1
|
Sung K, Gao Y, Yu LR, Chon J, Hiett KL, Line JE, Kweon O, Park M, Khan SA. Phenotypic, genotypic and proteomic variations between poor and robust colonizing Campylobacter jejuni strains. Microb Pathog 2024; 193:106766. [PMID: 38942248 DOI: 10.1016/j.micpath.2024.106766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024]
Abstract
Campylobacter jejuni is one of the major causes of bacterial gastrointestinal disease in humans worldwide. This foodborne pathogen colonizes the intestinal tracts of chickens, and consumption of chicken and poultry products is identified as a common route of transmission. We analyzed two C. jejuni strains after oral challenge with 105 CFU/ml of C. jejuni per chick; one strain was a robust colonizer (A74/C) and the other a poor colonizer (A74/O). We also found extensive phenotypic differences in growth rate, biofilm production, and in vitro adherence, invasion, intracellular survival, and transcytosis. Strains A74/C and A74/O were genotypically similar with respect to their whole genome alignment, core genome, and ribosomal MLST, MLST, flaA, porA, and PFGE typing. The global proteomes of the two congenic strains were quantitatively analyzed by ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) and 618 and 453 proteins were identified from A74/C and A74/O isolates, respectively. Cluster of Orthologous Groups (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses showed that carbon metabolism and motility proteins were distinctively overexpressed in strain A74/C. The robust colonizer also exhibited a unique proteome profile characterized by significantly increased expression of proteins linked to adhesion, invasion, chemotaxis, energy, protein synthesis, heat shock proteins, iron regulation, two-component regulatory systems, and multidrug efflux pump. Our study underlines phenotypic, genotypic, and proteomic variations of the poor and robust colonizing C. jejuni strains, suggesting that several factors may contribute to mediating the different colonization potentials of the isogenic isolates.
Collapse
Affiliation(s)
- Kidon Sung
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration (US FDA), Jefferson, AR, 72079, USA.
| | - Yuan Gao
- Division of Systems Biology, National Center for Toxicological Research, US FDA, Jefferson, AR, 72079, USA
| | - Li-Rong Yu
- Division of Systems Biology, National Center for Toxicological Research, US FDA, Jefferson, AR, 72079, USA
| | - Jungwhan Chon
- Department of Companion Animal Health, Inje University, Gimhae, South Korea
| | - Kelli L Hiett
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US FDA, Laurel, MD, 20708, USA
| | - J Eric Line
- Bacterial Epidemiology and Antimicrobial Resistance Research Unit, Agricultural Research Service, U.S. Department of Agriculture (USDA), Athens, GA, 30605, USA
| | - Ohgew Kweon
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration (US FDA), Jefferson, AR, 72079, USA
| | - Miseon Park
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration (US FDA), Jefferson, AR, 72079, USA
| | - Saeed A Khan
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration (US FDA), Jefferson, AR, 72079, USA
| |
Collapse
|
2
|
Chen H, Zhang Y, Pan Y, Wu L, Wang W, Zhang H, Lou H. Antibiotic-induced microbiome depletion promotes intestinal colonization by Campylobacter jejuni in mice. BMC Microbiol 2024; 24:156. [PMID: 38724913 PMCID: PMC11080253 DOI: 10.1186/s12866-024-03313-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
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.
Collapse
Affiliation(s)
- Haohao Chen
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China.
| | - Yanfang Zhang
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Yi Pan
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Lin Wu
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Wenqian Wang
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Hui Zhang
- Animal Center, Jinhua Food and Drug Inspection and Testing Research Institute, Jinhua, Zhejiang Province, P.R. China
| | - Hongqiang Lou
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China.
| |
Collapse
|
3
|
Fahim SM, Donowitz JR, Smirnova E, Jan NJ, Das S, Mahfuz M, Gaffar SMA, Petri WA, Marie C, Ahmed T. Small Intestine Bacterial Overgrowth is associated with increased Campylobacter and epithelial injury in duodenal biopsies of Bangladeshi children. PLoS Negl Trop Dis 2024; 18:e0012023. [PMID: 38536881 PMCID: PMC11020352 DOI: 10.1371/journal.pntd.0012023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 04/16/2024] [Accepted: 02/26/2024] [Indexed: 04/18/2024] Open
Abstract
Small intestine bacterial overgrowth (SIBO) has been associated with enteric inflammation, linear growth stunting, and neurodevelopmental delays in children from low-income countries. Little is known about the histologic changes or epithelial adherent microbiota associated with SIBO. We sought to describe these relationships in a cohort of impoverished Bangladeshi children. Undernourished 12-18-month-old children underwent both glucose hydrogen breath testing for SIBO and duodenoscopy with biopsy. Biopsy samples were subject to both histological scoring and 16s rRNA sequencing. 118 children were enrolled with 16s sequencing data available on 53. Of 11 histological features, we found that SIBO was associated with one, enterocyte injury in the second part of the duodenum (R = 0.21, p = 0.02). SIBO was also associated with a significant increase in Campylobacter by 16s rRNA analysis (Log 2-fold change of 4.43; adjusted p = 1.9 x 10-6). These findings support the growing body of literature showing an association between SIBO and enteric inflammation and enterocyte injury and further delineate the subgroup of children with environmental enteric dysfunction who have SIBO. Further, they show a novel association between SIBO and Campylobacter. Mechanistic work is needed to understand the relationship between SIBO, enterocyte injury, and Campylobacter.
Collapse
Affiliation(s)
- Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States of America
| | - Jeffrey R. Donowitz
- Division of Pediatric Infectious Diseases, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, Virginia, United States of America
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
- Division of Pediatric Infectious Diseases, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ekaterina Smirnova
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Ning-Juin Jan
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - S. M. Abdul Gaffar
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - William A. Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Chelsea Marie
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Office of the Executive Director, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| |
Collapse
|
4
|
Shayya NW, Bandick R, Busmann LV, Mousavi S, Bereswill S, Heimesaat MM. Metabolomic signatures of intestinal colonization resistance against Campylobacter jejuni in mice. Front Microbiol 2023; 14:1331114. [PMID: 38164399 PMCID: PMC10757985 DOI: 10.3389/fmicb.2023.1331114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Campylobacter jejuni stands out as one of the leading causes of bacterial enteritis. In contrast to humans, specific pathogen-free (SPF) laboratory mice display strict intestinal colonization resistance (CR) against C. jejuni, orchestrated by the specific murine intestinal microbiota, as shown by fecal microbiota transplantation (FMT) earlier. Methods Murine infection models, comprising SPF, SAB, hma, and mma mice were employed. FMT and microbiota depletion were confirmed by culture and culture-independent analyses. Targeted metabolome analyses of fecal samples provided insights into the associated metabolomic signatures. Results In comparison to hma mice, the murine intestinal microbiota of mma and SPF mice (with CR against C. jejuni) contained significantly elevated numbers of lactobacilli, and Mouse Intestinal Bacteroides, whereas numbers of enterobacteria, enterococci, and Clostridium coccoides group were reduced. Targeted metabolome analysis revealed that fecal samples from mice with CR contained increased levels of secondary bile acids and fatty acids with known antimicrobial activities, but reduced concentrations of amino acids essential for C. jejuni growth as compared to control animals without CR. Discussion The findings highlight the role of microbiota-mediated nutrient competition and antibacterial activities of intestinal metabolites in driving murine CR against C. jejuni. The study underscores the complex dynamics of host-microbiota-pathogen interactions and sets the stage for further investigations into the mechanisms driving CR against enteric infections.
Collapse
|
5
|
Bandick R, Busmann LV, Mousavi S, Shayya NW, Piwowarski JP, Granica S, Melzig MF, Bereswill S, Heimesaat MM. Therapeutic Effects of Oral Application of Menthol and Extracts from Tormentil ( Potentilla erecta), Raspberry Leaves ( Rubus idaeus), and Loosestrife ( Lythrum salicaria) during Acute Murine Campylobacteriosis. Pharmaceutics 2023; 15:2410. [PMID: 37896170 PMCID: PMC10610364 DOI: 10.3390/pharmaceutics15102410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Human food-borne infections with the enteropathogen Campylobacter jejuni are becoming increasingly prevalent worldwide. Since antibiotics are usually not indicated in campylobacteriosis, alternative treatment regimens are important. We here investigated potential disease-alleviating effects of menthol and of extracts from tormentil, raspberry leaves, and loosestrife in acute murine campylobacteriosis. Therefore, C. jejuni-infected microbiota-depleted IL-10-/- mice were orally treated with the compounds alone or all in combination from day 2 until day 6 post-infection. Whereas neither treatment regimen affected gastrointestinal pathogen loads, the combination of compounds alleviated C. jejuni-induced diarrheal symptoms in diseased mice on day 6 post-infection. Furthermore, the therapeutic application of tormentil and menthol alone and the combination of the four compounds resulted in lower colonic T cell numbers in infected mice when compared to placebo counterparts. Notably, pro-inflammatory cytokines measured in mesenteric lymph nodes taken from C. jejuni-infected mice following tormentil, menthol, and combination treatment did not differ from basal concentrations. However, neither treatment regimen could dampen extra-intestinal immune responses, including systemic pro-inflammatory cytokine secretion on day 6 post-infection. In conclusion, the combination of menthol and of extracts from tormentil, raspberry leaves, and loosestrife constitutes an antibiotic-independent approach to alleviate campylobacteriosis symptoms.
Collapse
Affiliation(s)
- Rasmus Bandick
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, D-12203 Berlin, Germany
| | - Lia V Busmann
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, D-12203 Berlin, Germany
| | - Soraya Mousavi
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, D-12203 Berlin, Germany
| | - Nizar W Shayya
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, D-12203 Berlin, Germany
| | - Jakub P Piwowarski
- Microbiota Lab, Department of Pharmaceutical Biology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Sebastian Granica
- Microbiota Lab, Department of Pharmaceutical Biology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Matthias F Melzig
- Institute of Pharmacy, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, D-12203 Berlin, Germany
| | - Markus M Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, D-12203 Berlin, Germany
| |
Collapse
|
6
|
Fonseca M, Heider LC, Stryhn H, McClure JT, Léger D, Rizzo D, Warder L, Dufour S, Roy JP, Kelton DF, Renaud D, Barkema HW, Sanchez J. Antimicrobial use and its association with the isolation of and antimicrobial resistance in Campylobacter spp. recovered from fecal samples from Canadian dairy herds: A cross-sectional study. Prev Vet Med 2023; 215:105925. [PMID: 37104967 DOI: 10.1016/j.prevetmed.2023.105925] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 04/07/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023]
Abstract
Campylobacteriosis is one of the most common zoonotic diseases in North America. As opposed to humans, animal infections caused by Campylobacter spp. are often asymptomatic. In this study, data collected through the Canadian Dairy Network for Antimicrobial Stewardship surveillance system were used to determine the proportion of Campylobacter spp. and antimicrobial resistant isolates recovered from dairy cattle herds. Additionally, the association of antimicrobial use (AMU) with fecal carriage and antimicrobial resistance (AMR) of Campylobacter spp. were investigated. Pooled fecal samples from 5 animals from each production phase (pre-weaned calves, post-weaned heifers, lactating cows), and a manure storage sample were collected from 140 dairy herds across Canada. Samples were cultured using selective media, and Campylobacter isolates were speciated using matrix-assisted laser desorption/ionization-time of flight mass spectrometry. Antimicrobial susceptibilities were determined using the minimum inhibitory concentration test, and interpretation was made according to the Clinical and Laboratory Standards Institute. Two multilevel logistic regression models were used to investigate the association between the AMU with the isolation and antimicrobial resistance in Campylobacter spp. Of 560 samples, 63.8% were positive for Campylobacter spp., and 96% of the participating farms had at least one sample source (i.e., calves, heifers, lactating cows, or manure storage) positive for Campylobacter spp. Overall, 54.3% of the Campylobacter spp. isolates were resistant to at least one antimicrobial. Resistance to tetracycline was observed in 49.7% of the Campylobacter spp. isolates, followed by ciprofloxacin (19.9%) and nalidixic acid (19.3%). The proportion of multi-drug resistant (≥3 antimicrobial classes) Campylobacter spp. isolates was low (0.3%); however, 15.6% were resistant to two different classes of antimicrobials. Samples collected from lactating cows, heifers, and manure storage were more likely to be positive for Campylobacter spp. compared to calves. Total AMU was associated with a decreased probability of recovering Campylobacter spp. In addition, AMR to either tetracycline or ciprofloxacin had an interaction with antimicrobial use. The probability of resistance to tetracycline increased for each unit increase in the total AMU (Defined Course Dose/100 animal-years), while the probability of resistance to ciprofloxacin decreased. Campylobacter coli isolates were more likely to be resistant to ciprofloxacin and tetracycline when compared to C. jejuni. Our study demonstrated that Campylobacter spp. is widespread among Canadian dairy farms, and a higher proportion of resistance to tetracycline was identified. The total AMU was associated with increased resistance to tetracycline in Campylobacter spp. isolates; however, for ciprofloxacin the AMU was associated with decreased resistance.
Collapse
Affiliation(s)
- Mariana Fonseca
- Department of Health Management, University of Prince Edward Island, Charlottetown, PE, Canada.
| | - Luke C Heider
- Department of Health Management, University of Prince Edward Island, Charlottetown, PE, Canada
| | - Henrik Stryhn
- Department of Health Management, University of Prince Edward Island, Charlottetown, PE, Canada
| | - J Trenton McClure
- Department of Health Management, University of Prince Edward Island, Charlottetown, PE, Canada
| | - David Léger
- Public Health Agency of Canada, Center for Foodborne, Environmental and Zoonotic Infectious Diseases, Guelph, ON, Canada
| | - Daniella Rizzo
- Public Health Agency of Canada, Center for Foodborne, Environmental and Zoonotic Infectious Diseases, Guelph, ON, Canada
| | - Landon Warder
- Department of Health Management, University of Prince Edward Island, Charlottetown, PE, Canada
| | - Simon Dufour
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Jean-Philippe Roy
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - David F Kelton
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - David Renaud
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Javier Sanchez
- Department of Health Management, University of Prince Edward Island, Charlottetown, PE, Canada
| |
Collapse
|
7
|
Abstract
The major function of the mammalian immune system is to prevent and control infections caused by enteropathogens that collectively have altered human destiny. In fact, as the gastrointestinal tissues are the major interface of mammals with the environment, up to 70% of the human immune system is dedicated to patrolling them The defenses are multi-tiered and include the endogenous microflora that mediate colonization resistance as well as physical barriers intended to compartmentalize infections. The gastrointestinal tract and associated lymphoid tissue are also protected by sophisticated interleaved arrays of active innate and adaptive immune defenses. Remarkably, some bacterial enteropathogens have acquired an arsenal of virulence factors with which they neutralize all these formidable barriers to infection, causing disease ranging from mild self-limiting gastroenteritis to in some cases devastating human disease.
Collapse
Affiliation(s)
- Micah J. Worley
- Department of Biology, University of Louisville, Louisville, Kentucky, USA,CONTACT Micah J. Worley Department of Biology, University of Louisville, 139 Life Sciences Bldg, Louisville, Kentucky, USA
| |
Collapse
|
8
|
Luo Y, Wang J, Wang C, Wang D, Li C, Zhang B, Zhong X, Chen L, Li H, Su H, Zheng Q, Zhu D, Tang H, Guo L. The fecal arsenic excretion, tissue arsenic accumulation, and metabolomics analysis in sub-chronic arsenic-exposed mice after in situ arsenic-induced fecal microbiota transplantation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 854:158583. [PMID: 36084774 DOI: 10.1016/j.scitotenv.2022.158583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 06/15/2023]
Abstract
Arsenic can be specifically enriched by rice, and the health hazards caused by high arsenic rice are gradually attracting attention. This study aimed to explore the potential of microbial detoxification via gut microbiome in the treatment of sub-chronic arsenic poisoning. We first exposed mice to high-dose arsenic feed (30 mg/kg, rice arsenic composition) for 60 days to promote arsenic-induced microbes in situ in the gastrointestinal tract, then transplanted their fecal microbiota (FMT) into another batch of healthy recipient mice, and dynamically monitored the microbial colonization by 16S rRNA sequencing and ITS sequencing. The results showed that in situ arsenic-induced fecal microbiome can stably colonized and interact with indigenous microbes in the recipient mice in two weeks, and established a more stable network of gut microbiome. Then, the recipient mice continued to receive high-dose arsenic exposure for 52 days. After above sub-chronic arsenic exposure, compared with the non-FMT group, fecal arsenic excretion, liver and plasma arsenic accumulation were significantly lower (P < 0.05), and that in kidney, hair, and thighbone present no significant differences. Metabolomics of feces- plasma-brain axis were also disturbed, some up-regulated metabolites in feces, plasma, and cerebral cortex may play positive roles for the host. Therefore, microbial detoxification has potential in the treatment of sub-chronic arsenic poisoning. However, gut flora is an extremely complex community with different microorganisms have different arsenic metabolizing abilities, and various microbial metabolites. Coupled with the matrix effects, these factors will have various effects on the efflux and accumulation of arsenic. The definite effects (detoxification or non-detoxification) could be not assured based on the current study, and more systematic and rigorous studies are needed in the future.
Collapse
Affiliation(s)
- Yu Luo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Jiating Wang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Chenfei Wang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518000, China
| | - Dongbin Wang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Chengji Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Bin Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Xiaoting Zhong
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Linkang Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Hao Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Hongtian Su
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Qiuyi Zheng
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Dajian Zhu
- Department of Surgery, Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan 528399, China.
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Lianxian Guo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
9
|
Herzog MKM, Cazzaniga M, Peters A, Shayya N, Beldi L, Hapfelmeier S, Heimesaat MM, Bereswill S, Frankel G, Gahan CG, Hardt WD. Mouse models for bacterial enteropathogen infections: insights into the role of colonization resistance. Gut Microbes 2023; 15:2172667. [PMID: 36794831 PMCID: PMC9980611 DOI: 10.1080/19490976.2023.2172667] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
Globally, enteropathogenic bacteria are a major cause of morbidity and mortality.1-3 Campylobacter, Salmonella, Shiga-toxin-producing Escherichia coli, and Listeria are among the top five most commonly reported zoonotic pathogens in the European Union.4 However, not all individuals naturally exposed to enteropathogens go on to develop disease. This protection is attributable to colonization resistance (CR) conferred by the gut microbiota, as well as an array of physical, chemical, and immunological barriers that limit infection. Despite their importance for human health, a detailed understanding of gastrointestinal barriers to infection is lacking, and further research is required to investigate the mechanisms that underpin inter-individual differences in resistance to gastrointestinal infection. Here, we discuss the current mouse models available to study infections by non-typhoidal Salmonella strains, Citrobacter rodentium (as a model for enteropathogenic and enterohemorrhagic E. coli), Listeria monocytogenes, and Campylobacter jejuni. Clostridioides difficile is included as another important cause of enteric disease in which resistance is dependent upon CR. We outline which parameters of human infection are recapitulated in these mouse models, including the impact of CR, disease pathology, disease progression, and mucosal immune response. This will showcase common virulence strategies, highlight mechanistic differences, and help researchers from microbiology, infectiology, microbiome research, and mucosal immunology to select the optimal mouse model.
Collapse
Affiliation(s)
- Mathias K.-M. Herzog
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Monica Cazzaniga
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Audrey Peters
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Nizar Shayya
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Luca Beldi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | - Markus M. Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Gad Frankel
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Cormac G.M. Gahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Heimesaat MM, Mousavi S, Bandick R, Bereswill S. Campylobacter jejuni infection induces acute enterocolitis in IL-10-/- mice pretreated with ampicillin plus sulbactam. Eur J Microbiol Immunol (Bp) 2022; 12:73-83. [PMID: 36069779 PMCID: PMC9530677 DOI: 10.1556/1886.2022.00014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/19/2022] [Indexed: 11/19/2022] Open
Abstract
Gut microbiota depletion is a pivotal prerequisite to warrant Campylobacter jejuni infection and induced inflammation in IL-10-/- mice used as acute campylobacteriosis model. We here assessed the impact of an 8-week antibiotic regimen of ampicillin, ciprofloxacin, imipenem, metronidazole, and vancomycin (ABx) as compared to ampicillin plus sulbactam (A/S) on gut microbiota depletion and immunopathological responses upon oral C. jejuni infection. Our obtained results revealed that both antibiotic regimens were comparably effective in depleting the murine gut microbiota facilitating similar pathogenic colonization alongside the gastrointestinal tract following oral infection. Irrespective of the preceding microbiota depletion regimen, mice were similarly compromised by acute C. jejuni induced enterocolitis as indicated by comparable clinical scores and macroscopic as well as microscopic sequelae such as colonic histopathology and apoptosis on day 6 post-infection. Furthermore, innate and adaptive immune cell responses in the large intestines were similar in both infected cohorts, which also held true for intestinal, extra-intestinal and even systemic secretion of pro-inflammatory cytokines such as TNF-α, IFN-γ, and IL-6. In conclusion, gut microbiota depletion in IL-10-/- mice by ampicillin plus sulbactam is sufficient to investigate both, C. jejuni infection and the immunopathological features of acute campylobacteriosis.
Collapse
Affiliation(s)
- Markus M. Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 12203, Berlin, Germany
| | - Soraya Mousavi
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 12203, Berlin, Germany
| | - Rasmus Bandick
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 12203, Berlin, Germany
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 12203, Berlin, Germany
| |
Collapse
|
11
|
Brudvig JM, Cluett MM, Gensterblum-Miller EU, Chen J, Bell JA, Mansfield LS. Th1/Th17-mediated Immunity and Protection from Peripheral Neuropathy in Wildtype and IL10 -/- BALB/c Mice Infected with a Guillain-Barré Syndrome-associated Campylobacter jejuni Strain. Comp Med 2022; 72:63-77. [PMID: 35272743 PMCID: PMC9084571 DOI: 10.30802/aalas-cm-21-000059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/07/2021] [Accepted: 09/03/2021] [Indexed: 11/05/2022]
Abstract
Campylobacter jejuni is an important cause of bacterial gastroenteritis worldwide and is linked to Guillain-Barré syndrome (GBS), a debilitating postinfectious polyneuropathy. The immunopathogenesis of GBS involves the generation of antibodies that are cross reactive to C. jejuni lipooligosaccharide and structurally similar peripheral nerve gangliosides. Both the C. jejuni infecting strain and host factors contribute to GBS development. GBS pathogenesis is associated with Th2-mediated responses in patients. Moreover, induction of IgG1 antiganglioside antibodies in association with colonic Th2-mediated immune responses has been reported in C. jejuni-infected C57BL/6 IL10-/- mice at 4 to 6 wk after infection. We hypothesized that, due to their Th2 immunologic bias, BALB/c mice would develop autoantibodies and signs of peripheral neuropathy after infection with a GBS patient-derived strain of C. jejuni (strain 260.94). WT and IL10-/- BALB/c mice were orally inoculated with C. jejuni 260.94, phenotyped weekly for neurologic deficits, and euthanized after 5 wk. Immune responses were assessed as C. jejuni-specific and antiganglioside antibodies in plasma and cytokine production and histologic lesions in the proximal colon. Peripheral nerve lesions were assessed in dorsal root ganglia and their afferent nerve fibers by scoring immunohistochemically labeled macrophages through morphometry. C. jejuni 260.94 stably colonized both WT and IL10-/- mice and induced systemic Th1/Th17-mediated immune responses with significant increases in C. jejuni-specific IgG2a, IgG2b, and IgG3 plasma antibodies. However, C. jejuni 260.94 did not induce IgG1 antiganglioside antibodies, colitis, or neurologic deficits or peripheral nerve lesions in WT or IL10-/- mice. Both WT and IL10-/- BALB/c mice showed relative protection from development of Th2-mediated immunity and antiganglioside antibodies as compared with C57BL/6 IL10-/- mice. Therefore, BALB/c mice infected with C. jejuni 260.94 are not an effective disease model but provide the opportunity to study the role of immune mechanisms and host genetic background in the susceptibility to post infectious GBS.
Collapse
Affiliation(s)
- Jean M Brudvig
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Matthew M Cluett
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Elizabeth U Gensterblum-Miller
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - James Chen
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - Julia A Bell
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - Linda S Mansfield
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan;,
| |
Collapse
|
12
|
Saha P, Banerjee A, Banerjee G, Bag PK. Inhibitory activities of Typhonium trilobatum (L.) Schott on virulence potential of multi-drug resistant toxigenic Vibrio cholerae. Microb Pathog 2022; 165:105485. [DOI: 10.1016/j.micpath.2022.105485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/29/2022]
|
13
|
A Review of the Advantages, Disadvantages and Limitations of Chemotaxis Assays for Campylobacter spp. Int J Mol Sci 2022; 23:ijms23031576. [PMID: 35163499 PMCID: PMC8836060 DOI: 10.3390/ijms23031576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 11/26/2022] Open
Abstract
Reproducible qualitative and quantitative assessment of bacterial chemotactic motility, particularly in response to chemorepellent effectors, is experimentally challenging. Here we compare several established chemotaxis assays currently used to investigate Campylobacter jejuni chemotaxis, with the aim of improving the correlation between different studies and establishing the best practices. We compare the methodologies of capillary, agar, and chamber-based assays, and discuss critical technical points, in terms of reproducibility, accuracy, and the advantages and limitations of each.
Collapse
|
14
|
Ruiz MJ, Sirini NE, Signorini ML, Etcheverría A, Zbrun MV, Soto LP, Zimmermann JA, Frizzo LS. Protective effect of Lactiplantibacillus plantarum LP5 in a murine model of colonisation by Campylobacter coli DSPV458. Benef Microbes 2021; 12:553-565. [PMID: 34590533 DOI: 10.3920/bm2021.0010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Thermotolerant Campylobacter species are the leading cause of foodborne bacterial diarrheal disease worldwide. Campylobacter coli, abundant in pigs and pork products, have been identified as a source of human infection. In this study, we propose the use of Lactiplantibacillus plantarum LP5 as a probiotic to reduce colonisation of this intestinal pathogen in a murine colonisation model of C. coli DSPV458. Six-week-old adult female Balb/cCmedc mice were housed in groups: Control, Campy and Pro-Campy. Control and Pro-Campy groups received antibiotics for 5 days and the Campy group for 12 days. Pro-Campy group was inoculated for 7 days with 8.78 log10 cfu total of L. plantarum LP5 suspended in De Man, Rogosa and Sharpe broth. All groups were inoculated with 6.72 log10 cfu of C. coli DSPV458 suspended in brain heart infusion broth. L. plantarum LP5 was recovered only in the Pro- Campy group. C. coli DSPV458 was recovered at higher levels in the Control and Campy groups. The differences with the Pro-Campy group were significant. As regards faeces, Control and Campy groups reached 7.41 and 7.84 log10 cfu/g, respectively, and the Pro-Campy group only 4.62 log10 cfu/g. In the caecum, Control and Campy groups reached 8.01 and 9.26 log10cfu/g, respectively, and the Pro-Campy group only 4.51 log10 cfu/g. In the ileum, Control and Campy groups reached 3.43 and 3.26 log10 cfu/g, respectively, and the Pro-Campy group did not show detectable levels. The reduction of C. coli DSPV458 in the Pro-Campy group compared to the Control group in faeces, caecum and ileum was 99.55, 99.98 and 100%, respectively. Animals were maintained under normal health conditions, and haematological parameters were within the standard values for Balb/cCmedc. The incorporation of a probiotic generated a protective effect in the mice colonisation model. The protective effect would also apply to intestinal colonisation by indigenous enterobacteria. Therefore, the strategy used in this study is of great importance to understand the protection mechanisms in a murine model, as well as its application in food-producing animals.
Collapse
Affiliation(s)
- M J Ruiz
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Animal Health and Preventive Medicine, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires, Tandil, Argentina
| | - N E Sirini
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - M L Signorini
- Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,National Council of Scientific and Technical Research, National Institute of Agricultural Technology EEA Rafaela, Ruta 34 Km 227, 2300 Rafaela, Province of Santa Fe, Argentina
| | - A Etcheverría
- Department of Animal Health and Preventive Medicine, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires, Tandil, Argentina
| | - M V Zbrun
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - L P Soto
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - J A Zimmermann
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - L S Frizzo
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| |
Collapse
|
15
|
Evidence of MHC class I and II influencing viral and helminth infection via the microbiome in a non-human primate. PLoS Pathog 2021; 17:e1009675. [PMID: 34748618 PMCID: PMC8601626 DOI: 10.1371/journal.ppat.1009675] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/18/2021] [Accepted: 10/05/2021] [Indexed: 01/04/2023] Open
Abstract
Until recently, the study of major histocompability complex (MHC) mediated immunity has focused on the direct link between MHC diversity and susceptibility to parasite infection. However, MHC genes can also influence host health indirectly through the sculpting of the bacterial community that in turn shape immune responses. We investigated the links between MHC class I and II gene diversity gut microbiome diversity and micro- (adenovirus, AdV) and macro- (helminth) parasite infection probabilities in a wild population of non-human primates, mouse lemurs of Madagascar. This setup encompasses a plethora of underlying interactions between parasites, microbes and adaptive immunity in natural populations. Both MHC classes explained shifts in microbiome composition and the effect was driven by a few select microbial taxa. Among them were three taxa (Odoribacter, Campylobacter and Prevotellaceae-UCG-001) which were in turn linked to AdV and helminth infection status, correlative evidence of the indirect effect of the MHC via the microbiome. Our study provides support for the coupled role of MHC diversity and microbial flora as contributing factors of parasite infection. The selective pressure of the major histocompatibility complex (MHC) on microbial communities, and the potential role of this interaction in driving parasite resistance has been largely neglected. Using a natural population of the primate Microcebus griseorufus, we provide correlative evidence of two outstanding findings: that MHCI and MHCII diversity shapes the composition of the gut microbiota; and that select taxa associated with MHC diversity predicted adenovirus and helminth infection status. Our study highlights the importance of incorporating the microbiome when investigating parasite-mediated MHC selection.
Collapse
|
16
|
Microbiota from Specific Pathogen-Free Mice Reduces Campylobacter jejuni Chicken Colonization. Pathogens 2021; 10:pathogens10111387. [PMID: 34832543 PMCID: PMC8621964 DOI: 10.3390/pathogens10111387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Campylobacter jejuni, a prevalent foodborne bacterial pathogen, is mainly transmitted from poultry with few effective prevention approaches. In this study, we aimed to investigate the role of microbiota on C. jejuni chicken colonization. Microbiota from specific pathogen-free (SPF) mouse stools were collected as SPF-Aerobe and SPF-Anaerobe. Birds were colonized with SPF-Aerobe or SPF-Anaerobe at day 0 and infected with C. jejuni AR101 at day 12. Notably, C. jejuni AR101 colonized at 5.3 and 5.6 log10 C. jejuni CFU/g chicken cecal digesta at days 21 and 28, respectively, while both SPF-Aerobe and SPF-Anaerobe microbiota reduced pathogen colonization. Notably, SPF-Aerobe and SPF-Anaerobe increased cecal phylum Bacteroidetes and reduced phylum Firmicutes compared to those in the nontransplanted birds. Interestingly, microbiota from noninfected chickens, SPF-Aerobe, or SPF-Anaerobe inhibited AR101 in vitro growth, whereas microbiota from infected birds alone failed to reduce pathogen growth. The bacterium Enterobacter102 isolated from infected birds transplanted with SPF-Aerobe inhibited AR101 in vitro growth and reduced pathogen gut colonization in chickens. Together, SPF mouse microbiota was able to colonize chicken gut and reduce C. jejuni chicken colonization. The findings may help the development of effective strategies to reduce C. jejuni chicken contamination and campylobacteriosis.
Collapse
|
17
|
Ruiz MJ, Soto LP, Sirini NE, Werning ML, Olivero CR, Zimmermann JA, Zbrun MV, Acosta FF, Signorini ML, Frizzo LS. Murine colonization model by Campylobacter coli DSPV458. J Appl Microbiol 2021; 132:1457-1466. [PMID: 34465011 DOI: 10.1111/jam.15272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/11/2021] [Accepted: 08/24/2021] [Indexed: 11/29/2022]
Abstract
AIMS To generate a murine experimental model of colonization by Campylobacter coli DSPV458. METHODS AND RESULTS Twelve adult Balb/cCmedc female mice were housed in a treated group (T-G) and a control group (C-G) for 4 weeks. Both experimental groups received antibiotics for 5 days during the first week. The T-G was administered with 6.68log10 CFU of C. coli DSPV458 by oesophageal gavage. Necropsies were performed weekly to evaluate translocation and intestinal colonization in the spleen and liver and in the ileum and cecum respectively. Samples were cultured to quantify intestinal microbiota members. Faeces were cultured weekly for a C. coli DSPV458 count. Campylobacter coli DSPV458 was isolated from all the inoculated mice. The recovered level of C. coli DSPV458 was, on average, 6.9 log10 CFUg-1 , 8.0 log10 CFUg-1 and 1.6 log10 CFUg-1 in faeces, cecum and ileum respectively. Colonization by C. coli DSPV458 does not alter the normal clinical and physiological status. CONCLUSIONS Campylobacter coli DSPV458 does not have an invasive capacity, and the model is suitable for evaluating strategies to reduce intestinal loads. SIGNIFICANCE AND IMPACT OF STUDY Farm animals have an important impact on thermotolerant Campylobacter transmission to humans. Extremely few colonization models by C. coli have been reported to date. In food-producing animals, infection is mild or absent and thermotolerant Campylobacter colonize the intestines of animals. Colonization models are specific models that do not cause infection as they do not generally result in diarrhoea or other signs of disease. Therefore, this model will allow to evaluate the evolution of colonization by thermotolerant Campylobacter and the alternative tools development to antibiotics that limit their colonization in food-producing animals.
Collapse
Affiliation(s)
- M J Ruiz
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Animal Health and Preventive Medicine, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires, Argentina
| | - L P Soto
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina
| | - N E Sirini
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - M L Werning
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - C R Olivero
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - J A Zimmermann
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina
| | - M V Zbrun
- Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina.,National Council of Scientific and Technical Research, National Institute of Agricultural Technology EEA Rafaela, Rafaela, Province of Santa Fe, Argentina
| | - F F Acosta
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - M L Signorini
- Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina.,National Council of Scientific and Technical Research, National Institute of Agricultural Technology EEA Rafaela, Rafaela, Province of Santa Fe, Argentina
| | - L S Frizzo
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina
| |
Collapse
|
18
|
Gorain C, Khan A, Singh A, Mondal S, Mallick AI. Bioengineering of LAB vector expressing Haemolysin co-regulated protein (Hcp): a strategic approach to control gut colonization of Campylobacter jejuni in a murine model. Gut Pathog 2021; 13:48. [PMID: 34330327 PMCID: PMC8323230 DOI: 10.1186/s13099-021-00444-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/20/2021] [Indexed: 01/02/2023] Open
Abstract
Background Campylobacter jejuni (C. jejuni) is accountable for more than 400 million cases of gastroenteritis each year and is listed as a high-priority gut pathogen by the World Health Organization (WHO). Although the acute infection of C. jejuni (campylobacteriosis) is commonly treated with macrolides and fluoroquinolones, the emergence of antibiotic resistance among C. jejuni warrants the need for an alternative approach to control campylobacteriosis in humans. To this end, vaccines remain a safe, effective, and widely accepted strategy for controlling emerging and re-emerging infectious diseases. In search of a suitable vaccine against campylobacteriosis, recently, we demonstrated the potential of recombinant Haemolysin co-regulated protein (Hcp) of C. jejuni Type VI secretion system (T6SS) in imparting significant immune-protection against cecal colonization of C. jejuni; however, in the avian model. Since clinical features of human campylobacteriosis are more complicated than the avians, we explored the potential of Hcp as a T6SS targeted vaccine in a murine model as a more reliable and reproducible experimental host to study vaccine-induced immune-protection against C. jejuni. Because C. jejuni primarily utilizes the mucosal route for host pathogenesis, we analyzed the immunogenicity of a mucosally deliverable bioengineered Lactic acid bacteria (LAB), Lactococcus lactis (L. lactis), expressing Hcp. Considering the role of Hcp in both structural (membrane-bound) and functional (effector protein) exhibition of C. jejuni T6SS, a head-to-head comparison of two different forms of recombinant LAB vectors (cell wall anchored and secreted form of Hcp) were tested and assessed for the immune phenotypes of each modality in BALB/c mice. Results We show that regardless of the Hcp protein localization, mucosal delivery of bioengineered LAB vector expressing Hcp induced high-level production of antigen-specific neutralizing antibody (sIgA) in the gut with the potential to reduce the cecal load of C. jejuni in mice. Conclusion Together with the non-commensal nature of L. lactis, short gut transit time in humans, and the ability to express the heterologous protein in the gut, the present study highlights the benefits of bioengineered LAB vectors based mucosal vaccine modality against C. jejuni without the risk of immunotolerance. Supplementary Information The online version contains supplementary material available at 10.1186/s13099-021-00444-2.
Collapse
Affiliation(s)
- Chandan Gorain
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Afruja Khan
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Ankita Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Samiran Mondal
- Department of Veterinary Pathology, West Bengal University of Animal and Fishery Sciences, Belgachia, Kolkata, West Bengal, 700037, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India.
| |
Collapse
|
19
|
Wymore Brand M, Sahin O, Hostetter JM, Trachsel J, Zhang Q, Wannemuehler MJ. Campylobacter jejuni persistently colonizes gnotobiotic altered Schaedler flora C3H/HeN mice and induces mild colitis. FEMS Microbiol Lett 2021; 367:5937419. [PMID: 33098301 DOI: 10.1093/femsle/fnaa163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/05/2020] [Indexed: 11/13/2022] Open
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.
Collapse
Affiliation(s)
- Meghan Wymore Brand
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Orhan Sahin
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Jesse M Hostetter
- Department of Veterinary Pathology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Drive, Athens, GA 30602, USA
| | - Julian Trachsel
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Qijing Zhang
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| |
Collapse
|
20
|
The Host Cellular Immune Response to Infection by Campylobacter Spp. and Its Role in Disease. Infect Immun 2021; 89:e0011621. [PMID: 34031129 DOI: 10.1128/iai.00116-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
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.
Collapse
|
21
|
Ruiz MJ, Zbrun MV, Signorini ML, Zimmermann JA, Soto LP, Rosmini MR, Frizzo LS. In vitro screening and in vivo colonization pilot model of Lactobacillus plantarum LP5 and Campylobacter coli DSPV 458 in mice. Arch Microbiol 2021; 203:4161-4171. [PMID: 34061232 DOI: 10.1007/s00203-021-02385-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/23/2021] [Accepted: 05/18/2021] [Indexed: 11/29/2022]
Abstract
The objective of this work was to determine the antibacterial effect of Lactobacillus plantarum strains of pork origin against Campylobacter coli strains, and to conduct experimental colonization pilot models in mice for both microorganisms. Inhibition assays allowed evaluation and selection of L. plantarum LP5 as the strain with the highest antagonistic activity against C. coli and with the best potential to be used in in vivo study. Adult 6-week-old female Balb/cCmedc mice were lodged in two groups. The treated group was administered with 9.4 log10CFU/2 times/wk of L. plantarum LP5. L. plantarum LP5 was recovered from the feces and cecum of the inoculated mice. However, when bacteria stopped being administered, probiotic counts decreased. Experimental colonization with C. coli was carried out in five groups of mice. All animals were treated with antibiotics in their drinking water to weaken the indigenous microbiota and to allow colonization of C. coli. Four groups were administered once with different C. coli strains (DSPV458: 8.49 log10CFU; DSPV567: 8.09 log10CFU; DSPV570: 8.46 log10CFU; DSPV541: 8.86 log10CFU, respectively). After 8 h, mice inoculated with different C. coli strains were colonized because the pathogen was detected in their feces. L. plantarum LP5 tolerated the gastrointestinal conditions of murine model without generating adverse effects on the animals. C. coli DSPV458 colonized the mice without causing infection by lodging in their digestive tract, thus generating a reproducible colonization model. Both models combined could be used as protection murine models against pathogens to test alternative control tools to antibiotics.
Collapse
Affiliation(s)
- M J Ruiz
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Animal Health and Preventive Medicine, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires (UNCPBA), Tandil, Province of Buenos Aires, Argentina
| | - M V Zbrun
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University (DSPV-FCV-UNL), Esperanza, Province of Santa Fe, Argentina
| | - M L Signorini
- Department of Public Health, Faculty of Veterinary Science, Litoral National University (DSPV-FCV-UNL), Esperanza, Province of Santa Fe, Argentina.,National Council of Scientific and Technical Research, National Institute of Agricultural Technology EEA Rafaela (CONICET/INTA), Rafaela, Province of Santa Fe, Argentina
| | - J A Zimmermann
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University (DSPV-FCV-UNL), Esperanza, Province of Santa Fe, Argentina
| | - L P Soto
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University (DSPV-FCV-UNL), Esperanza, Province of Santa Fe, Argentina
| | - M R Rosmini
- Department of Public Health, Faculty of Veterinary Science, Litoral National University (DSPV-FCV-UNL), Esperanza, Province of Santa Fe, Argentina
| | - L S Frizzo
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina. .,Department of Public Health, Faculty of Veterinary Science, Litoral National University (DSPV-FCV-UNL), Esperanza, Province of Santa Fe, Argentina.
| |
Collapse
|
22
|
Abstract
Campylobacter jejuni and Campylobacter coli can be frequently isolated from poultry and poultry-derived products, and in combination these two species cause a large portion of human bacterial gastroenteritis cases. While birds are typically colonized by these Campylobacter species without clinical symptoms, in humans they cause (foodborne) infections at high frequencies, estimated to cost billions of dollars worldwide every year. The clinical outcome of Campylobacter infections comprises malaise, diarrhea, abdominal pain and fever. Symptoms may continue for up to two weeks and are generally self-limiting, though occasionally the disease can be more severe or result in post-infection sequelae. The virulence properties of these pathogens have been best-characterized for C. jejuni, and their actions are reviewed here. Various virulence-associated bacterial determinants include the flagellum, numerous flagellar secreted factors, protein adhesins, cytolethal distending toxin (CDT), lipooligosaccharide (LOS), serine protease HtrA and others. These factors are involved in several pathogenicity-linked properties that can be divided into bacterial chemotaxis, motility, attachment, invasion, survival, cellular transmigration and spread to deeper tissue. All of these steps require intimate interactions between bacteria and host cells (including immune cells), enabled by the collection of bacterial and host factors that have already been identified. The assortment of pathogenicity-associated factors now recognized for C. jejuni, their function and the proposed host cell factors that are involved in crucial steps leading to disease are discussed in detail.
Collapse
|
23
|
Mousavi S, Bereswill S, Heimesaat MM. Murine Models for the Investigation of Colonization Resistance and Innate Immune Responses in Campylobacter Jejuni Infections. Curr Top Microbiol Immunol 2021; 431:233-263. [PMID: 33620654 DOI: 10.1007/978-3-030-65481-8_9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Human infections with the food-borne pathogen Campylobacter jejuni are progressively increasing worldwide and constitute a significant socioeconomic burden to mankind. Intestinal campylobacteriosis in humans is characterized by bloody diarrhea, fever, abdominal pain, and severe malaise. Some individuals develop chronic post-infectious sequelae including neurological and autoimmune diseases such as reactive arthritis and Guillain-Barré syndrome. Studies unraveling the molecular mechanisms underlying campylobacteriosis and post-infectious sequelae have been hampered by the scarcity of appropriate experimental in vivo models. Particularly, conventional laboratory mice are protected from C. jejuni infection due to the physiological colonization resistance exerted by the murine gut microbiota composition. Additionally, as compared to humans, mice are up to 10,000 times more resistant to C. jejuni lipooligosaccharide (LOS) constituting a major pathogenicity factor responsible for the immunopathological host responses during campylobacteriosis. In this chapter, we summarize the recent progress that has been made in overcoming these fundamental obstacles in Campylobacter research in mice. Modification of the murine host-specific gut microbiota composition and sensitization of the mice to C. jejuni LOS by deletion of genes encoding interleukin-10 or a single IL-1 receptor-related molecule as well as by dietary zinc depletion have yielded reliable murine infection models resembling key features of human campylobacteriosis. These substantial improvements pave the way for a better understanding of the molecular mechanisms underlying pathogen-host interactions. The ongoing validation and standardization of these novel murine infection models will provide the basis for the development of innovative treatment and prevention strategies to combat human campylobacteriosis and collateral damages of C. jejuni infections.
Collapse
Affiliation(s)
- Soraya Mousavi
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University of Berlin, Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University of Berlin, Berlin Institute of Health, Berlin, Germany
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University of Berlin, Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
24
|
Negretti NM, Ye Y, Malavasi LM, Pokharel SM, Huynh S, Noh S, Klima CL, Gourley CR, Ragle CA, Bose S, Looft T, Parker CT, Clair G, Adkins JN, Konkel ME. A porcine ligated loop model reveals new insight into the host immune response against Campylobacter jejuni. Gut Microbes 2020; 12:1-25. [PMID: 32887530 PMCID: PMC7524355 DOI: 10.1080/19490976.2020.1814121] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 02/03/2023] Open
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.
Collapse
Affiliation(s)
- Nicholas M Negretti
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Yinyin Ye
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lais M Malavasi
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Swechha M Pokharel
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Steven Huynh
- Produce Safety and Microbiology, United States Department of Agriculture-Agricultural Research Service, Albany, CA, USA
| | - Susan Noh
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, USA
- Washington Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Cassidy L Klima
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, USA
| | - Christopher R Gourley
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Claude A Ragle
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Torey Looft
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, USA
| | - Craig T Parker
- Produce Safety and Microbiology, United States Department of Agriculture-Agricultural Research Service, Albany, CA, USA
| | - Geremy Clair
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joshua N Adkins
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Michael E Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
25
|
Perruzza L, Jaconi S, Lombardo G, Pinna D, Strati F, Morone D, Seehusen F, Hu Y, Bajoria S, Xiong J, Kumru OS, Joshi SB, Volkin DB, Piantanida R, Benigni F, Grassi F, Corti D, Pizzuto MS. Prophylactic Activity of Orally Administered FliD-Reactive Monoclonal SIgA Against Campylobacter Infection. Front Immunol 2020; 11:1011. [PMID: 32582158 PMCID: PMC7296071 DOI: 10.3389/fimmu.2020.01011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
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.
Collapse
Affiliation(s)
- Lisa Perruzza
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Stefano Jaconi
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Gloria Lombardo
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Debora Pinna
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Francesco Strati
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Diego Morone
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Frauke Seehusen
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Yue Hu
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Jian Xiong
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Ozan Selahattin Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Sangeeta Bagai Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - David Bernard Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Renato Piantanida
- Department of Otolaryngology-Head and Neck Surgery, Ospedale Regionale di Lugano, Lugano, Switzerland
| | - Fabio Benigni
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Fabio Grassi
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Davide Corti
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | | |
Collapse
|
26
|
Vendrik KEW, Ooijevaar RE, de Jong PRC, Laman JD, van Oosten BW, van Hilten JJ, Ducarmon QR, Keller JJ, Kuijper EJ, Contarino MF. Fecal Microbiota Transplantation in Neurological Disorders. Front Cell Infect Microbiol 2020; 10:98. [PMID: 32266160 PMCID: PMC7105733 DOI: 10.3389/fcimb.2020.00098] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Several studies suggested an important role of the gut microbiota in the pathophysiology of neurological disorders, implying that alteration of the gut microbiota might serve as a treatment strategy. Fecal microbiota transplantation (FMT) is currently the most effective gut microbiota intervention and an accepted treatment for recurrent Clostridioides difficile infections. To evaluate indications of FMT for patients with neurological disorders, we summarized the available literature on FMT. In addition, we provide suggestions for future directions. Methods: In July 2019, five main databases were searched for studies and case descriptions on FMT in neurological disorders in humans or animal models. In addition, the ClinicalTrials.gov website was consulted for registered planned and ongoing trials. Results: Of 541 identified studies, 34 were included in the analysis. Clinical trials with FMT have been performed in patients with autism spectrum disorder and showed beneficial effects on neurological symptoms. For multiple sclerosis and Parkinson's disease, several animal studies suggested a positive effect of FMT, supported by some human case reports. For epilepsy, Tourette syndrome, and diabetic neuropathy some studies suggested a beneficial effect of FMT, but evidence was restricted to case reports and limited numbers of animal studies. For stroke, Alzheimer's disease and Guillain-Barré syndrome only studies with animal models were identified. These studies suggested a potential beneficial effect of healthy donor FMT. In contrast, one study with an animal model for stroke showed increased mortality after FMT. For Guillain-Barré only one study was identified. Whether positive findings from animal studies can be confirmed in the treatment of human diseases awaits to be seen. Several trials with FMT as treatment for the above mentioned neurological disorders are planned or ongoing, as well as for amyotrophic lateral sclerosis. Conclusions: Preliminary literature suggests that FMT may be a promising treatment option for several neurological disorders. However, available evidence is still scanty and some contrasting results were observed. A limited number of studies in humans have been performed or are ongoing, while for some disorders only animal experiments have been conducted. Large double-blinded randomized controlled trials are needed to further elucidate the effect of FMT in neurological disorders.
Collapse
Affiliation(s)
- Karuna E W Vendrik
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Centre for Infectious Disease Control, National Institute for Public Health and the Environment (Rijksinstituut voor Volksgezondheid en Milieu, RIVM), Bilthoven, Netherlands
| | - Rogier E Ooijevaar
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, Netherlands
| | - Pieter R C de Jong
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Jon D Laman
- Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, Groningen, Netherlands
| | - Bob W van Oosten
- Department of Neurology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, Netherlands
| | | | - Quinten R Ducarmon
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
| | - Josbert J Keller
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology, Haaglanden Medical Center, The Hague, Netherlands
| | - Eduard J Kuijper
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Centre for Infectious Disease Control, National Institute for Public Health and the Environment (Rijksinstituut voor Volksgezondheid en Milieu, RIVM), Bilthoven, Netherlands.,Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Fiorella Contarino
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands.,Department of Neurology, Haga Teaching Hospital, The Hague, Netherlands
| |
Collapse
|
27
|
Igwaran A, Okoh AI. Human campylobacteriosis: A public health concern of global importance. Heliyon 2019; 5:e02814. [PMID: 31763476 PMCID: PMC6861584 DOI: 10.1016/j.heliyon.2019.e02814] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 06/07/2019] [Accepted: 11/07/2019] [Indexed: 01/18/2023] Open
Abstract
Campylobacter species are among the leading cause of bacterial foodborne and waterborne infections. In addition, Campylobacter is one of the major causative agent of bacterial gastrointestinal infections and the rise in the incidence of Campylobacter infections have been reported worldwide. Also, the emergence of some Campylobacter species as one of the main causative agent of diarrhea and the propensity of these bacteria species to resist the actions of antimicrobial agents; position them as a serious threat to the public health. This paper reviews Campylobacter pathogenicity, infections, isolation and diagnosis, their reservoirs, transmission pathways, epidemiology of Campylobacter outbreaks, prevention and treatment option, antibiotics resistance and control of antibiotics use.
Collapse
Affiliation(s)
- Aboi Igwaran
- SAMRC Microbial Water Quality Monitoring Centre, University of Fort Hare, Alice, 5700, South Africa
- Applied and Environmental Microbiology Research Group (AEMREG), Department of Biochemistry and Microbiology, University of Fort Hare, Private Bag X1314, Alice, 5700, Eastern Cape, South Africa
| | - Anthony Ifeanyi Okoh
- SAMRC Microbial Water Quality Monitoring Centre, University of Fort Hare, Alice, 5700, South Africa
- Applied and Environmental Microbiology Research Group (AEMREG), Department of Biochemistry and Microbiology, University of Fort Hare, Private Bag X1314, Alice, 5700, Eastern Cape, South Africa
| |
Collapse
|
28
|
Brooks PT, Bell JA, Bejcek CE, Malik A, Mansfield LS. An antibiotic depleted microbiome drives severe Campylobacter jejuni-mediated Type 1/17 colitis, Type 2 autoimmunity and neurologic sequelae in a mouse model. J Neuroimmunol 2019; 337:577048. [PMID: 31678855 DOI: 10.1016/j.jneuroim.2019.577048] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/01/2019] [Accepted: 09/02/2019] [Indexed: 10/26/2022]
Abstract
The peripheral neuropathy Guillain-Barré Syndrome can follow Campylobacter jejuni infection when outer core lipooligosaccharides induce production of neurotoxic anti-ganglioside antibodies. We hypothesized that gut microbiota depletion with an antibiotic would increase C. jejuni colonization, severity of gastroenteritis, and GBS. Microbiota depletion increased C. jejuni colonization, invasion, and colitis with Type 1/17 T cells in gut lamina propria. It also stimulated Type 1/17 anti-C. jejuni and -antiganglioside-antibodies, Type 2 anti-C. jejuni and -antiganglioside antibodies, and neurologic phenotypes. Results indicate that both C. jejuni strain and gut microbiota affect development of inflammation and GBS and suggest that probiotics following C. jejuni infection may ameliorate inflammation and autoimmune disease.
Collapse
Affiliation(s)
- Phillip T Brooks
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Comparative Medicine Integrative Biology Graduate Program, Michigan State University, East Lansing, MI, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Julia A Bell
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Christopher E Bejcek
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Ankit Malik
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Linda S Mansfield
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
29
|
Effects of antibiotic resistance (AR) and microbiota shifts on Campylobacter jejuni-mediated diseases. Anim Health Res Rev 2019; 18:99-111. [PMID: 29665882 DOI: 10.1017/s1466252318000014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Campylobacter jejuni is an important zoonotic pathogen recently designated a serious antimicrobial resistant (AR) threat. While most patients with C. jejuni experience hemorrhagic colitis, serious autoimmune conditions can follow including inflammatory bowel disease (IBD) and the acute neuropathy Guillain Barré Syndrome (GBS). This review examines inter-relationships among factors mediating C. jejuni diarrheal versus autoimmune disease especially AR C. jejuni and microbiome shifts. Because both susceptible and AR C. jejuni are acquired from animals or their products, we consider their role in harboring strains. Inter-relationships among factors mediating C. jejuni colonization, diarrheal and autoimmune disease include C. jejuni virulence factors and AR, the enteric microbiome, and host responses. Because AR C. jejuni have been suggested to affect the severity of disease, length of infections and propensity to develop GBS, it is important to understand how these interactions occur when strains are under selection by antimicrobials. More work is needed to elucidate host-pathogen interactions of AR C. jejuni compared with susceptible strains and how AR C. jejuni are maintained and evolve in animal reservoirs and the extent of transmission to humans. These knowledge gaps impair the development of effective strategies to prevent the emergence of AR C. jejuni in reservoir species and human populations.
Collapse
|
30
|
Ungureanu VA, Stratakos AC, Gundogdu O, Stef L, Pet I, Pet E, Pacala N, Corcionivoschi N. Virulence of a T6SS Campylobacter jejuni chicken isolate from North Romania. BMC Res Notes 2019; 12:180. [PMID: 30922352 PMCID: PMC6437841 DOI: 10.1186/s13104-019-4201-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/16/2019] [Indexed: 11/25/2022] Open
Abstract
Objectives In this study we have investigated the in vitro and in vivo virulence characteristics of a new T6SS positive Campylobacter jejuni chicken isolate (SV12) originating from a poultry population in North Romania. A detailed phenotypic characterization was performed and compared to the T6SS negative C. jejuni 81–176 wild strain. Results Our results indicate that the significantly higher capacity to attach and invade HCT-8 cells of C. jejuni SV12 isolate is associated with increased motility, increased resistance to bile salts and serum resistance, when compared to C. jejuni strain 81–76. Mice infected with the SV12 isolate showed statistically higher levels of colonization at both 7- and 14-days post-inoculation and in the stomach, caecum, duodenum and large intestine. Infection with the SV12 strain induced a stronger immune response as the gene transcript levels of IL-17, TNFα and IFNγ were more pronouncedly up-regulated compared to the C. jejuni strain 81–176. The present study showed that the new isolate SV12 had an enhanced virulence capacity compared to the wild strain which was evident in vivo as well. This work also provides an insight on the colonization pattern and host immune response differences between T6SS positive and T6SS negative C. jejuni.
Collapse
Affiliation(s)
- Vlad A Ungureanu
- School of Bioengineering and Animal Resources, Banat University of Animal Sciences and Veterinary Medicine, King Michael I of Romania, Timisoara, Romania
| | - Alexandros Ch Stratakos
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Newforge Lane, Belfast, BT9 5GB, Northern Ireland, UK.,Auranta, NovaUCD, Dublin, Ireland
| | - Ozan Gundogdu
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, 11 Keppel Street, London, WC1E 7HT, UK
| | - Lavinia Stef
- School of Bioengineering and Animal Resources, Banat University of Animal Sciences and Veterinary Medicine, King Michael I of Romania, Timisoara, Romania
| | - Ioan Pet
- School of Bioengineering and Animal Resources, Banat University of Animal Sciences and Veterinary Medicine, King Michael I of Romania, Timisoara, Romania
| | - Elena Pet
- School Management and Rural Tourism, Banat University of Animal Sciences and Veterinary Medicine, King Michael I of Romania, Timisoara, Romania
| | - Nicolae Pacala
- School of Bioengineering and Animal Resources, Banat University of Animal Sciences and Veterinary Medicine, King Michael I of Romania, Timisoara, Romania
| | - Nicolae Corcionivoschi
- School of Bioengineering and Animal Resources, Banat University of Animal Sciences and Veterinary Medicine, King Michael I of Romania, Timisoara, Romania. .,Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Newforge Lane, Belfast, BT9 5GB, Northern Ireland, UK.
| |
Collapse
|
31
|
In vivo fluid accumulation-inhibitory, anticolonization and anti-inflammatory and in vitro biofilm-inhibitory activities of methyl gallate isolated from Terminalia chebula against fluoroquinolones resistant Vibrio cholerae. Microb Pathog 2019; 128:41-46. [DOI: 10.1016/j.micpath.2018.12.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 12/02/2017] [Accepted: 12/18/2018] [Indexed: 11/21/2022]
|
32
|
Shariati A, Fallah F, Pormohammad A, Taghipour A, Safari H, Chirani AS, Sabour S, Alizadeh-Sani M, Azimi T. The possible role of bacteria, viruses, and parasites in initiation and exacerbation of irritable bowel syndrome. J Cell Physiol 2018; 234:8550-8569. [PMID: 30480810 DOI: 10.1002/jcp.27828] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022]
Abstract
Irritable bowel syndrome (IBS) is a prolonged and disabling functional gastrointestinal disorder with the incidence rate of 18% in the world. IBS could seriously affect lifetime of patients and cause high economic burden on the community. The pathophysiology of the IBS is hardly understood, whereas several possible mechanisms, such as visceral hypersensitivity, irregular gut motility, abnormal brain-gut relations, and the role of infectious agents, are implicated in initiation and development of this syndrome. Different studies demonstrated an alteration in B-lymphocytes, mast cells (MC), T-lymphocytes, and cytokine concentrations in intestinal mucosa or systemic circulation that are likely to contribute to the formation of the IBS. Therefore, IBS could be developed in those with genetic predisposition. Infections' role in initiation and exacerbation of IBS has been investigated by quite several clinical studies; moreover, the possible role of some pathogens in development and exacerbation of this disease has been described. It appears that the main obligatory pathogens correspond with the IBS disease, Clostridium difficile, Escherichia coli, Mycobacterium avium subspecies paratuberculosis, Campylobacter concisus, Campylobacter jejuni, Chlamydia trachomatis, Helicobacter pylori, Pseudomonas aeruginosa, Salmonella spp, Shigella spp, and viruses, particularly noroviruses. A number of pathogenic parasites (Blastocystis, Dientamoeba fragilis, and Giardia lamblia) may also be involved in the progression and exacerbation of the disease. Based on the current knowledge, the current study concludes that the most common bacterial, viral, and parasitic pathogens may be involved in the development and progression of IBS.
Collapse
Affiliation(s)
- Aref Shariati
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Fallah
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Pormohammad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Taghipour
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Safari
- Health Promotion Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Salami Chirani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Sabour
- Department of Microbiology, School of Medicine, Ardebil University of Medical Science, Ardebil, Iran
| | - Mahmood Alizadeh-Sani
- Student Research Committee, Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Azimi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Korolik V. The role of chemotaxis during Campylobacter jejuni colonisation and pathogenesis. Curr Opin Microbiol 2018; 47:32-37. [PMID: 30476739 DOI: 10.1016/j.mib.2018.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 01/25/2023]
Abstract
Campylobacter jejuni is a ubiquitous gastrointestinal pathogen, transmitted to humans from birds and animals, where C. jejuni is part of normal intestinal flora. In C. jejuni, similar to other motile bacteria, chemotaxis pathway and the array of chemosensors sense and respond to external stimuli with unique precision and sensitivity and are considered to be critical for bacterial colonisation and pathogenicity. Disruption of any component of the signal transduction pathway consisting of receptor-CheA/CheW-CheY-flagella cascade, the signal adaptation system, and even a loss of a single chemosensory receptor, dramatically reduce the ability of C. jejuni to colonise various animal hosts and to cause disease.
Collapse
Affiliation(s)
- Victoria Korolik
- Institute for Glycomics, Griffith University, Gold Coast, 4222, QLD, Australia.
| |
Collapse
|
34
|
Selective colonization ability of human fecal microbes in different mouse gut environments. ISME JOURNAL 2018; 13:805-823. [PMID: 30442907 DOI: 10.1038/s41396-018-0312-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/10/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022]
Abstract
Mammalian hosts constantly interact with diverse exogenous microbes, but only a subset of the microbes manage to colonize due to selective colonization resistance exerted by host genetic factors as well as the native microbiota of the host. An important question in microbial ecology and medical science is if such colonization resistance can discriminate closely related microbial species, or even closely related strains of the same species. Using human-mouse fecal microbiota transplantation and metagenomic shotgun sequencing, we reconstructed colonization patterns of human fecal microbes in mice with different genotypes (C57BL6/J vs. NSG) and with or without an intact gut microbiota. We found that mouse genotypes and the native mouse gut microbiota both exerted different selective pressures on exogenous colonizers: human fecal Bacteroides successfully established in the mice gut, however, different species of Bacteroides selectively enriched under different gut conditions, potentially due to a multitude of functional differences, ranging from versatility in nutrient acquisition to stress responses. Additionally, different clades of Bacteroides cellulosilyticus strains were selectively enriched in different gut conditions, suggesting that the fitness of conspecific microbial strains in a novel host environment could differ.
Collapse
|
35
|
Matilla MA, Krell T. The effect of bacterial chemotaxis on host infection and pathogenicity. FEMS Microbiol Rev 2018; 42:4563582. [PMID: 29069367 DOI: 10.1093/femsre/fux052] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022] Open
Abstract
Chemotaxis enables microorganisms to move according to chemical gradients. Although this process requires substantial cellular energy, it also affords key physiological benefits, including enhanced access to growth substrates. Another important implication of chemotaxis is that it also plays an important role in infection and disease, as chemotaxis signalling pathways are broadly distributed across a variety of pathogenic bacteria. Furthermore, current research indicates that chemotaxis is essential for the initial stages of infection in different human, animal and plant pathogens. This review focuses on recent findings that have identified specific bacterial chemoreceptors and corresponding chemoeffectors associated with pathogenicity. Pathogenicity-related chemoeffectors are either host and niche-specific signals or intermediates of the host general metabolism. Plant pathogens were found to contain an elevated number of chemotaxis signalling genes and functional studies demonstrate that these genes are critical for their ability to enter the host. The expanding body of knowledge of the mechanisms underlying chemotaxis in pathogens provides a foundation for the development of new therapeutic strategies capable of blocking infection and preventing disease by interfering with chemotactic signalling pathways.
Collapse
Affiliation(s)
- Miguel A Matilla
- Department of Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain
| | - Tino Krell
- Department of Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain
| |
Collapse
|
36
|
Ren F, Li X, Tang H, Jiang Q, Yun X, Fang L, Huang P, Tang Y, Li Q, Huang J, Jiao XA. Insights into the impact of flhF inactivation on Campylobacter jejuni colonization of chick and mice gut. BMC Microbiol 2018; 18:149. [PMID: 30348090 PMCID: PMC6196472 DOI: 10.1186/s12866-018-1318-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/15/2018] [Indexed: 11/23/2022] Open
Abstract
Background Campylobacter jejuni (C. jejuni) is a leading cause of foodborne gastroenteritis worldwide. This bacterium lacks many of the classical virulence factors, and flagellum-associated persistent colonization has been shown to be crucial for its pathogenesis. The flagellum plays a multifunctional role in C. jejuni pathogenesis, and different flagellar elements make diverse contributions. The flhF gene encodes the flagellar biosynthesis regulator, which is important for flagellar biosynthesis. In this study, the influence of flhF on C. jejuni colonization was systematically studied, and the possible mechanisms were also analyzed. Results The flhF gene has a significant influence on C. jejuni colonization, and its inactivation resulted in severe defects in the commensal colonization of chicks, with approximately 104- to 107-fold reductions (for NCTC 11168 and a C. jejuni isolate respectively) observed in the bacterial caecal loads. Similar effects were observed in mice where the flhF mutant strain completely lost the ability to continuously colonize mice, which cleared the isolate at 7 days post inoculation. Characterization of the phenotypic properties of C. jejuni that influence colonization showed that the adhesion and invasion abilities of the C. jejuni flhF mutant were reduced to approximately 52 and 27% of that of the wild-type strain, respectively. The autoagglutination and biofilm-formation abilities of the flhF mutant strain were also significantly decreased. Further genetic investigation revealed that flhF is continuously upregulated during the infection process, which indicates a close association of this gene with C. jejuni pathogenesis. The transcription of some other infection-related genes that are not directly involved in flagellar assembly were also influenced by its inactivation, with the flagellar coexpressed determinants (Feds) being apparently affected. Conclusions Inactivation of flhF has a significant influence on C. jejuni colonization in both birds and mammals. This defect may be caused by the decreased adhesion, invasion, autoagglutination and biofilm-formation abilities of the flhF mutant strain, as well as the influence on the transcription of other infection related genes, which provides insights into this virulence factor and the flagellum mediated co-regulation of C. jejuni pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12866-018-1318-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fangzhe Ren
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
| | - Xiaofei Li
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China
| | - Haiyan Tang
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China
| | - Qidong Jiang
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China
| | - Xi Yun
- Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Lin Fang
- Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Pingyu Huang
- Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Yuanyue Tang
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China.,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Qiuchun Li
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China.,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Jinlin Huang
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China. .,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China. .,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China.
| | - Xin-An Jiao
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China. .,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China. .,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
37
|
Elgamoudi BA, Ketley JM, Korolik V. New approach to distinguishing chemoattractants, chemorepellents and catabolised chemoeffectors for Campylobacter jejuni. J Microbiol Methods 2018; 146:83-91. [PMID: 29428740 DOI: 10.1016/j.mimet.2018.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/07/2018] [Accepted: 02/07/2018] [Indexed: 10/18/2022]
Abstract
Chemotactic behaviour is an important part of the lifestyle of motile bacteria and enables cells to respond to various environmental stimuli. The Hard Agar Plug (HAP) method is used to study the chemotactic behaviour of bacteria, including the fastidious microaerophile Campylobacter jejuni, an intestinal pathogen of humans. However, the traditional HAP assay is not quantitative, is unsuitable for chemotaxis observation over short time periods and for the investigation of repellent taxis, and is prone to false-positive and -negative results. Here we report an accurate, rapid, and quantitative HAP-based chemotaxis assay, tHAP, for the investigation of bacterial chemotactic responses. The critical component of the new assay is the addition of triphenyltetrazolium chloride (TTC). Enzymatic reduction of TTC to TFP-Red (1, 3, 5-Triphenylformazan) enables colourimetric detection of actively metabolising bacterial cells. Quantitative assessment of chemotaxis is achieved by colourimetric measurement or viability count over a period of 10 min to 3 h. Using the tHAP assay, we observed the dose-responsive chemotactic motility of C. jejuni cells along different concentrations of attractants aspartate and serine. Importantly, we have also designed a competitive tHAP assay to differentiate between repellents and attractants and to identify chemoeffectors that do not activate metabolism. IMPORTANCE The modified tHAP assay described here enables the exploration of the chemoresponse of Campylobacter jejuni towards chemorepellents, and catabolizable and non-catabolizable chemoattractants.
Collapse
Affiliation(s)
- Bassam A Elgamoudi
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Australia; Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Julian M Ketley
- Department of Genetics, University of Leicester, Leicester, United Kingdom.
| | - Victoria Korolik
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Australia.
| |
Collapse
|
38
|
Campylobacter jejuni and associated immune mechanisms: short-term effects and long-term implications for infants in low-income countries. Curr Opin Infect Dis 2018; 30:322-328. [PMID: 28157786 DOI: 10.1097/qco.0000000000000364] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Campylobacter jejuni is recognized as one of the most common causes of food-borne gastrointestinal illness worldwide, resulting in a self-limiting dysentery in developed countries. However, it is increasingly gaining attention due to its association with postinfectious complications such as Guillain-Barré Syndrome and recently recognized importance in early childhood diarrhea in developing countries. We hypothesize that the inflammation mediated by C. jejuni infection causes environmental enteric dysfunction, and with contribution from diet and the host, microbiome may be responsible for growth faltering in children and developmental disability. RECENT FINDINGS Diet plays a major role in the impact of C. jejuni infection, both by availability of micronutrients for the bacteria and host as well as shaping the microbiome that affords resistance. Early childhood repeated exposure to the bacterium results in inflammation that affords long-term immunity but, in the short term, can lead to malabsorption, oral vaccine failure, cognitive delay and increased under-5 mortality. SUMMARY As interest in C. jejuni increases, our understanding of its virulence mechanisms has improved. However, much work remains to be done to fully understand the implications of immune-mediated inflammation and its potential role in diseases such as environmental enteric dysfunction.
Collapse
|
39
|
Brooks PT, Brakel KA, Bell JA, Bejcek CE, Gilpin T, Brudvig JM, Mansfield LS. Transplanted human fecal microbiota enhanced Guillain Barré syndrome autoantibody responses after Campylobacter jejuni infection in C57BL/6 mice. MICROBIOME 2017; 5:92. [PMID: 28789710 PMCID: PMC5547673 DOI: 10.1186/s40168-017-0284-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/05/2017] [Indexed: 05/10/2023]
Abstract
BACKGROUND Campylobacter jejuni is the leading antecedent infection to the autoimmune neuropathy Guillain-Barré syndrome (GBS), which is accompanied by an autoimmune anti-ganglioside antibody attack on peripheral nerves. Previously, we showed that contrasting immune responses mediate C. jejuni induced colitis and autoimmunity in interleukin-10 (IL-10)-deficient mice, dependent upon the infecting strain. Strains from colitis patients elicited T helper 1 (TH1)-dependent inflammatory responses while strains from GBS patients elicited TH2-dependent autoantibody production. Both syndromes were exacerbated by antibiotic depletion of the microbiota, but other factors controlling susceptibility to GBS are unknown. METHODS Using 16S rRNA gene high-throughput sequencing, we examined whether structure of the gut microbial community alters host (1) gastrointestinal inflammation or (2) anti-ganglioside antibody responses after infection with C. jejuni strains from colitis or GBS patients. We compared these responses in C57BL/6 mice with either (1) stable human gut microbiota (Humicrobiota) transplants or (2) conventional mouse microbiota (Convmicrobiota). RESULTS Inoculating germ-free C57BL/6 wild-type (WT) mice with a mixed human fecal slurry provided a murine model that stably passed its microbiota over >20 generations. Mice were housed in specific pathogen-free (SPF) facilities, while extra precautions of having caretakers wear sterile garb along with limited access ensured that no mouse pathogens were acquired. Humicrobiota conferred many changes upon the WT model in contrast to previous results, which showed only colonization with no disease after C. jejuni challenge. When compared to Convmicrobiota mice for susceptibility to C. jejuni enteric or GBS patient strains, infected Humicrobiota mice had (1) 10-100 fold increases in C. jejuni colonization of both strains, (2) pathologic change in draining lymph nodes but only mild changes in colon or cecal lamina propria, (3) significantly lower Th1/Th17-dependent anti-C. jejuni responses, (4) significantly higher IL-4 responses at 5 but not 7 weeks post infection (PI), (5) significantly higher Th2-dependent anti-C. jejuni responses, and (6) significantly elevated anti-ganglioside autoantibodies after C. jejuni infection. These responses in Humicrobiota mice were correlated with a dominant Bacteroidetes and Firmicutes microbiota. CONCLUSIONS These data demonstrate that Humicrobiota altered host-pathogen interactions in infected mice, increasing colonization and Th-2 and autoimmune responses in a C. jejuni strain-dependent manner. Thus, microbiota composition is another factor controlling susceptibility to GBS.
Collapse
Affiliation(s)
- Phillip T Brooks
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Kelsey A Brakel
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Julia A Bell
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Christopher E Bejcek
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
| | - Trey Gilpin
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
| | - Jean M Brudvig
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Linda S Mansfield
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA.
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI, USA.
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA.
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA.
- Department of Microbiology and Molecular Genetics, Michigan State University, 181 Food Safety Building; 1129 Farm Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
40
|
King RM, Korolik V. Characterization of Ligand-Receptor Interactions: Chemotaxis, Biofilm, Cell Culture Assays, and Animal Model Methodologies. Methods Mol Biol 2017; 1512:149-161. [PMID: 27885605 DOI: 10.1007/978-1-4939-6536-6_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Chemotactic motility is an essential virulence factor for the pathogenesis of Campylobacter spp. infection. In Chapter 6 , we described technologies that enable initial screening and identification of ligands able to interact with chemoreceptor sensory domains. These include amino acid and glycan arrays, NMR, and SPR that are utilized to identify potential ligands interacting with Campylobacter jejuni. Here we describe techniques that enable the characterization and evaluation of ligand-receptor binding in chemotaxis through the assessment of motility and directed chemotactic motility as well as the associated phenotypes-autoagglutination behavior, biofilm formation, ability to adhere and invade cultured mammalian cells, and colonization ability in avian hosts.
Collapse
Affiliation(s)
- Rebecca M King
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Australia
| | - Victoria Korolik
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Australia.
| |
Collapse
|
41
|
Stahl M, Graef FA, Vallance BA. Mouse Models for Campylobacter jejuni Colonization and Infection. Methods Mol Biol 2017; 1512:171-188. [PMID: 27885607 DOI: 10.1007/978-1-4939-6536-6_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Relevant animal models for Campylobacter jejuni infection have been difficult to establish due to C. jejuni's inability to cause disease in many common animal research models. Fortunately, recent work has proven successful in developing several new and relevant mouse models of C. jejuni infection, including the SIGIRR-deficient mouse strain that develops acute enterocolitis in response to C. jejuni. Here we describe how to properly infect mice with C. jejuni, as well as a number of accompanying histological techniques to aid in studying C. jejuni colonization and infection in mice.
Collapse
Affiliation(s)
- Martin Stahl
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, V6H 3V4, Canada
| | - Franziska A Graef
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, V6H 3V4, Canada
| | - Bruce A Vallance
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, V6H 3V4, Canada.
| |
Collapse
|
42
|
Fiebiger U, Bereswill S, Heimesaat MM. Dissecting the Interplay Between Intestinal Microbiota and Host Immunity in Health and Disease: Lessons Learned from Germfree and Gnotobiotic Animal Models. Eur J Microbiol Immunol (Bp) 2016; 6:253-271. [PMID: 27980855 PMCID: PMC5146645 DOI: 10.1556/1886.2016.00036] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 02/06/2023] Open
Abstract
This review elaborates the development of germfree and gnotobiotic animal models and their application in the scientific field to unravel mechanisms underlying host-microbe interactions and distinct diseases. Strictly germfree animals are raised in isolators and not colonized by any organism at all. The germfree state is continuously maintained by birth, raising, housing and breeding under strict sterile conditions. However, isolator raised germfree mice are exposed to a stressful environment and exert an underdeveloped immune system. To circumvent these physiological disadvantages depletion of the bacterial microbiota in conventionally raised and housed mice by antibiotic treatment has become an alternative approach. While fungi and parasites are not affected by antibiosis, the bacterial microbiota in these "secondary abiotic mice" have been shown to be virtually eradicated. Recolonization of isolator raised germfree animals or secondary abiotic mice results in a gnotobiotic state. Both, germfree and gnotobiotic mice have been successfully used to investigate biological functions of the conventional microbiota in health and disease. Particularly for the development of novel clinical applications germfree mice are widely used tools, as summarized in this review further focusing on the modulation of bacterial microbiota in laboratory mice to better mimic conditions in the human host.
Collapse
Affiliation(s)
| | | | - Markus M. Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology and Hygiene, Charité – University Medicine Berlin, Campus Benjamin Franklin
| |
Collapse
|
43
|
Yu ZT, Nanthakumar NN, Newburg DS. The Human Milk Oligosaccharide 2'-Fucosyllactose Quenches Campylobacter jejuni-Induced Inflammation in Human Epithelial Cells HEp-2 and HT-29 and in Mouse Intestinal Mucosa. J Nutr 2016; 146:1980-1990. [PMID: 27629573 PMCID: PMC5037868 DOI: 10.3945/jn.116.230706] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/09/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Campylobacter jejuni causes diarrhea worldwide; young children are most susceptible. Binding of virulent C. jejuni to the intestinal mucosa is inhibited ex vivo by α1,2-fucosylated carbohydrate moieties, including human milk oligosaccharides (HMOSs). OBJECTIVE The simplest α1,2-fucosylated HMOS structure, 2'-fucosyllactose (2'-FL), can be predominant at ≤5 g/L milk. Although 2'-FL inhibits C. jejuni binding ex vivo and in vivo, the effects of 2'FL on the cell invasion central to C. jejuni pathogenesis have not been tested. Clinical isolates of C. jejuni infect humans, birds, and ferrets, limiting studies on its mammalian pathobiology. METHODS Human epithelial cells HEp-2 and HT-29 infected with the virulent C. jejuni strain 81-176 human isolate were treated with 5 g 2'-FL/L, and the degree of infection and inflammatory response was measured. Four-week-old male wild-type C57BL/6 mice were fed antibiotics to reduce their intestinal microbiota and were inoculated with C. jejuni strain 81-176. The sensitivity of the resulting acute transient enteric infection and immune response to inhibition by 2'-FL ingestion was tested. RESULTS In HEp-2 and HT-29 cells, 2'-FL attenuated 80% of C. jejuni invasion (P < 0.05) and suppressed the release of mucosal proinflammatory signals of interleukin (IL) 8 by 60-70%, IL-1β by 80-90%, and the neutrophil chemoattractant macrophage inflammatory protein 2 (MIP-2) by 50% (P < 0.05). Ingestion of 2'-FL by mice reduced C. jejuni colonization by 80%, weight loss by 5%, histologic features of intestinal inflammation by 50-70%, and induction of inflammatory signaling molecules of the acute-phase mucosal immune response by 50-60% (P < 0.05). This acute model did not induce IL-17 (adaptive T cell response), a chronic response. CONCLUSIONS In human cells in vitro (HEp-2, HT-29) and in a mouse infection model that recapitulated key pathologic features of C. jejuni clinical disease, 2'-FL inhibited pathogenesis and its sequelae. These data strongly support the hypothesis that 2'-FL represents a new class of oral agent for prevention, and potentially for treatment, of specific enteric infectious diseases.
Collapse
Affiliation(s)
- Zhuo-Teng Yu
- Department of Biology, Boston College, Chestnut Hill, MA; and
| | - N Nanda Nanthakumar
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - David S Newburg
- Department of Biology, Boston College, Chestnut Hill, MA; and
| |
Collapse
|
44
|
Du X, Wang N, Ren F, Tang H, Jiao X, Huang J. cj0371: A Novel Virulence-Associated Gene of Campylobacter jejuni. Front Microbiol 2016; 7:1094. [PMID: 27471500 PMCID: PMC4944492 DOI: 10.3389/fmicb.2016.01094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/30/2016] [Indexed: 01/03/2023] Open
Abstract
Campylobacter jejuni is the major cause of human bacterial diarrhea worldwide. Its pathogenic mechanism remains poorly understood. cj0371 is a novel gene that was uncovered using immunoscreening. There have been no previous reports regarding its function. In this study, we constructed an insertion mutant and complement of this gene in C. jejuni and examined changes in virulence. We observed that the cj0371 mutant showed significantly increased invasion and colonization ability. We also investigated the role of cj0371 in motility, chemotaxis, and growth kinetics to further study its function. We found that the cj0371 mutant displays hypermotility, enhanced chemotaxis, and enhanced growth kinetics. In addition, we localized the Cj0371 protein at the poles of C. jejuni by fluorescence microscopy. We present data that collectively significantly proves our hypothesis that cj0371 is a new virulence-associated gene and through the influence of chemotaxis plays a negative role in C. jejuni pathogenicity.
Collapse
Affiliation(s)
- Xueqing Du
- Jiangsu Key Lab of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou, China
| | - Nan Wang
- Jiangsu Key Lab of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou, China
| | - Fangzhe Ren
- Jiangsu Key Lab of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou, China
| | - Hong Tang
- Jiangsu Key Lab of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou, China
| | - Xinan Jiao
- Jiangsu Key Lab of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou, China
| | - Jinlin Huang
- Jiangsu Key Lab of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou, China
| |
Collapse
|
45
|
Dwivedi R, Nothaft H, Garber J, Xin Kin L, Stahl M, Flint A, van Vliet AHM, Stintzi A, Szymanski CM. L-fucose influences chemotaxis and biofilm formation in Campylobacter jejuni. Mol Microbiol 2016; 101:575-89. [PMID: 27145048 DOI: 10.1111/mmi.13409] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 04/15/2016] [Indexed: 11/28/2022]
Abstract
Campylobacter jejuni and Campylobacter coli are zoonotic pathogens once considered asaccharolytic, but are now known to encode pathways for glucose and fucose uptake/metabolism. For C. jejuni, strains with the fuc locus possess a competitive advantage in animal colonization models. We demonstrate that this locus is present in > 50% of genome-sequenced strains and is prevalent in livestock-associated isolates of both species. To better understand how these campylobacters sense nutrient availability, we examined biofilm formation and chemotaxis to fucose. C. jejuni NCTC11168 forms less biofilms in the presence of fucose, although its fucose permease mutant (fucP) shows no change. In a newly developed chemotaxis assay, both wild-type and the fucP mutant are chemotactic towards fucose. C. jejuni 81-176 naturally lacks the fuc locus and is unable to swim towards fucose. Transfer of the NCTC11168 locus into 81-176 activated fucose uptake and chemotaxis. Fucose chemotaxis also correlated with possession of the pathway for C. jejuni RM1221 (fuc+) and 81116 (fuc-). Systematic mutation of the NCTC11168 locus revealed that Cj0485 is necessary for fucose metabolism and chemotaxis. This study suggests that components for fucose chemotaxis are encoded within the fuc locus, but downstream signals only in fuc + strains, are involved in coordinating fucose availability with biofilm development.
Collapse
Affiliation(s)
- Ritika Dwivedi
- Alberta Glycomics Centre and Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Harald Nothaft
- Alberta Glycomics Centre and Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Jolene Garber
- Alberta Glycomics Centre and Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Lin Xin Kin
- Alberta Glycomics Centre and Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Martin Stahl
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Annika Flint
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Arnoud H M van Vliet
- Institute of Food Research, Gut Health and Food Safety Programme, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Alain Stintzi
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Christine M Szymanski
- Alberta Glycomics Centre and Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| |
Collapse
|
46
|
Robust bioengineered 3D functional human intestinal epithelium. Sci Rep 2015; 5:13708. [PMID: 26374193 PMCID: PMC4571649 DOI: 10.1038/srep13708] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 08/04/2015] [Indexed: 12/29/2022] Open
Abstract
Intestinal functions are central to human physiology, health and disease. Options to study these functions with direct relevance to the human condition remain severely limited when using conventional cell cultures, microfluidic systems, organoids, animal surrogates or human studies. To replicate in vitro the tissue architecture and microenvironments of native intestine, we developed a 3D porous protein scaffolding system, containing a geometrically-engineered hollow lumen, with adaptability to both large and small intestines. These intestinal tissues demonstrated representative human responses by permitting continuous accumulation of mucous secretions on the epithelial surface, establishing low oxygen tension in the lumen, and interacting with gut-colonizing bacteria. The newly developed 3D intestine model enabled months-long sustained access to these intestinal functions in vitro, readily integrable with a multitude of different organ mimics and will therefore ensure a reliable ex vivo tissue system for studies in a broad context of human intestinal diseases and treatments.
Collapse
|
47
|
Chandrashekhar K, Gangaiah D, Pina-Mimbela R, Kassem II, Jeon BH, Rajashekara G. Transducer like proteins of Campylobacter jejuni 81-176: role in chemotaxis and colonization of the chicken gastrointestinal tract. Front Cell Infect Microbiol 2015; 5:46. [PMID: 26075188 PMCID: PMC4444964 DOI: 10.3389/fcimb.2015.00046] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 05/11/2015] [Indexed: 01/06/2023] Open
Abstract
Transducer Like Proteins (Tlps), also known as methyl accepting chemotaxis proteins (MCP), enable enteric pathogens to respond to changing nutrient levels in the environment by mediating taxis toward or away from specific chemoeffector molecules. Despite recent advances in the characterization of chemotaxis responses in Campylobacter jejuni, the impact of Tlps on the adaptation of this pathogen to disparate niches and hosts is not fully characterized. The latter is particularly evident in the case of C. jejuni 81-176, a strain that is known to be highly invasive. Furthermore, the cytoplasmic group C Tlps (Tlp5, 6, and 8) were not extensively evaluated. Here, we investigated the role of C. jejuni 81-176 Tlps in chemotaxis toward various substrates, biofilm formation, in vitro interaction with human intestinal cells, and chicken colonization. We found that the Δtlp6 and Δtlp10 mutants exhibited decreased chemotaxis toward aspartate, whereas the Δtlp6 mutant displayed a decreased chemotaxis toward Tri-Carboxylic Acid (TCA) cycle intermediates such as pyruvate, isocitrate, and succinate. Our findings also corroborated that more than one Tlp is involved in mediating chemotaxis toward the same nutrient. The deletion of tlps affected important phenotypes such as motility, biofilm formation, and invasion of human intestinal epithelial cells (INT-407). The Δtlp8 mutant displayed increased motility in soft agar and showed decreased biofilm formation. The Δtlp8 and Δtlp9 mutants were significantly defective in invasion in INT-407 cells. The Δtlp10 mutant was defective in colonization of the chicken proximal and distal gastrointestinal tract, while the Δtlp6 and Δtlp8 mutants showed reduced colonization of the duodenum and jejunum. Our results highlight the importance of Tlps in C. jejuni's adaptation and pathobiology.
Collapse
Affiliation(s)
- Kshipra Chandrashekhar
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research Development Center, The Ohio State University Wooster, OH, USA
| | - Dharanesh Gangaiah
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research Development Center, The Ohio State University Wooster, OH, USA
| | - Ruby Pina-Mimbela
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research Development Center, The Ohio State University Wooster, OH, USA
| | - Issmat I Kassem
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research Development Center, The Ohio State University Wooster, OH, USA
| | - Byeong H Jeon
- Department of Environmental Health Sciences, School of Public Health, University of Alberta Edmonton, AB, Canada
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research Development Center, The Ohio State University Wooster, OH, USA
| |
Collapse
|
48
|
The Intestinal Microbiota Influences Campylobacter jejuni Colonization and Extraintestinal Dissemination in Mice. Appl Environ Microbiol 2015; 81:4642-50. [PMID: 25934624 DOI: 10.1128/aem.00281-15] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/24/2015] [Indexed: 12/15/2022] Open
Abstract
Campylobacter jejuni is a leading cause of human foodborne gastroenteritis worldwide. The interactions between this pathogen and the intestinal microbiome within a host are of interest as endogenous intestinal microbiota mediates a form of resistance to the pathogen. This resistance, termed colonization resistance, is the ability of commensal microbiota to prevent colonization by exogenous pathogens or opportunistic commensals. Although mice normally demonstrate colonization resistance to C. jejuni, we found that mice treated with ampicillin are colonized by C. jejuni, with recovery of Campylobacter from the colon, mesenteric lymph nodes, and spleen. Furthermore, there was a significant reduction in recovery of C. jejuni from ampicillin-treated mice inoculated with a C. jejuni virulence mutant (ΔflgL strain) compared to recovery of mice inoculated with the C. jejuni wild-type strain or the C. jejuni complemented isolate (ΔflgL/flgL). Comparative analysis of the microbiota from nontreated and ampicillin-treated CBA/J mice led to the identification of a lactic acid-fermenting isolate of Enterococcus faecalis that prevented C. jejuni growth in vitro and limited C. jejuni colonization of mice. Next-generation sequencing of DNA from fecal pellets that were collected from ampicillin-treated CBA/J mice revealed a significant decrease in diversity of operational taxonomic units (OTUs) compared to that in control (nontreated) mice. Taken together, we have demonstrated that treatment of mice with ampicillin alters the intestinal microbiota and permits C. jejuni colonization. These findings provide valuable insights for researchers using mice to investigate C. jejuni colonization factors, virulence determinants, or the mechanistic basis of probiotics.
Collapse
|
49
|
Stahl M, Vallance BA. Insights into Campylobacter jejuni colonization of the mammalian intestinal tract using a novel mouse model of infection. Gut Microbes 2015; 6:143-8. [PMID: 25831043 PMCID: PMC4615362 DOI: 10.1080/19490976.2015.1016691] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A lack of relevant disease models for Campylobacter jejuni has long been an obstacle to research into this common enteric pathogen. We recently published that mice deficient in Single IgG Interleukin-1 related receptor (SIGIRR), a repressor of MyD88-dependent innate immune signaling, were highly susceptible to enteric infection by murine bacterial pathogens. Subsequently, we successfully employed these mice as an animal model for the human pathogen C. jejuni and gained substantial new insights into infection by this pathogen. The infected mice developed significant intestinal inflammation, primarily via TLR4 stimulation. Furthermore, the resulting gastroenteritis was dependent on C. jejuni pathogenesis as bacterial strains suffering mutations in key virulence factors were attenuated in causing disease. The ability to infect SIGIRR-deficient mice with C. jejuni sheds new light onto how these bacteria colonize the mucus layer of the intestinal tract, invade epithelial cells, and raises new prospects for studying the virulence strategies and pathogenesis of C. jejuni.
Collapse
Affiliation(s)
- Martin Stahl
- Division of Gastroenterology; BC's Children's Hospital; The Child and Family Research Institute and The University of British Columbia; Vancouver, BC Canada
| | - Bruce A Vallance
- Division of Gastroenterology; BC's Children's Hospital; The Child and Family Research Institute and The University of British Columbia; Vancouver, BC Canada,Correspondence to: Bruce A Vallance;
| |
Collapse
|
50
|
Bolton DJ. Campylobacter virulence and survival factors. Food Microbiol 2014; 48:99-108. [PMID: 25790997 DOI: 10.1016/j.fm.2014.11.017] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/26/2014] [Accepted: 11/30/2014] [Indexed: 10/24/2022]
Abstract
Despite over 30 years of research, campylobacteriosis is the most prevalent foodborne bacterial infection in many countries including in the European Union and the United States of America. However, relatively little is known about the virulence factors in Campylobacter or how an apparently fragile organism can survive in the food chain, often with enhanced pathogenicity. This review collates information on the virulence and survival determinants including motility, chemotaxis, adhesion, invasion, multidrug resistance, bile resistance and stress response factors. It discusses their function in transition through the food processing environment and human infection. In doing so it provides a fundamental understanding of Campylobacter, critical for improved diagnosis, surveillance and control.
Collapse
Affiliation(s)
- Declan J Bolton
- Food Safety Department, Teagasc Food Research Centre, Ashtown, Dublin 15, Ireland.
| |
Collapse
|