritis. In an immunocompetent weaned mouse model, a single oral administration of FliD-reactive SIgA CAA1 or CCG4 at 2 h before infection significantly enhances Campylobacter clearance at early stages post-infection, reducing the levels of inflammation markers associated with epithelial damage and polymorphonuclear (PMN) cells infiltration in the cecum lamina propria. Our data indicate that the prophylactic activity of CAA1 and CCG4 is not only dependent on the specificity to FliD but also on the use of the SIgA format, as the immunoglobulin G (IgG) versions of the same antibodies did not confer a comparable protective effect. Our work emphasizes the potential of FliD as a target for the development of vaccines and supports the concept that orally administered FliD-reactive SIgA can be developed to prevent or mitigate the severity of Campylobacter infections as well as the development of post-infection syndromes.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Campylobacter Infections/immunology , Campylobacter/physiology , Gastroenteritis/immunology , Immunotherapy/methods , Intestinal Mucosa/immunology , Neutrophils/immunology , Animals , Antibodies, Bacterial/metabolism , Antibodies, Monoclonal/metabolism , Disease Models, Animal , Disease Resistance , Female , Humans , Immunoglobulin A/metabolism , Mice , Mice, Inbred C57BL
14.
J Reprod Immunol ; 141: 103153, 2020 09.
Article in English | MEDLINE | ID: mdl-32570105

ABSTRACT

Campylobacter jejuni is recognized as a cause of miscarriage in animals, but rarely in humans. We describe here a case of spontaneous miscarriage at 12 weeks of gestation associated with Campylobacter jejuni bacteremia following digestive disorders. The patient was treated with azithromycin with good clinical evolution and underwent uterine aspiration during hospitalization. In our review of the literature, we found only 12 other miscarriages due to C. jejuni infections. Clinicians should consider this cause of miscarriage in febrile pregnant women, as the bacterium is resistant to many beta-lactam antibiotics, and macrolides are the first-line treatment.


Subject(s)
Abortion, Spontaneous/immunology , Bacteremia/complications , Campylobacter Infections/complications , Pregnancy Complications, Infectious/immunology , Abortion, Spontaneous/microbiology , Adult , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteremia/drug therapy , Bacteremia/immunology , Bacteremia/microbiology , Campylobacter Infections/drug therapy , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Campylobacter jejuni/immunology , Campylobacter jejuni/isolation & purification , Drug Resistance, Bacterial , Female , Humans , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/microbiology
15.
Neurogastroenterol Motil ; 32(10): e13875, 2020 10.
Article in English | MEDLINE | ID: mdl-32436301

ABSTRACT

BACKGROUND: Recent data substantiate the importance of acute gastroenteritis in the development of irritable bowel syndrome (IBS). An animal model of postinfectious IBS determined the importance of cytolethal distending toxin B (CdtB) during live Campylobacter jejuni infection and its development of autoimmunity to vinculin. In this study, we examine whether subcutaneous exposure to CdtB alone is sufficient to produce the postinfectious IBS effect and autoimmunity. METHODS: Sixty adult Sprague Dawley rats were randomized into 2 groups to receive subcutaneous injection of either CdtB or vehicle and administered a booster injection of the same product 3 weeks later. Serum was collected for anti-CdtB and anti-vinculin titers. Duodenal and ileal luminal contents for total eubacterial qPCR, and ileal bowel segments were harvested for vinculin and ileal expression. In a second experiment, 4 adult, Sprague Dawley rats were injected with either Cy7-labeled anti-CdtB and anti-vinculin antibodies were injected into the tail vein and imaged to determine organ localization of the antibodies. KEY RESULTS: Rats that received CdtB increased in serum anti-CdtB after injection. CdtB exposure also precipitated significant elevation in anti-vinculin antibodies (P < .001). This was associated with a reduction in intestinal vinculin expression (P < .001) that negatively correlated with serum anti-CdtB levels. CdtB exposure was also associated with greater levels of duodenal (P < .001) and ileal (P < .01) bacteria by qPCR that positively correlated with anti-CdtB levels. CONCLUSIONS AND INFERENCES: Rats injected with CdtB developed a postinfectious IBS-like phenotype and autoimmunity to vinculin with corresponding reduction in intestinal vinculin expression.


Subject(s)
Autoantibodies/immunology , Campylobacter Infections/immunology , Campylobacter jejuni/immunology , Intestine, Small/immunology , Irritable Bowel Syndrome/immunology , Vinculin/immunology , Animals , Autoantibodies/administration & dosage , Campylobacter Infections/complications , Immunization/methods , Intestine, Small/microbiology , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/microbiology , Male , Rats , Rats, Sprague-Dawley
16.
Cell Microbiol ; 22(8): e13210, 2020 08.
Article in English | MEDLINE | ID: mdl-32329205

ABSTRACT

Campylobacter jejuni is the leading cause of bacterial-derived gastroenteritis worldwide and can lead to several post-infectious inflammatory disorders. Despite the prevalence and health impacts of the bacterium, interactions between the host innate immune system and C. jejuni remain poorly understood. To expand on earlier work demonstrating that neutrophils traffic to the site of infection in an animal model of campylobacteriosis, we identified significant increases in several predominantly neutrophil-derived proteins in the faeces of C. jejuni-infected patients, including lipocalin-2, myeloperoxidase and neutrophil elastase. In addition to demonstrating that these proteins significantly inhibited C. jejuni growth, we determined they are released during formation of C. jejuni-induced neutrophil extracellular traps (NETs). Using quantitative and qualitative methods, we found that purified human neutrophils are activated by C. jejuni and exhibit signatures of NET generation, including presence of protein arginine deiminase-4, histone citrullination, myeloperoxidase, neutrophil elastase release and DNA extrusion. Production of NETs correlated with C. jejuni phagocytosis/endocytosis and invasion of neutrophils suggesting that host- and bacterial-mediated activities are responsible for NET induction. Further, NET-like structures were observed within intestinal tissue of C. jejuni-infected ferrets. Finally, induction of NETs significantly increased human colonocyte cytotoxicity, indicating that NET formation during C. jejuni infection may contribute to observed tissue pathology. These findings provide further understanding of C. jejuni-neutrophil interactions and inflammatory responses during campylobacteriosis.


Subject(s)
Campylobacter jejuni/immunology , Campylobacter jejuni/physiology , Extracellular Traps/immunology , Extracellular Traps/microbiology , Feces/chemistry , Host Microbial Interactions/immunology , Neutrophils/immunology , Animals , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Cells, Cultured , Colon/cytology , Colon/microbiology , Colon/pathology , Ferrets , Humans , Inflammation , Leukocyte Elastase/metabolism , Male , Neutrophils/chemistry , Neutrophils/microbiology , Phagocytosis
17.
Poult Sci ; 99(4): 2157-2161, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32241501

ABSTRACT

Oral administration of antibodies is a promising strategy against various infectious diseases. Previously, it was demonstrated that passive immunization by providing hyperimmune egg yolk through the feed reduces Campylobacter jejuni colonization in broilers. Campylobacteriosis is the most commonly reported bacterial foodborne zoonosis worldwide, and poultry products are the number one origin of these bacteria for human infection. To date, no effective control measures exist to limit Campylobacter colonization in the chicken's intestinal tract. Here, the effect of lyophilization of hyperimmune egg yolk on protection of broilers against C. jejuni was investigated. During an in vivo trial, broiler chickens were prophylactically given feed with lyophilized hyperimmune or non-immunized egg yolk powder starting from day 1 after hatch. At day 11, broilers were inoculated with C. jejuni according to a seeder model. Five days later, all broilers were euthanized and cecal content was examined for C. jejuni colonization. No decrease in C. jejuni colonization was found. The freeze-drying resulted in a 16-fold decrease of the antibody titer in the yolk powder compared to the fresh yolks, presumably caused by structural changes in the antibodies. In conclusion, applying freeze-dried hyperimmune egg yolk failed to protect broilers against C. jejuni colonization, possibly because lyophilization affected the antibodies' functionality.


Subject(s)
Antibodies, Bacterial/administration & dosage , Campylobacter Infections/veterinary , Chickens , Egg Yolk/immunology , Freeze Drying/veterinary , Immunization, Passive/veterinary , Poultry Diseases/prevention & control , Administration, Oral , Animals , Campylobacter/physiology , Campylobacter Infections/immunology , Campylobacter Infections/prevention & control , Female , Male , Poultry Diseases/immunology , Random Allocation
18.
Foodborne Pathog Dis ; 17(8): 485-493, 2020 08.
Article in English | MEDLINE | ID: mdl-31977245

ABSTRACT

This investigation was performed to assess the supplementation of probiotics on cytokine expression and lymphocyte subpopulation in Campylobacter coli challenged chickens. Thirty-six individuals were equally separated into four experimental treatments: C = untreated chickens, LB = probiotic control (Lactobacillus fermentum), Cc = Campylobacter-challenged control, LBCc = probiotic + Cc. All chicks were slaughtered and cecum samples were collected on day 4 postinfection. Gene expression analysis, using reverse transcription quantitative PCR (RT-qPCR), revealed significant differences in cytokine transcript expression between untreated and probiotic-treated chickens. In addition, flow cytometry was used to quantitate the levels of lymphocyte subpopulations. Principal component analysis showed that probiotic administration induced an overall downregulation of cytokine expression. C. coli exposure provoked a similar response to that of L. fermentum but to a lesser extent. Colonization of C. coli in the presence of the probiotic evoked a complex response with an upregulation of some type II cytokines, including interleukin IL-4 and IL-13, which could explain the increased presence of antibodies in both lamina propria and epithelium. Moreover, despite that the percentage of CD8 intraepithelial lymphocytes (IELs) was found to be higher, downregulation of proinflammatory cytokines IL-15, IL-16, and interferon γ was observed. This suggests that the detected CD8 are not effector cells but induced IELs, which release antimicrobial peptides, and are ready to be primed upon encountering antigen. These outcomes demonstrate that probiotic administration promotes a humoral response to a C. coli infection while dampening any potential inflammation mediated by effector T cells in 1-week-old chicks.


Subject(s)
Campylobacter Infections/veterinary , Chickens/immunology , Cytokines/immunology , Limosilactobacillus fermentum , Lymphocyte Subsets , Poultry Diseases/microbiology , Probiotics/therapeutic use , Animals , Campylobacter Infections/immunology , Campylobacter coli , Chickens/microbiology , Male , Poultry Diseases/immunology
19.
Front Immunol ; 11: 579615, 2020.
Article in English | MEDLINE | ID: mdl-33488580

ABSTRACT

Human pathogen Campylobacter jejuni is a significant risk factor for the development of long-term intestinal dysfunction although the cellular and molecular mechanisms remain scantily defined. IL-23 is an emerging therapeutic target for the treatment of inflammatory intestinal diseases, however its role in C. jejuni-driven intestinal pathology is not fully understood. IL-10 deficient mice represent a robust model to study the pathogenesis of C. jejuni infection because C. jejuni infection of mice lacking IL-10 results in symptoms and pathology that resemble human campylobacteriosis. To determine the role of IL-23 in C. jejuni-driven intestinal inflammation, we studied the disease pathogenesis in IL-23-/- mice with inhibited IL-10Rα signaling. These mice exhibited reduced intestinal pathology independent from bacterial clearance. Further, levels of IFNγ, IL-17, IL-22, TNF, and IL-6 were reduced and associated with reduced accumulation of neutrophils, monocytes and macrophages in the colon. Flow cytometry analysis revealed reduced production of IL-17 and IFNγ by group 1 and 3 innate lymphoid cells. Thus, our data suggest that IL-23 contributes to intestinal inflammation in C. jejuni infected mice by promoting IL-17 and IFNγ production by innate lymphoid cells.


Subject(s)
Campylobacter Infections/immunology , Campylobacter jejuni/physiology , Colitis/immunology , Interleukin-23/metabolism , Intestines/pathology , Lymphocytes/immunology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Immunity, Innate , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-17/metabolism , Interleukin-23/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
20.
Sci Rep ; 9(1): 19797, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875037

ABSTRACT

Conventional mice are protected from Campylobacter jejuni infection by the murine host-specific gut microbiota composition. We here addressed whether peroral fecal microbiota transplantation (FMT) might be an antibiotics-independent option to lower even high gastrointestinal C. jejuni loads in the infected vertebrate host. To address this, secondary abiotic mice were generated by broad-spectrum antibiotic treatment and perorally infected with C. jejuni by gavage. One week later, mice were stably colonized with more than 109 C. jejuni and subjected to peroral FMT from murine donors on three consecutive days. Two weeks post-intervention, gastrointestinal C. jejuni loads were up to 7.5 orders of magnitude lower following murine FMT versus mock challenge. Remarkably, FMT reversed C. jejuni induced colonic epithelial apoptosis, but enhanced proliferative and regenerative responses in the colon thereby counteracting pathogenic cell damage. Furthermore, FMT dampened both, innate and adaptive immune cell responses in the large intestines upon C. jejuni infection that were accompanied by less C. jejuni-induced colonic nitric oxide secretion. Our study provides strong evidence that novel probiotic formulations developed as alternative option to FMT in severe intestinal inflammatory morbidities including Clostridoides difficile infection might be effective to treat campylobacteriosis and lower pathogen loads in colonized vertebrates including farm animals.


Subject(s)
Anti-Bacterial Agents/pharmacology , Campylobacter Infections/immunology , Colon/microbiology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Animals , Apoptosis , Bacterial Load , Campylobacter jejuni , Colon/pathology , Epithelium/metabolism , Female , Immune System , Inflammation , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...