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
|
Elmi A, Nasher F, Dorrell N, Wren B, Gundogdu O. Revisiting Campylobacter jejuni Virulence and Fitness Factors: Role in Sensing, Adapting, and Competing. Front Cell Infect Microbiol 2021; 10:607704. [PMID: 33614526 PMCID: PMC7887314 DOI: 10.3389/fcimb.2020.607704] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial foodborne gastroenteritis world wide and represents a major public health concern. Over the past two decades, significant progress in functional genomics, proteomics, enzymatic-based virulence profiling (EBVP), and the cellular biology of C. jejuni have improved our basic understanding of this important pathogen. We review key advances in our understanding of the multitude of emerging virulence factors that influence the outcome of C. jejuni–mediated infections. We highlight, the spatial and temporal dynamics of factors that promote C. jejuni to sense, adapt and survive in multiple hosts. Finally, we propose cohesive research directions to obtain a comprehensive understanding of C. jejuni virulence mechanisms.
Collapse
Affiliation(s)
- Abdi Elmi
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Fauzy Nasher
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nick Dorrell
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Brendan Wren
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
3
|
Kelley BR, Lu J, Haley KP, Gaddy JA, Johnson JG. Metal homeostasis in pathogenic Epsilonproteobacteria: mechanisms of acquisition, efflux, and regulation. Metallomics 2021; 13:mfaa002. [PMID: 33570133 PMCID: PMC8043183 DOI: 10.1093/mtomcs/mfaa002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/25/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022]
Abstract
Epsilonproteobacteria are a diverse class of eubacteria within the Proteobacteria phylum that includes environmental sulfur-reducing bacteria and the human pathogens, Campylobacter jejuni and Helicobacter pylori. These pathogens infect and proliferate within the gastrointestinal tracts of multiple animal hosts, including humans, and cause a variety of disease outcomes. While infection of these hosts provides nutrients for the pathogenic Epsilonproteobacteria, many hosts have evolved a variety of strategies to either sequester metals from the invading pathogen or exploit the toxicity of metals and drive their accumulation as an antimicrobial strategy. As a result, C. jejuni and H. pylori have developed mechanisms to sense changes in metal availability and regulate their physiology in order to respond to either metal limitation or accumulation. In this review, we will discuss the challenges of metal availability at the host-pathogen interface during infection with C. jejuni and H. pylori and describe what is currently known about how these organisms alter their gene expression and/or deploy bacterial virulence factors in response to these environments.
Collapse
Affiliation(s)
- Brittni R Kelley
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Kathryn P Haley
- Department of Biology, Grand Valley State University, Grand Rapids, MI, USA
| | - Jennifer A Gaddy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
- Tennessee Valley Healthcare Systems, Department of Veterans Affairs, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | |
Collapse
|
4
|
Klančnik A, Šimunović K, Kovac J, Sahin O, Wu Z, Vučković D, Abram M, Zhang Q, Smole Možina S. The Anti- Campylobacter Activity and Mechanisms of Pinocembrin Action. Microorganisms 2019; 7:E675. [PMID: 31835624 PMCID: PMC6955772 DOI: 10.3390/microorganisms7120675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/05/2019] [Accepted: 12/07/2019] [Indexed: 11/11/2022] Open
Abstract
We investigated the anti-Campylobacter activity of pinocembrin and its mechanism of action, as well as Campylobacter responses to pinocembrin treatment at the genetic and phenotypic levels, using C. jejuni NCTC 11168 and a multidrug efflux system repressor mutant (11168ΔcmeR). At its minimal inhibitory concentration, pinocembrin significantly increased cell membrane permeability of Campylobacter. Interestingly, at sub-inhibitory concentrations, pinocembrin did not significantly alter membrane functionality and it increased bacterial fitness. Treatment with pinocembrin evoked decreased expression of ribosomal proteins and down-regulation of several NADH dehydrogenase I chain subunits and proteins involved in iron uptake. This suggests altered protein production and redox cycle and iron metabolism. Interestingly, the chelation of Fe ions during the treatment with pinocembrin increased C. jejuni survival, although there was no increase in the formation of reactive oxygen species. Pre-treatment of C. jejuni with sub-inhibitory concentrations of pinocembrin for 2 h resulted in a 1 log decrease in C. jejuni colony forming units in mice liver at 8 days post-infection, compared to untreated C. jejuni. These findings suggest that pinocembrin modulates the metabolic activity of C. jejuni and that pre-treatment of C. jejuni with pinocembrin influences its virulence potential in mice. This anti-Campylobacter potential of pinocembrin warrants further investigation.
Collapse
Affiliation(s)
- Anja Klančnik
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia; (K.Š.); (J.K.); (S.S.M.)
| | - Katarina Šimunović
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia; (K.Š.); (J.K.); (S.S.M.)
| | - Jasna Kovac
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia; (K.Š.); (J.K.); (S.S.M.)
- Department of Food Science, The Pennsylvania State University, University Park, PA 16802, USA
| | - Orhan Sahin
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (O.S.); (Z.W.); (Q.Z.)
| | - Zuowei Wu
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (O.S.); (Z.W.); (Q.Z.)
| | - Darinka Vučković
- Department of Microbiology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (D.V.); (M.A.)
| | - Maja Abram
- Department of Microbiology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (D.V.); (M.A.)
| | - Qijing Zhang
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; (O.S.); (Z.W.); (Q.Z.)
| | - Sonja Smole Možina
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia; (K.Š.); (J.K.); (S.S.M.)
| |
Collapse
|
5
|
Askoura M, Youns M, Halim Hegazy WA. Investigating the influence of iron on Campylobacter jejuni transcriptome in response to acid stress. Microb Pathog 2019; 138:103777. [PMID: 31600543 DOI: 10.1016/j.micpath.2019.103777] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 01/12/2023]
Abstract
The capacity of C. jejuni to survive acid and capture iron is a requirement for C. jejuni to colonize host and cause infection. Herein, we aimed to characterize the influence of iron on Campylobacter acid response. The capacity of C. jejuni to survive acid stress was greatly enhanced in presence of iron. Moreover, the acid stimulon of C. jejuni under iron-enriched condition was investigated using the microarray approach. A total of 211 genes were differentially expressed in C. jejuni. Differentially expressed genes were included in 21 functional groups that control Campylobacter physiology. C. jejuni induced expression of many genes that were previously shown to be important for Campylobacter acid survival such as flagella biogenesis genes and genes involved in cell envelope biogenesis. The microarray results were validated using RT-qPCR where there was a great similarity in data obtained by both techniques. Finally, comparative analysis with previous studies showed that acid exposure induced expression of many genes in C. jejuni that were not detected in other studies such as genes encoding for the heat shock proteins GroEL and GroES. Current data could help us understand the mechanism of C. jejuni acid survival and consequently overcome infection by this enteric pathogen.
Collapse
Affiliation(s)
- Momen Askoura
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | - Mahmoud Youns
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt; Department of Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg, Germany.
| | - Wael Abdel Halim Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt; Department of Pharmaceutics, College of Pharmacy, University of Florida, USA.
| |
Collapse
|
6
|
Van TTH, Lacey JA, Vezina B, Phung C, Anwar A, Scott PC, Moore RJ. Survival Mechanisms of Campylobacter hepaticus Identified by Genomic Analysis and Comparative Transcriptomic Analysis of in vivo and in vitro Derived Bacteria. Front Microbiol 2019; 10:107. [PMID: 30804905 PMCID: PMC6371046 DOI: 10.3389/fmicb.2019.00107] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/18/2019] [Indexed: 01/01/2023] Open
Abstract
Chickens infected with Campylobacter jejuni or Campylobacter coli are largely asymptomatic, however, infection with the closely related species, Campylobacter hepaticus, can result in Spotty Liver Disease (SLD). C. hepaticus has been detected in the liver, bile, small intestine and caecum of SLD affected chickens. The survival and colonization mechanisms that C. hepaticus uses to colonize chickens remain unknown. In this study, we compared the genome sequences of 14 newly sequenced Australian isolates of C. hepaticus, isolates from outbreaks in the United Kingdom, and reference strains of C. jejuni and C. coli, with the aim of identifying virulence genes associated with SLD. We also carried out global comparative transcriptomic analysis between C. hepaticus recovered from the bile of SLD infected chickens and C. hepaticus grown in vitro. This revealed how the bacteria adapt to proliferate in the challenging host environment in which they are found. Additionally, biochemical experiments confirmed some in silico metabolic predictions. We found that, unlike other Campylobacter sp., C. hepaticus encodes glucose and polyhydroxybutyrate metabolism pathways. This study demonstrated the metabolic plasticity of C. hepaticus, which may contribute to survival in the competitive, nutrient and energy-limited environment of the chicken. Transcriptomic analysis indicated that gene clusters associated with glucose utilization, stress response, hydrogen metabolism, and sialic acid modification may play an important role in the pathogenicity of C. hepaticus. An understanding of the survival and virulence mechanisms that C. hepaticus uses will help to direct the development of effective intervention methods to protect birds from the debilitating effects of SLD.
Collapse
Affiliation(s)
- Thi Thu Hao Van
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Jake A Lacey
- Doherty Department, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ben Vezina
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Canh Phung
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Arif Anwar
- Scolexia Pty Ltd., Moonee Ponds, VIC, Australia
| | | | - Robert J Moore
- School of Science, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
7
|
Teren M, Turonova Michova H, Vondrakova L, Demnerova K. Molecules Autoinducer 2 and cjA and Their Impact on Gene Expression in Campylobacter jejuni. J Mol Microbiol Biotechnol 2019; 28:207-215. [DOI: 10.1159/000495411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 11/14/2018] [Indexed: 11/19/2022] Open
Abstract
Quorum sensing is a widespread form of cell-to-cell communication, which is based on the production of signaling molecules known as autoinducers (AIs). The first group contains highly species-specific N-acyl homoserine lactones (N-AHLs), generally known as AI-1, which are produced by AHL synthase. The second group, possessing the characteristic structure of a furanone ring, are known as AI-2. The enzyme responsible for their production is S-ribosylhomocysteine lyase (LuxS). In <i>Campylobacter jejuni</i>, AI-2 and LuxS play a role in many important processes, including biofilm formation, stress response, motility, expression of virulence factors, and colonization. However, neither the receptor protein nor the exact structure of the AI-2 molecule have been identified to date. Similarly, little is known about the possible existence of AHL-synthase producing AI-1 and its impact on gene expression. Recently, an analogue of homoserine lactone, called cjA, was isolated from a cell-free supernatant of <i>C. jejuni</i> strain<i></i> 81–176 and from the food isolate c11. The molecule cjA particularly impacted the expression of virulence factors and biofilm formation. This review summarizes the role of AI-2 and cjA in the context of biofilm formation, motility, stress responses, and expression of virulence factors.
Collapse
|
8
|
Gomes CN, Passaglia J, Vilela FP, Pereira da Silva FM, Duque SS, Falcão JP. High survival rates of Campylobacter coli under different stress conditions suggest that more rigorous food control measures might be needed in Brazil. Food Microbiol 2018. [DOI: 10.1016/j.fm.2018.02.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
9
|
Lourenço M, De Sordi L, Debarbieux L. The Diversity of Bacterial Lifestyles Hampers Bacteriophage Tenacity. Viruses 2018; 10:v10060327. [PMID: 29914064 PMCID: PMC6024678 DOI: 10.3390/v10060327] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
Phage therapy is based on a simple concept: the use of a virus (bacteriophage) that is capable of killing specific pathogenic bacteria to treat bacterial infections. Since the pioneering work of Félix d’Herelle, bacteriophages (phages) isolated in vitro have been shown to be of therapeutic value. Over decades of study, a large number of rather complex mechanisms that are used by phages to hijack bacterial resources and to produce their progeny have been deciphered. While these mechanisms have been identified and have been studied under optimal conditions in vitro, much less is known about the requirements for successful viral infections in relevant natural conditions. This is particularly true in the context of phage therapy. Here, we highlight the parameters affecting phage replication in both in vitro and in vivo environments, focusing, in particular, on the mammalian digestive tract. We propose avenues for increasing the knowledge-guided implementation of phages as therapeutic tools.
Collapse
Affiliation(s)
- Marta Lourenço
- Department of Microbiology, Institut Pasteur, F-75015 Paris, France.
- Collège Doctoral, Sorbonne Université, F-75005 Paris, France.
| | - Luisa De Sordi
- Department of Microbiology, Institut Pasteur, F-75015 Paris, France.
| | | |
Collapse
|
10
|
Trigui H, Lee K, Thibodeau A, Lévesque S, Mendis N, Fravalo P, Letellier A, Faucher SP. Phenotypic and Transcriptomic Responses of Campylobacter jejuni Suspended in an Artificial Freshwater Medium. Front Microbiol 2017; 8:1781. [PMID: 28979243 PMCID: PMC5611540 DOI: 10.3389/fmicb.2017.01781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/01/2017] [Indexed: 12/24/2022] Open
Abstract
Campylobacter jejuni is the leading cause of campylobacteriosis in the developed world. Although most cases are caused by consumption of contaminated meat, a significant proportion is linked to ingestion of contaminated water. The differences between C. jejuni strains originating from food products and those isolated from water are poorly understood. Working under the hypothesis that water-borne C. jejuni strains are better equipped at surviving the nutrient-poor aquatic environment than food-borne strains, the present study aims to characterize these differences using outbreak strains 81116 and 81-176. Strain 81116 caused a campylobacteriosis outbreak linked to consumption of water, while strain 81-176 was linked to consumption of raw milk. CFU counts and viability assays showed that 81116 survives better than 81-176 at 4°C in a defined freshwater medium (Fraquil). Moreover, 81116 was significantly more resistant to oxidative stress and bile salt than strain 81-176 in Fraquil. To better understand the genetic response of 81116 to water, a transcriptomic profiling study was undertaken using microarrays. Compared to rich broth, strain 81116 represses genes involved in amino acid uptake and metabolism, as well as genes involved in costly biosynthetic processes such as replication, translation, flagellum synthesis and virulence in response to Fraquil. In accordance with the observed increase in stress resistance in Fraquil, 81116 induces genes involved in resistance to oxidative stress and bile salt. Interestingly, genes responsible for cell wall synthesis were also induced upon Fraquil exposure. Finally, twelve unique genes were expressed in Fraquil; however, analysis of their distribution in animal and water isolates showed that they are not uniquely and ubiquitously present in water isolates, and thus, unlikely to play a major role in adaptation to water. Our results show that some C. jejuni strains are more resilient than others, thereby challenging current water management practices. The response of 81116 to Fraquil serves as a starting point to understand the adaptation of C. jejuni to water and its subsequent transmission.
Collapse
Affiliation(s)
- Hana Trigui
- Department of Natural Resource Sciences, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-BellevueQC, Canada
| | - Kristen Lee
- Department of Natural Resource Sciences, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-BellevueQC, Canada
| | - Alexandre Thibodeau
- Research Chair in Meat Safety, Department of Pathology and Microbiology, University of Montreal, Saint-HyacintheQC, Canada
| | - Simon Lévesque
- Laboratoire de Santé Publique du Québec (LSPQ)/Institut National de Santé Publique du Québec, Sainte-Anne-de-BellevueQC, Canada
| | - Nilmini Mendis
- Department of Natural Resource Sciences, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-BellevueQC, Canada
| | - Philippe Fravalo
- Research Chair in Meat Safety, Department of Pathology and Microbiology, University of Montreal, Saint-HyacintheQC, Canada
| | - Ann Letellier
- Research Chair in Meat Safety, Department of Pathology and Microbiology, University of Montreal, Saint-HyacintheQC, Canada
| | - Sébastien P. Faucher
- Department of Natural Resource Sciences, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-BellevueQC, Canada
| |
Collapse
|
11
|
van der Stel AX, Boogerd FC, Huynh S, Parker CT, van Dijk L, van Putten JPM, Wösten MMSM. Generation of the membrane potential and its impact on the motility, ATP production and growth in Campylobacter jejuni. Mol Microbiol 2017; 105:637-651. [PMID: 28586527 DOI: 10.1111/mmi.13723] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/29/2017] [Accepted: 06/04/2017] [Indexed: 02/06/2023]
Abstract
The generation of a membrane potential (Δψ), the major constituent of the proton motive force (pmf), is crucial for ATP synthesis, transport of nutrients and flagellar rotation. Campylobacter jejuni harbors a branched electron transport chain, enabling respiration with different electron donors and acceptors. Here, we demonstrate that a relatively high Δψ is only generated in the presence of either formate as electron donor or oxygen as electron acceptor, in combination with an acceptor/donor respectively. We show the necessity of the pmf for motility and growth of C. jejuni. ATP generation is not only accomplished by oxidative phosphorylation via the pmf, but also by substrate-level phosphorylation via the enzyme AckA. In response to a low oxygen tension, C. jejuni increases the transcription and activity of the donor complexes formate dehydrogenase (FdhABC) and hydrogenase (HydABCD) as well as the transcription of the alternative respiratory acceptor complexes. Our findings suggest that in the gut of warm-blooded animals, C. jejuni depends on at least formate or hydrogen as donor (in the anaerobic lumen) or oxygen as acceptor (near the epithelial cells) to generate a pmf that sustains efficient motility and growth for colonization and pathogenesis.
Collapse
Affiliation(s)
| | - Fred C Boogerd
- Department of Molecular Cell Biology, VU University Amsterdam, Amsterdam, The Netherlands
| | - Steven Huynh
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, CA, USA
| | - Craig T Parker
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, CA, USA
| | - Linda van Dijk
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Marc M S M Wösten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
12
|
Kreuder AJ, Schleining JA, Yaeger M, Zhang Q, Plummer PJ. RNAseq Reveals Complex Response of Campylobacter jejuni to Ovine Bile and In vivo Gallbladder Environment. Front Microbiol 2017; 8:940. [PMID: 28611744 PMCID: PMC5447181 DOI: 10.3389/fmicb.2017.00940] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/10/2017] [Indexed: 02/01/2023] Open
Abstract
Colonization of the gallbladder by enteric pathogens such as Salmonella typhi, Listeria monocytogenes, and Campylobacter jejuni is thought to play a key role in transmission and persistence of these important zoonotic agents; however, little is known about the molecular mechanisms that allow for bacterial survival within this harsh environment. Recently, a highly virulent C. jejuni sheep abortion (SA) clone represented by the clinical isolate IA3902 has emerged as the dominant cause for sheep abortion in the United States. Previous studies have indicated that the C. jejuni clone SA can frequently be isolated from the gallbladders of otherwise healthy sheep, suggesting that the gallbladder may serve as an important reservoir for infection. To begin to understand the molecular mechanisms associated with survival in the host gallbladder, C. jejuni IA3902 was exposed for up to 24 h to both the natural ovine host in vivo gallbladder environment, as well as ovine bile in vitro. Following exposure, total RNA was isolated from the bile and high throughput deep sequencing of strand specific rRNA-depleted total RNA was used to characterize the transcriptome of IA3902 under these conditions. Our results demonstrated for the first time the complete transcriptome of C. jejuni IA3902 during exposure to an important host environment, the sheep gallbladder. Exposure to the host environment as compared to in vitro bile alone provided a more robust picture of the complexity of gene regulation required for survival in the host gallbladder. A subset of genes including a large number of protein coding genes as well as seven previously identified non-coding RNAs were confirmed to be differentially expressed within our data, suggesting that they may play a key role in adaptation upon exposure to these conditions. This research provides valuable insights into the molecular mechanisms that may be utilized by C. jejuni IA3902 to colonize and survive within the inhospitable gallbladder environment.
Collapse
Affiliation(s)
- Amanda J Kreuder
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State UniversityAmes, IA, United States.,Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State UniversityAmes, IA, United States
| | - Jennifer A Schleining
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State UniversityAmes, IA, United States
| | - Michael Yaeger
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State UniversityAmes, IA, United States
| | - Qijing Zhang
- Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State UniversityAmes, IA, United States
| | - Paul J Plummer
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State UniversityAmes, IA, United States.,Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State UniversityAmes, IA, United States
| |
Collapse
|
13
|
Rossi CC, de Oliveira LL, de Carvalho Rodrigues D, Ürményi TP, Laport MS, Giambiagi-deMarval M. Expression of the stress-response regulators CtsR and HrcA in the uropathogen Staphylococcus saprophyticus during heat shock. Antonie Van Leeuwenhoek 2017; 110:1105-1111. [PMID: 28455762 DOI: 10.1007/s10482-017-0881-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/22/2017] [Indexed: 11/28/2022]
Abstract
The uropathogen Staphylococcus saprophyticus is an ubiquitous bacterium but little is known about mechanisms that allow its persistence in diverse environments. Here we evaluated S. saprophyticus growth and survival during heat shock, the expression of stress response regulators ctsR and hrcA through qRT-PCR and heat shock protein synthesis through 35S-Met metabolic labeling. S. saprophyticus does not tolerate temperatures much higher than the optimal 37 °C, as its growth is greatly affected at 42 °C, though viability is maintained up to 48 °C. At 42 °C, the expression of ctsR and hrcA repressor genes approximately triple when compared to 37 °C and continue to increase together with temperature till 48 °C. Expression of hrcA peaks after 20 min of heat shock and decreases significantly after 30 min, indicating that heat stress response regulated by this gene may last 20-30 min. An increase in temperature is accompanied by the synthesis of at least eight proteins, three of which are likely the chaperones DnaK, GroEL and ClpB. In silico analysis indicate that the groEL gene may be regulated by HrcA, clpB by CtsR and dnaK by both repressors. This is the first work to discuss heat stress response in S. saprophyticus and a step forward in the understanding of mechanisms that make this a widespread and emergent pathogen.
Collapse
Affiliation(s)
- Ciro César Rossi
- Instituto de Microbiologia Paulo de Góes, Rio de Janeiro, Brazil
| | | | - Deivid de Carvalho Rodrigues
- Instituto de Biofísica Carlos Chagas Filho, Universidade do Rio de Janeiro, Rio de Janeiro, Brazil.,Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Turán Peter Ürményi
- Instituto de Biofísica Carlos Chagas Filho, Universidade do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
14
|
Insights into Campylobacter jejuni colonization and enteritis using a novel infant rabbit model. Sci Rep 2016; 6:28737. [PMID: 27357336 PMCID: PMC4928045 DOI: 10.1038/srep28737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/07/2016] [Indexed: 01/26/2023] Open
Abstract
A lack of relevant disease models for Campylobacter jejuni has long been an obstacle to research into this common enteric pathogen. Here we used an infant rabbit to study C. jejuni infection, which enables us to define several previously unknown but key features of the organism. C. jejuni is capable of systemic invasion in the rabbit, and developed a diarrhea symptom that mimicked that observed in many human campylobacteriosis. The large intestine was the most consistently colonized site and produced intestinal inflammation, where specific cytokines were induced. Genes preferentially expressed during C. jejuni infection were screened, and acs, cj1385, cj0259 seem to be responsible for C. jejuni invasion. Our results demonstrates that the infant rabbit can be used as an alternative experimental model for the study of diarrheagenic Campylobacter species and will be useful in exploring the pathogenesis of other related pathogens.
Collapse
|
15
|
Fields JA, Li J, Gulbronson CJ, Hendrixson DR, Thompson SA. Campylobacter jejuni CsrA Regulates Metabolic and Virulence Associated Proteins and Is Necessary for Mouse Colonization. PLoS One 2016; 11:e0156932. [PMID: 27257952 PMCID: PMC4892619 DOI: 10.1371/journal.pone.0156932] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/20/2016] [Indexed: 12/28/2022] Open
Abstract
Campylobacter jejuni infection is a leading bacterial cause of gastroenteritis and a common antecedent leading to Gullian-Barré syndrome. Our previous data suggested that the RNA-binding protein CsrA plays an important role in regulating several important phenotypes including motility, biofilm formation, and oxidative stress resistance. In this study, we compared the proteomes of wild type, csrA mutant, and complemented csrA mutant C. jejuni strains in an effort to elucidate the mechanisms by which CsrA affects virulence phenotypes. The putative CsrA regulon was more pronounced at stationary phase (111 regulated proteins) than at mid-log phase (25 regulated proteins). Proteins displaying altered expression in the csrA mutant included diverse metabolic functions, with roles in amino acid metabolism, TCA cycle, acetate metabolism, and various other cell processes, as well as pathogenesis-associated characteristics such as motility, chemotaxis, oxidative stress resistance, and fibronectin binding. The csrA mutant strain also showed altered autoagglutination kinetics when compared to the wild type. CsrA specifically bound the 5' end of flaA mRNA, and we demonstrated that CsrA is a growth-phase dependent repressor of FlaA expression. Finally, the csrA mutant exhibited reduced ability to colonize in a mouse model when in competition with the wild type, further underscoring the role of CsrA in C. jejuni colonization and pathogenesis.
Collapse
Affiliation(s)
- Joshua A. Fields
- Department of Medicine, Division of Infectious Diseases, Augusta University, Augusta, GA, 30912, United States of America
- Department of Natural Sciences, Georgia Military College - Augusta, Augusta, GA, 30907, United States of America
| | - Jiaqi Li
- Department of Medicine, Division of Infectious Diseases, Augusta University, Augusta, GA, 30912, United States of America
| | - Connor J. Gulbronson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, United States of America
| | - David R. Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, United States of America
| | - Stuart A. Thompson
- Department of Medicine, Division of Infectious Diseases, Augusta University, Augusta, GA, 30912, United States of America
- * E-mail:
| |
Collapse
|
16
|
Wallace N, Zani A, Abrams E, Sun Y. The Impact of Oxygen on Bacterial Enteric Pathogens. ADVANCES IN APPLIED MICROBIOLOGY 2016; 95:179-204. [PMID: 27261784 DOI: 10.1016/bs.aambs.2016.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bacterial enteric pathogens are responsible for a tremendous amount of foodborne illnesses every year through the consumption of contaminated food products. During their transit from contaminated food sources to the host gastrointestinal tract, these pathogens are exposed and must adapt to fluctuating oxygen levels to successfully colonize the host and cause diseases. However, the majority of enteric infection research has been conducted under aerobic conditions. To raise awareness of the importance in understanding the impact of oxygen, or lack of oxygen, on enteric pathogenesis, we describe in this review the metabolic and physiological responses of nine bacterial enteric pathogens exposed to environments with different oxygen levels. We further discuss the effects of oxygen levels on virulence regulation to establish potential connections between metabolic adaptations and bacterial pathogenesis. While not providing an exhaustive list of all bacterial pathogens, we highlight key differences and similarities among nine facultative anaerobic and microaerobic pathogens in this review to argue for a more in-depth understanding of the diverse impact oxygen levels have on enteric pathogenesis.
Collapse
Affiliation(s)
- N Wallace
- University of Dayton, Dayton, OH, United States
| | - A Zani
- University of Dayton, Dayton, OH, United States
| | - E Abrams
- University of Dayton, Dayton, OH, United States
| | - Y Sun
- University of Dayton, Dayton, OH, United States
| |
Collapse
|
17
|
The Campylobacter jejuni Ferric Uptake Regulator Promotes Acid Survival and Cross-Protection against Oxidative Stress. Infect Immun 2016; 84:1287-1300. [PMID: 26883589 DOI: 10.1128/iai.01377-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/05/2016] [Indexed: 12/29/2022] Open
Abstract
Campylobacter jejuni is a prevalent cause of bacterial gastroenteritis in humans worldwide. The mechanisms by which C. jejuni survives stomach acidity remain undefined. In the present study, we demonstrated that the C. jejuni ferric uptake regulator (Fur) plays an important role in C. jejuni acid survival and acid-induced cross-protection against oxidative stress. A C. jejuni Δfur mutant was more sensitive to acid than the wild-type strain. Profiling of the acid stimulon of the C. jejuni Δfur mutant allowed us to uncover Fur-regulated genes under acidic conditions. In particular, Fur was found to upregulate genes involved in flagellar and cell envelope biogenesis upon acid stress, and mutants with deletions of these genes were found to be defective in surviving acid stress. Interestingly, prior acid exposure of C. jejuni cross-protected against oxidative stress in a catalase (KatA)- and Fur-dependent manner. Western blotting and reverse transcription-quantitative PCR revealed increased expression of KatA upon acid stress. Electrophoretic mobility shift assays (EMSAs) demonstrated that the binding affinity between Fur and the katA promoter is reduced in vitro under conditions of low pH, rationalizing the higher levels of expression of katA under acidic conditions. Strikingly, the Δfur mutant exhibited reduced virulence in both human epithelial cells and the Galleria mellonella infection model. Altogether, this is the first study showing that, in addition to its role in iron metabolism, Fur is an important regulator of C. jejuni acid responses and this function cross-protects against oxidative stress. Moreover, our results clearly demonstrate Fur's important role in C. jejuni pathogenesis.
Collapse
|
18
|
Mobley HLT. Measuring Escherichia coli Gene Expression during Human Urinary Tract Infections. Pathogens 2016; 5:pathogens5010007. [PMID: 26784237 PMCID: PMC4810128 DOI: 10.3390/pathogens5010007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/13/2016] [Accepted: 01/13/2016] [Indexed: 11/25/2022] Open
Abstract
Extraintestinal Escherichiacoli (E. coli) evolved by acquisition of pathogenicity islands, phage, plasmids, and DNA segments by horizontal gene transfer. Strains are heterogeneous but virulent uropathogenic isolates more often have specific fimbriae, toxins, and iron receptors than commensal strains. One may ask whether it is the virulence factors alone that are required to establish infection. While these virulence factors clearly contribute strongly to pathogenesis, bacteria must survive by metabolizing nutrients available to them. By constructing mutants in all major metabolic pathways and co-challenging mice transurethrally with each mutant and the wild type strain, we identified which major metabolic pathways are required to infect the urinary tract. We must also ask what else is E. coli doing in vivo? To answer this question, we examined the transcriptome of E. coli CFT073 in the murine model of urinary tract infection (UTI) as well as for E. coli strains collected and analyzed directly from the urine of patients attending either a urology clinic or a university health clinic for symptoms of UTI. Using microarrays and RNA-seq, we measured in vivo gene expression for these uropathogenic E. coli strains, identifying genes upregulated during murine and human UTI. Our findings allow us to propose a new definition of bacterial virulence.
Collapse
Affiliation(s)
- Harry L T Mobley
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
19
|
Cloning, Expression, Purification, Regulation, and Subcellular Localization of a Mini-protein from Campylobacter jejuni. Curr Microbiol 2016; 72:511-7. [PMID: 26750120 DOI: 10.1007/s00284-015-0980-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/28/2015] [Indexed: 10/22/2022]
Abstract
The Cj1169c-encoded putative protein of Campylobacter was expressed and purified from E. coli after sequence optimization. The purified protein allowed the production of a specific rabbit antiserum that was used to study the protein expression in vitro and its subcellular localization in the bacterial cell and putative interactions with other proteins. This protein is produced in Campylobacter and it clearly localizes into the periplasmic space. The level of protein production depends on factors, including pH, temperature, osmolarity, and growth phase suggesting a role in the Campylobacter environmental adaptation. The cysteine residues present in the sequence are probably involved in disulfide bridges, which may promote covalent interactions with other proteins of the Campylobacter envelope.
Collapse
|
20
|
Yeh H, Hiett KL, Line JE, Jagne JF, Lauer DC. Seroprevalence in Chickens against
Campylobacter jejuni
Flagellar Capping Protein (FliD) in Selected Areas of the United States. Zoonoses Public Health 2015; 63:265-70. [DOI: 10.1111/zph.12237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Indexed: 01/27/2023]
Affiliation(s)
- H.‐Y. Yeh
- United States Department of Agriculture Agricultural Research Service U.S. National Poultry Research Center Poultry Microbiological Safety and Processing Research Unit Athens GA USA
| | - K. L. Hiett
- United States Department of Agriculture Agricultural Research Service U.S. National Poultry Research Center Poultry Microbiological Safety and Processing Research Unit Athens GA USA
| | - J. E. Line
- United States Department of Agriculture Agricultural Research Service U.S. National Poultry Research Center Poultry Microbiological Safety and Processing Research Unit Athens GA USA
| | - J. F. Jagne
- Animal Health Diagnostic Center Cornell University Ithaca NY USA
| | - D. C. Lauer
- Minnesota Poultry Testing Laboratory University of Minnesota Willmar MN USA
| |
Collapse
|
21
|
Reuter M, Periago PM, Mulholland F, Brown HL, van Vliet AHM. A PAS domain-containing regulator controls flagella-flagella interactions in Campylobacter jejuni. Front Microbiol 2015; 6:770. [PMID: 26284050 PMCID: PMC4519771 DOI: 10.3389/fmicb.2015.00770] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/14/2015] [Indexed: 01/09/2023] Open
Abstract
The bipolar flagella of the foodborne bacterial pathogen Campylobacter jejuni confer motility, which is essential for virulence. The flagella of C. jejuni are post-translationally modified, but how this process is controlled is not well understood. In this work, we have identified a novel PAS-domain containing regulatory system, which modulates flagella-flagella interactions in C. jejuni. Inactivation of the cj1387c gene, encoding a YheO-like PAS6 domain linked to a helix-turn-helix domain, resulted in the generation of a tightly associated “cell-train” morphotype, where up to four cells were connected by their flagella. The morphotype was fully motile, resistant to vortexing, accompanied by increased autoagglutination, and was not observed in aflagellated cells. The Δcj1387c mutant displayed increased expression of the adjacent Cj1388 protein, which comprises of a single endoribonuclease L-PSP domain. Comparative genomics showed that cj1387c (yheO) orthologs in bacterial genomes are commonly linked to an adjacent cj1388 ortholog, with some bacteria, including C. jejuni, containing another cj1388-like gene (cj0327). Inactivation of the cj1388 and cj0327 genes resulted in decreased autoagglutination in Tween-20-supplemented media. The Δcj1388 and Δcj0327 mutants were also attenuated in a Galleria larvae-based infection model. Finally, substituting the sole cysteine in Cj1388 for serine prevented Cj1388 dimerization in non-reducing conditions, and resulted in decreased autoagglutination in the presence of Tween-20. We hypothesize that Cj1388 and Cj0327 modulate post-translational modification of the flagella through yet unidentified mechanisms, and propose naming Cj1387 the Campylobacter Flagella Interaction Regulator CfiR, and the Cj1388 and Cj0327 protein as CfiP and CfiQ, respectively.
Collapse
Affiliation(s)
- Mark Reuter
- Institute of Food Research, Gut Health and Food Safety Programme Norwich, UK
| | - Paula M Periago
- Departamento Ingeniería de Alimentos y del Equipamiento Agrícola, Campus de Excelencia Internacional Regional "Campus Mare Nostrum," Escuela Técnica Superior de Ingeniería Agronómica, Universidad Politécnica de Cartagena Cartagena, Spain ; Instituto de Biotecnología Vegetal, Campus de Excelencia Internacional Regional "Campus Mare Nostrum," Universidad Politécnica de Cartagena Cartagena, Spain
| | - Francis Mulholland
- Institute of Food Research, Gut Health and Food Safety Programme Norwich, UK
| | - Helen L Brown
- Institute of Food Research, Gut Health and Food Safety Programme Norwich, UK ; Cardiff School of Health Sciences, Cardiff Metropolitan University Cardiff, UK
| | | |
Collapse
|
22
|
Frank KL, Colomer-Winter C, Grindle SM, Lemos JA, Schlievert PM, Dunny GM. Transcriptome analysis of Enterococcus faecalis during mammalian infection shows cells undergo adaptation and exist in a stringent response state. PLoS One 2014; 9:e115839. [PMID: 25545155 PMCID: PMC4278851 DOI: 10.1371/journal.pone.0115839] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 12/01/2014] [Indexed: 11/30/2022] Open
Abstract
As both a commensal and a major cause of healthcare-associated infections in humans, Enterococcus faecalis is a remarkably adaptable organism. We investigated how E. faecalis adapts in a mammalian host as a pathogen by characterizing changes in the transcriptome during infection in a rabbit model of subdermal abscess formation using transcriptional microarrays. The microarray experiments detected 222 and 291 differentially regulated genes in E. faecalis OG1RF at two and eight hours after subdermal chamber inoculation, respectively. The profile of significantly regulated genes at two hours post-inoculation included genes involved in stress response, metabolism, nutrient acquisition, and cell surface components, suggesting genome-wide adaptation to growth in an altered environment. At eight hours post-inoculation, 88% of the differentially expressed genes were down-regulated and matched a transcriptional profile consistent with a (p)ppGpp-mediated stringent response. Subsequent subdermal abscess infections with E. faecalis mutants lacking the (p)ppGpp synthetase/hydrolase RSH, the small synthetase RelQ, or both enzymes, suggest that intracellular (p)ppGpp levels, but not stringent response activation, influence persistence in the model. The ability of cells to synthesize (p)ppGpp was also found to be important for growth in human serum and whole blood. The data presented in this report provide the first genome-wide insights on E. faecalis in vivo gene expression and regulation measured by transcriptional profiling during infection in a mammalian host and show that (p)ppGpp levels affect viability of E. faecalis in multiple conditions relevant to mammalian infection. The subdermal abscess model can serve as a novel experimental system for studying the E. faecalis stringent response in the context of the mammalian immune system.
Collapse
Affiliation(s)
- Kristi L. Frank
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Cristina Colomer-Winter
- Center for Oral Biology and Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Suzanne M. Grindle
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - José A. Lemos
- Center for Oral Biology and Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Patrick M. Schlievert
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Gary M. Dunny
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| |
Collapse
|
23
|
Hofreuter D. Defining the metabolic requirements for the growth and colonization capacity of Campylobacter jejuni. Front Cell Infect Microbiol 2014; 4:137. [PMID: 25325018 PMCID: PMC4178425 DOI: 10.3389/fcimb.2014.00137] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/11/2014] [Indexed: 01/27/2023] Open
Abstract
During the last decade Campylobacter jejuni has been recognized as the leading cause of bacterial gastroenteritis worldwide. This facultative intracellular pathogen is a member of the Epsilonproteobacteria and requires microaerobic atmosphere and nutrient rich media for efficient proliferation in vitro. Its catabolic capacity is highly restricted in contrast to Salmonella Typhimurium and other enteropathogenic bacteria because several common pathways for carbohydrate utilization are either missing or incomplete. Despite these metabolic limitations, C. jejuni efficiently colonizes various animal hosts as a commensal intestinal inhabitant. Moreover, C. jejuni is tremendously successful in competing with the human intestinal microbiota; an infectious dose of few hundreds bacteria is sufficient to overcome the colonization resistance of humans and can lead to campylobacteriosis. Besides the importance and clear clinical manifestation of this disease, the pathogenesis mechanisms of C. jejuni infections are still poorly understood. In recent years comparative genome sequence, transcriptome and metabolome analyses as well as mutagenesis studies combined with animal infection models have provided a new understanding of how the specific metabolic capacity of C. jejuni drives its persistence in the intestinal habitat of various hosts. Furthermore, new insights into the metabolic requirements that support the intracellular survival of C. jejuni were obtained. Because C. jejuni harbors distinct properties in establishing an infection in comparison to pathogenic Enterobacteriaceae, it represents an excellent organism for elucidating new aspects of the dynamic interaction and metabolic cross talk between a bacterial pathogen, the microbiota and the host.
Collapse
Affiliation(s)
- Dirk Hofreuter
- Hannover Medical School, Institute for Medical Microbiology and Hospital Epidemiology Hannover, Germany
| |
Collapse
|
24
|
Scott NE, Marzook NB, Cain JA, Solis N, Thaysen-Andersen M, Djordjevic SP, Packer NH, Larsen MR, Cordwell SJ. Comparative Proteomics and Glycoproteomics Reveal Increased N-Linked Glycosylation and Relaxed Sequon Specificity in Campylobacter jejuni NCTC11168 O. J Proteome Res 2014; 13:5136-50. [DOI: 10.1021/pr5005554] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | - Morten Thaysen-Andersen
- Department
of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Steven P. Djordjevic
- i3
Institute, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Nicolle H. Packer
- Department
of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Martin R. Larsen
- Protein
Research Group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense 5000, Denmark
| | - Stuart J. Cordwell
- Discipline
of Pathology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
25
|
van der Stel AX, van Mourik A, Heijmen-van Dijk L, Parker CT, Kelly DJ, van de Lest CHA, van Putten JPM, Wösten MMSM. TheCampylobacter jejuni RacRS system regulates fumarate utilization in a low oxygen environment. Environ Microbiol 2014; 17:1049-64. [DOI: 10.1111/1462-2920.12476] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 02/23/2014] [Accepted: 03/30/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Anne-Xander van der Stel
- Department of Infectious Diseases and Immunology; Utrecht University; Utrecht 3584CL The Netherlands
| | - Andries van Mourik
- Department of Infectious Diseases and Immunology; Utrecht University; Utrecht 3584CL The Netherlands
| | - Linda Heijmen-van Dijk
- Department of Infectious Diseases and Immunology; Utrecht University; Utrecht 3584CL The Netherlands
| | - Craig T. Parker
- Produce Safety and Microbiology Research Unit; Agricultural Research Service; U.S. Department of Agriculture; Albany CA 94710 USA
| | - David J. Kelly
- Department of Molecular Biology and Biotechnology; The University of Sheffield; Sheffield S10 2TN UK
| | - Chris H. A. van de Lest
- Department of Biochemistry and Cell Biology; Utrecht University; Utrecht 3584CL The Netherlands
| | - Jos P. M. van Putten
- Department of Infectious Diseases and Immunology; Utrecht University; Utrecht 3584CL The Netherlands
| | - Marc M. S. M. Wösten
- Department of Infectious Diseases and Immunology; Utrecht University; Utrecht 3584CL The Netherlands
| |
Collapse
|
26
|
Comparative genomics of Campylobacter concisus isolates reveals genetic diversity and provides insights into disease association. BMC Genomics 2013; 14:585. [PMID: 23984967 PMCID: PMC3765806 DOI: 10.1186/1471-2164-14-585] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/17/2013] [Indexed: 12/17/2022] Open
Abstract
Background In spite of its association with gastroenteritis and inflammatory bowel diseases, the isolation of Campylobacter concisus from both diseased and healthy individuals has led to controversy regarding its role as an intestinal pathogen. One proposed reason for this is the presence of high genetic diversity among the genomes of C. concisus strains. Results In this study the genomes of six C. concisus strains were sequenced, assembled and annotated including two strains isolated from Crohn’s disease patients (UNSW2 and UNSW3), three from gastroenteritis patients (UNSW1, UNSWCS and ATCC 51562) and one from a healthy individual (ATCC 51561). The genomes of C. concisus BAA-1457 and UNSWCD, available from NCBI, were included in subsequent comparative genomic analyses. The Pan and Core genomes for the sequenced C. concisus strains consisted of 3254 and 1556 protein coding genes, respectively. Conclusion Genes were identified with specific conservation in C. concisus strains grouped by phenotypes such as invasiveness, adherence, motility and diseased states. Phylogenetic trees based on ribosomal RNA sequences and concatenated host-related pathways for the eight C. concisus strains were generated using the neighbor-joining method, of which the 16S rRNA gene and peptidoglycan biosynthesis grouped the C. concisus strains according to their pathogenic phenotypes. Furthermore, 25 non-synonymous amino acid changes with 14 affecting functional domains, were identified within proteins of conserved host-related pathways, which had possible associations with the pathogenic potential of C. concisus strains. Finally, the genomes of the eight C. concisus strains were compared to the nine available genomes of the well-established pathogen Campylobacter jejuni, which identified several important differences in the respiration pathways of these two species. Our findings indicate that C. concisus strains are genetically diverse, and suggest the genomes of this bacterium contain respiration pathways and modifications in the peptidoglycan layer that may play an important role in its virulence.
Collapse
|
27
|
Taveirne ME, Theriot CM, Livny J, DiRita VJ. The complete Campylobacter jejuni transcriptome during colonization of a natural host determined by RNAseq. PLoS One 2013; 8:e73586. [PMID: 23991199 PMCID: PMC3749233 DOI: 10.1371/journal.pone.0073586] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/22/2013] [Indexed: 11/19/2022] Open
Abstract
Campylobacter jejuni is a major human pathogen and a leading cause of bacterial derived gastroenteritis worldwide. C. jejuni regulates gene expression under various environmental conditions and stresses, indicative of its ability to survive in diverse niches. Despite this ability to highly regulate gene transcription, C. jejuni encodes few transcription factors and its genome lacks many canonical transcriptional regulators. High throughput deep sequencing of mRNA transcripts (termed RNAseq) has been used to study the transcriptome of many different organisms, including C. jejuni; however, this technology has yet to be applied to defining the transcriptome of C. jejuni during in vivo colonization of its natural host, the chicken. In addition to its use in profiling the abundance of annotated genes, RNAseq is a powerful tool for identifying and quantifying, as-of-yet, unknown transcripts including non-coding regulatory RNAs, 5’ untranslated regulatory elements, and anti-sense transcripts. Here we report the complete transcriptome of C. jejuni during colonization of the chicken cecum and in two different in vitro growth phases using strand-specific RNAseq. Through this study, we identified over 250 genes differentially expressed in vivo in addition to numerous putative regulatory RNAs, including trans-acting non-coding RNAs and anti-sense transcripts. These latter potential regulatory elements were not identified in two prior studies using ORF-based microarrays, highlighting the power and value of the RNAseq approach. Our results provide new insights into how C. jejuni responds and adapts to the cecal environment and reveals new functions involved in colonization of its natural host.
Collapse
Affiliation(s)
- Michael E. Taveirne
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Casey M. Theriot
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, Division of Pulmonary and Critical Care, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jonathan Livny
- Genome Sequencing and Analysis Program, Broad Institute, Cambridge, Massachusetts, United States of America
- * E-mail: (VJD); (JL)
| | - Victor J. DiRita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail: (VJD); (JL)
| |
Collapse
|
28
|
Adaptive strategies and pathogenesis of Clostridium difficile from in vivo transcriptomics. Infect Immun 2013; 81:3757-69. [PMID: 23897605 DOI: 10.1128/iai.00515-13] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Clostridium difficile is currently the major cause of nosocomial intestinal diseases associated with antibiotic therapy in adults. In order to improve our knowledge of C. difficile-host interactions, we analyzed the genome-wide temporal expression of C. difficile 630 genes during the first 38 h of mouse colonization to identify genes whose expression is modulated in vivo, suggesting that they may play a role in facilitating the colonization process. In the ceca of the C. difficile-monoassociated mice, 549 genes of the C. difficile genome were differentially expressed compared to their expression during in vitro growth, and they were distributed in several functional categories. Overall, our results emphasize the roles of genes involved in host adaptation. Colonization results in a metabolic shift, with genes responsible for the fermentation as well as several other metabolic pathways being regulated inversely to those involved in carbon metabolism. In addition, several genes involved in stress responses, such as ferrous iron uptake or the response to oxidative stress, were regulated in vivo. Interestingly, many genes encoding conserved hypothetical proteins (CHP) were highly and specifically upregulated in vivo. Moreover, genes for all stages of sporulation were quickly induced in vivo, highlighting the observation that sporulation is central to the persistence of C. difficile in the gut and to its ability to spread in the environment. Finally, we inactivated two genes that were differentially expressed in vivo and evaluated the relative colonization fitness of the wild-type and mutant strains in coinfection experiments. We identified a CHP as a putative colonization factor, supporting the suggestion that the in vivo transcriptomic approach can unravel new C. difficile virulence genes.
Collapse
|
29
|
Enhanced, sialoadhesin-dependent uptake of Guillain-Barre syndrome-associated Campylobacter jejuni strains by human macrophages. Infect Immun 2013; 81:2095-103. [PMID: 23529622 DOI: 10.1128/iai.01437-12] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Molecular mimicry between Campylobacter jejuni sialylated lipooligosaccharides (LOS) and human nerve gangliosides can trigger the production of cross-reactive antibodies which induce Guillain-Barré syndrome (GBS). To better understand the immune events leading to GBS, it is essential to know how sialylated LOS are recognized by the immune system. Here, we show that GBS-associated C. jejuni strains bind to human sialoadhesin (hSn), a conserved, mainly macrophage-restricted I-type lectin. Using hSn-transduced THP-1 cells, we observed that C. jejuni strains with α(2,3)-sialylated LOS, including strains expressing GM1a- and GD1a-like epitopes, bind to hSn. This observation is of importance, as these epitopes are frequently the targets of the cross-reactive antibodies detected in GBS patients. Interestingly, the Sn binding domains were not constitutively exposed on the surface of C. jejuni. Heat inactivation and the environmental conditions which food-borne C. jejuni encounters during its passage through the intestinal tract, such as low pH and contact with bile constituents, exposed LOS and facilitated Sn binding. Sn binding enhanced bacterial uptake and increased the production of interleukin-6 (IL-6) by primary human Sn-expressing monocyte-derived macrophages compared to control conditions, where Sn was blocked using neutralizing antibodies or when nonsialylated C. jejuni was used. Sn-mediated uptake has been reported to enhance humoral immune responses. As C. jejuni strains expressing ganglioside mimics GD1a and GM1a are closely associated with GBS, Sn binding may be a determining event in the production of cross-reactive antibodies and the development of GBS.
Collapse
|
30
|
Saxena M, John B, Mu M, Van TTH, Taki A, Coloe PJ, Smooker PM. Strategies to Reduce Campylobacter Colonisation in Chickens. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.provac.2013.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
31
|
Bell JA, Jerome J, Plovanich-Jones AE, Smith EJ, Gettings JR, Kim HY, Landgraf JR, Lefébure T, Kopper JJ, Rathinam VA, St. Charles JL, Buffa BA, Brooks AP, Poe SA, Eaton KA, Stanhope MJ, Mansfield LS. Outcome of infection of C57BL/6 IL-10(-/-) mice with Campylobacter jejuni strains is correlated with genome content of open reading frames up- and down-regulated in vivo. Microb Pathog 2013; 54:1-19. [PMID: 22960579 PMCID: PMC4118490 DOI: 10.1016/j.micpath.2012.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 08/07/2012] [Indexed: 12/12/2022]
Abstract
Human Campylobacter jejuni infection can result in an asymptomatic carrier state, watery or bloody diarrhea, bacteremia, meningitis, or autoimmune neurological sequelae. Infection outcomes of C57BL/6 IL-10(-/-) mice orally infected with twenty-two phylogenetically diverse C. jejuni strains were evaluated to correlate colonization and disease phenotypes with genetic composition of the strains. Variation between strains was observed in colonization, timing of development of clinical signs, and occurrence of enteric lesions. Five pathotypes of C. jejuni in C57BL/6 IL-10(-/-) mice were delineated: little or no colonization, colonization without disease, colonization with enteritis, colonization with hemorrhagic enteritis, and colonization with neurological signs with or without enteritis. Virulence gene content of ten sequenced strains was compared in silico; virulence gene content of twelve additional strains was compared using a C. jejuni pan-genome microarray. Neither total nor virulence gene content predicted pathotype; nor was pathotype correlated with multilocus sequence type. Each strain was unique with regard to absences of known virulence-related loci and/or possession of point mutations and indels, including phase variation, in virulence-related genes. An experiment in C. jejuni 11168-infected germ-free mice showed that expression levels of ninety open reading frames (ORFs) were significantly up- or down-regulated in the mouse cecum at least two-fold compared to in vitro growth. Genomic content of these ninety C. jejuni 11168 ORFs was significantly correlated with the capacity to colonize and cause enteritis in C57BL/6 IL-10(-/-) mice. Differences in gene expression levels and patterns are thus an important determinant of pathotype in C. jejuni strains in this mouse model.
Collapse
Affiliation(s)
- J. A. Bell
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - J.P. Jerome
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Department of Microbiology and Molecular Genetics, College of Natural Sciences, Michigan State University, East Lansing, MI 48824
| | - A. E. Plovanich-Jones
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - E. J. Smith
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - J. R. Gettings
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - H. Y. Kim
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - J. R. Landgraf
- Research Technology Support Facility, Michigan State University, East Lansing, MI 48824
| | - T. Lefébure
- Department of Population Medicine and Diagnostic Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - J. J. Kopper
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Cell and Molecular Biology Program, College of Natural Sciences, Michigan State University, East Lansing, MI 48824
| | - V. A. Rathinam
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - J. L. St. Charles
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - B. A. Buffa
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - A. P. Brooks
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - S. A. Poe
- Laboratory Animal Medicine Unit, University of Michigan Medical School, Ann Arbor, MI 48109; Laboratory Animal Medicine Unit, University of Michigan Medical School, Ann Arbor, MI 48109
| | - K. A. Eaton
- Laboratory Animal Medicine Unit, University of Michigan Medical School, Ann Arbor, MI 48109; Laboratory Animal Medicine Unit, University of Michigan Medical School, Ann Arbor, MI 48109
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - M. J. Stanhope
- Department of Population Medicine and Diagnostic Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - L. S. Mansfield
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
32
|
Fields JA, Thompson SA. Campylobacter jejuni CsrA complements an Escherichia coli csrA mutation for the regulation of biofilm formation, motility and cellular morphology but not glycogen accumulation. BMC Microbiol 2012; 12:233. [PMID: 23051923 PMCID: PMC3534301 DOI: 10.1186/1471-2180-12-233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 10/01/2012] [Indexed: 11/12/2022] Open
Abstract
Background Although Campylobacter jejuni is consistently ranked as one of the leading causes of bacterial diarrhea worldwide, the mechanisms by which C. jejuni causes disease and how they are regulated have yet to be clearly defined. The global regulator, CsrA, has been well characterized in several bacterial genera and is known to regulate a number of independent pathways via a post transcriptional mechanism, but remains relatively uncharacterized in the genus Campylobacter. Previously, we reported data illustrating the requirement for CsrA in several virulence related phenotypes of C. jejuni strain 81–176, indicating that the Csr pathway is important for Campylobacter pathogenesis. Results We compared the Escherichia coli and C. jejuni orthologs of CsrA and characterized the ability of the C. jejuni CsrA protein to functionally complement an E. coli csrA mutant. Phylogenetic comparison of E. coli CsrA to orthologs from several pathogenic bacteria demonstrated variability in C. jejuni CsrA relative to the known RNA binding domains of E. coli CsrA and in several amino acids reported to be involved in E. coli CsrA-mediated gene regulation. When expressed in an E. coli csrA mutant, C. jejuni CsrA succeeded in recovering defects in motility, biofilm formation, and cellular morphology; however, it failed to return excess glycogen accumulation to wild type levels. Conclusions These findings suggest that C. jejuni CsrA is capable of efficiently binding some E. coli CsrA binding sites, but not others, and provide insight into the biochemistry of C. jejuni CsrA.
Collapse
Affiliation(s)
- Joshua A Fields
- Department of Biochemistry and Molecular Biology, Georgia Health Sciences University, 1120 15th Street, Augusta, GA 30912, USA
| | | |
Collapse
|
33
|
Vannucci FA, Foster DN, Gebhart CJ. Comparative transcriptional analysis of homologous pathogenic and non-pathogenic Lawsonia intracellularis isolates in infected porcine cells. PLoS One 2012; 7:e46708. [PMID: 23056413 PMCID: PMC3463550 DOI: 10.1371/journal.pone.0046708] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 09/04/2012] [Indexed: 11/19/2022] Open
Abstract
Lawsonia intracellularis is the causative agent of proliferative enteropathy. This disease affects various animal species, including nonhuman primates, has been endemic in pigs, and is an emerging concern in horses. Non-pathogenic variants obtained through multiple passages in vitro do not induce disease, but bacterial isolates at low passage induce clinical and pathological changes. We hypothesize that genes differentially expressed between pathogenic (passage 10) and non-pathogenic (passage 60) L. intracellularis isolates encode potential bacterial virulence factors. The present study used high-throughput sequencing technology to characterize the transcriptional profiling of a pathogenic and a non-pathogenic homologous L. intracellularis variant during in vitro infection. A total of 401 genes were exclusively expressed by the pathogenic variant. Plasmid-encoded genes and those involved in membrane transporter (e.g. ATP-binding cassette), adaptation and stress response (e.g. transcriptional regulators) were the categories mostly responsible for this wider transcriptional landscape. The entire gene repertoire of plasmid A was repressed in the non-pathogenic variant suggesting its relevant role in the virulence phenotype of the pathogenic variant. Of the 319 genes which were commonly expressed in both pathogenic and non-pathogenic variants, no significant difference was observed by comparing their normalized transcription levels (fold change±2; p<0.05). Unexpectedly, these genes demonstrated a positive correlation (r(2) = 0.81; p<0.05), indicating the involvement of gene silencing (switching off) mechanisms to attenuate virulence properties of the pathogenic variant during multiple cell passages. Following the validation of these results by reverse transcriptase-quantitative PCR using ten selected genes, the present study represents the first report characterizing the transcriptional profile of L. intracellularis. The complexity of the virulence phenotype was demonstrated by the diversity of genes exclusively expressed in the pathogenic isolate. The results support our hypothesis and provide the basis for prospective mechanistic studies regarding specific roles of target genes involved in the pathogenesis, diagnosis and control of proliferative enteropathy.
Collapse
Affiliation(s)
- Fabio A Vannucci
- Department of Veterinary and Biomedical Science, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America.
| | | | | |
Collapse
|
34
|
Abstract
The diarrheal pathogen Campylobacter jejuni and other gastrointestinal bacteria encounter changes in osmolarity in the environment, through exposure to food processing, and upon entering host organisms, where osmotic adaptation can be associated with virulence. In this study, growth profiles, transcriptomics, and phenotypic, mutant, and single-cell analyses were used to explore the effects of hyperosmotic stress exposure on C. jejuni. Increased growth inhibition correlated with increased osmotic concentration, with both ionic and nonionic stressors inhibiting growth at 0.620 total osmol liter(-1). C. jejuni adaptation to a range of osmotic stressors and concentrations was accompanied by severe filamentation in subpopulations, with microscopy indicating septum formation and phenotypic diversity between individual cells in a filament. Population heterogeneity was also exemplified by the bifurcation of colony morphology into small and large variants on salt stress plates. Flow cytometry of C. jejuni harboring green fluorescent protein (GFP) fused to the ATP synthase promoter likewise revealed bimodal subpopulations under hyperosmotic stress. We also identified frequent hyperosmotic stress-sensitive variants within the clonal wild-type population propagated on standard laboratory medium. Microarray analysis following hyperosmotic upshift revealed enhanced expression of heat shock genes and genes encoding enzymes for synthesis of potential osmoprotectants and cross-protective induction of oxidative stress genes. The capsule export gene kpsM was also upregulated, and an acapsular mutant was defective for growth under hyperosmotic stress. For C. jejuni, an organism lacking most conventional osmotic response factors, these data suggest an unusual hyperosmotic stress response, including likely "bet-hedging" survival strategies relying on the presence of stress-fit individuals in a heterogeneous population.
Collapse
|
35
|
FdhTU-modulated formate dehydrogenase expression and electron donor availability enhance recovery of Campylobacter jejuni following host cell infection. J Bacteriol 2012; 194:3803-13. [PMID: 22636777 DOI: 10.1128/jb.06665-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Campylobacter jejuni is a food-borne bacterial pathogen that colonizes the intestinal tract and causes severe gastroenteritis. Interaction with host epithelial cells is thought to enhance severity of disease, and the ability of C. jejuni to modulate its metabolism in different in vivo and environmental niches contributes to its success as a pathogen. A C. jejuni operon comprising two genes that we designated fdhT (CJJ81176_1492) and fdhU (CJJ81176_1493) is conserved in many bacterial species. Deletion of fdhT or fdhU in C. jejuni resulted in apparent defects in adherence and/or invasion of Caco-2 epithelial cells when assessed by CFU enumeration on standard Mueller-Hinton agar. However, fluorescence microscopy indicated that each mutant invaded cells at wild-type levels, instead suggesting roles for FdhTU in either intracellular survival or postinvasion recovery. The loss of fdhU caused reduced mRNA levels of formate dehydrogenase (FDH) genes and a severe defect in FDH activity. Cell infection phenotypes of a mutant deleted for the FdhA subunit of FDH and an ΔfdhU ΔfdhA double mutant were similar to those of a ΔfdhU mutant, which likewise suggested that FdhU and FdhA function in the same pathway. Cell infection assays followed by CFU enumeration on plates supplemented with sodium sulfite abolished the ΔfdhU and ΔfdhA mutant defects and resulted in significantly enhanced recovery of all strains, including wild type, at the invasion and intracellular survival time points. Collectively, our data indicate that FdhTU and FDH are required for optimal recovery following cell infection and suggest that C. jejuni alters its metabolic potential in the intracellular environment.
Collapse
|
36
|
Culligan EP, Sleator RD, Marchesi JR, Hill C. Functional metagenomics reveals novel salt tolerance loci from the human gut microbiome. ISME JOURNAL 2012; 6:1916-25. [PMID: 22534607 DOI: 10.1038/ismej.2012.38] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Metagenomics is a powerful tool that allows for the culture-independent analysis of complex microbial communities. One of the most complex and dense microbial ecosystems known is that of the human distal colon, with cell densities reaching up to 10(12) per gram of faeces. With the majority of species as yet uncultured, there are an enormous number of novel genes awaiting discovery. In the current study, we conducted a functional screen of a metagenomic library of the human gut microbiota for potential salt-tolerant clones. Using transposon mutagenesis, three genes were identified from a single clone exhibiting high levels of identity to a species from the genus Collinsella (closest relative being Collinsella aerofaciens) (COLAER_01955, COLAER_01957 and COLAER_01981), a high G+C, Gram-positive member of the Actinobacteria commonly found in the human gut. The encoded proteins exhibit a strong similarity to GalE, MurB and MazG. Furthermore, pyrosequencing and bioinformatic analysis of two additional fosmid clones revealed the presence of an additional galE and mazG gene, with the highest level of genetic identity to Akkermansia muciniphila and Eggerthella sp. YY7918, respectively. Cloning and heterologous expression of the genes in the osmosensitive strain, Escherichia coli MKH13, resulted in increased salt tolerance of the transformed cells. It is hoped that the identification of atypical salt tolerance genes will help to further elucidate novel salt tolerance mechanisms, and will assist our increased understanding how resident bacteria cope with the osmolarity of the gastrointestinal tract.
Collapse
Affiliation(s)
- Eamonn P Culligan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
37
|
Neal-McKinney JM, Konkel ME. The Campylobacter jejuni CiaC virulence protein is secreted from the flagellum and delivered to the cytosol of host cells. Front Cell Infect Microbiol 2012; 2:31. [PMID: 22919623 PMCID: PMC3417660 DOI: 10.3389/fcimb.2012.00031] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/29/2012] [Indexed: 12/04/2022] Open
Abstract
Campylobacter jejuni is a leading cause of bacterial gastroenteritis worldwide. Acute C. jejuni-mediated disease (campylobacteriosis) involves C. jejuni invasion of host epithelial cells using adhesins (e.g., CadF and FlpA) and secreted proteins [e.g., the Campylobacter invasion antigens (Cia)]. The genes encoding the Cia proteins are up-regulated upon co-culture of C. jejuni with epithelial cells. One of the Cia proteins, CiaC, is required for maximal invasion of host cells by C. jejuni. Previous work has also revealed that CiaC is, in part, responsible for host cell cytoskeletal rearrangements that result in membrane ruffling. This study was performed to test the hypothesis that CiaC is delivered to the cytosol of host cells. To detect the delivery of CiaC into cultured epithelial cells, we used the adenylate cyclase domain (ACD) of Bordetella pertussis CyaA as a reporter. In this study, we found that export and delivery of the C. jejuni Cia proteins into human INT 407 epithelial cells required a functional flagellar hook complex composed of FlgE, FlgK, and FlgL. Assays performed with bacterial culture supernatants supported the hypothesis that CiaC delivery requires bacteria-host cell contact. We also found that CiaC was delivered to host cells by cell-associated (bound) bacteria, as judged by experiments performed with inhibitors that specifically target the cell signaling pathways utilized by C. jejuni for cell invasion. Interestingly, the C. jejuni flgL mutant, which is incapable of exporting and delivering the Cia proteins, did not induce INT 407 cell membrane ruffles. Complementation of the flgL mutant with plasmid-encoded flgL restored the motility and membrane ruffling. These data support the hypothesis that the C. jejuni Cia proteins, which are exported from the flagellum, are delivered to the cytosol of host cells.
Collapse
|
38
|
Stahl M, Butcher J, Stintzi A. Nutrient acquisition and metabolism by Campylobacter jejuni. Front Cell Infect Microbiol 2012; 2:5. [PMID: 22919597 PMCID: PMC3417520 DOI: 10.3389/fcimb.2012.00005] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/21/2012] [Indexed: 12/03/2022] Open
Abstract
The gastrointestinal pathogen Campylobacter jejuni is able to colonize numerous different hosts and compete against the gut microbiota. To do this, it must be able to efficiently acquire sufficient nutrients from its environment to support its survival and rapid growth in the intestine. However, despite almost 50 years of research, many aspects as to how C. jejuni accomplishes this feat remain poorly understood. C. jejuni lacks many of the common metabolic pathways necessary for the use of glucose, galactose, or other carbohydrates upon which most other microbes thrive. It does however make efficient use of citric acid cycle intermediates and various amino acids. C. jejuni readily uses the amino acids aspartate, glutamate, serine, and proline, with certain strains also possessing additional pathways allowing for the use of glutamine and asparagine. More recent work has revealed that some C. jejuni strains can metabolize the sugar l-fucose. This finding has upset years of dogma that C. jejuni is an asaccharolytic organism. C. jejuni also possesses diverse mechanisms for the acquisition of various transition metals that are required for metabolic activities. In particular, iron acquisition is critical for the formation of iron–sulfur complexes. C. jejuni is also unique in possessing both molybdate and tungsten cofactored proteins and thus has an unusual regulatory scheme for these metals. Together these various metabolic and acquisition pathways help C. jejuni to compete and thrive in wide variety of hosts and environments.
Collapse
Affiliation(s)
- Martin Stahl
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa Ottawa, ON, Canada
| | | | | |
Collapse
|
39
|
CmeR-dependent gene Cj0561c is induced more effectively by bile salts than the CmeABC efflux pump in both human and poultry Campylobacter jejuni strains. Res Microbiol 2011; 162:991-8. [DOI: 10.1016/j.resmic.2011.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 08/05/2011] [Indexed: 11/23/2022]
|
40
|
Grabowska AD, Wandel MP, Łasica AM, Nesteruk M, Roszczenko P, Wyszyńska A, Godlewska R, Jagusztyn-Krynicka EK. Campylobacter jejuni dsb gene expression is regulated by iron in a Fur-dependent manner and by a translational coupling mechanism. BMC Microbiol 2011; 11:166. [PMID: 21787430 PMCID: PMC3167755 DOI: 10.1186/1471-2180-11-166] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 07/25/2011] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Many bacterial extracytoplasmic proteins are stabilized by intramolecular disulfide bridges that are formed post-translationally between their cysteine residues. This protein modification plays an important role in bacterial pathogenesis, and is facilitated by the Dsb (disulfide bond) family of the redox proteins. These proteins function in two parallel pathways in the periplasmic space: an oxidation pathway and an isomerization pathway. The Dsb oxidative pathway in Campylobacter jejuni is more complex than the one in the laboratory E. coli K-12 strain. RESULTS In the C. jejuni 81-176 genome, the dsb genes of the oxidative pathway are arranged in three transcriptional units: dsbA2-dsbB-astA, dsbA1 and dba-dsbI. Their transcription responds to an environmental stimulus - iron availability - and is regulated in a Fur-dependent manner. Fur involvement in dsb gene regulation was proven by a reporter gene study in a C. jejuni wild type strain and its isogenic fur mutant. An electrophoretic mobility shift assay (EMSA) confirmed that analyzed genes are members of the Fur regulon but each of them is regulated by a disparate mechanism, and both the iron-free and the iron-complexed Fur are able to bind in vitro to the C. jejuni promoter regions. This study led to identification of a new iron- and Fur-regulated promoter that drives dsbA1 gene expression in an indirect way. Moreover, the present work documents that synthesis of DsbI oxidoreductase is controlled by the mechanism of translational coupling. The importance of a secondary dba-dsbI mRNA structure for dsbI mRNA translation was verified by estimating individual dsbI gene expression from its own promoter. CONCLUSIONS The present work shows that iron concentration is a significant factor in dsb gene transcription. These results support the concept that iron concentration - also through its influence on dsb gene expression - might control the abundance of extracytoplasmic proteins during different stages of infection. Our work further shows that synthesis of the DsbI membrane oxidoreductase is controlled by a translational coupling mechanism. The dba expression is not only essential for the translation of the downstream dsbI gene, but also Dba protein that is produced might regulate the activity and/or stability of DsbI.
Collapse
Affiliation(s)
- Anna D Grabowska
- Department of Bacterial Genetics, Institute of Microbiology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Routh MD, Zalucki Y, Su CC, Zhang Q, Shafer WM, Yu EW. Efflux pumps of the resistance-nodulation-division family: a perspective of their structure, function, and regulation in gram-negative bacteria. ADVANCES IN ENZYMOLOGY AND RELATED AREAS OF MOLECULAR BIOLOGY 2011; 77:109-46. [PMID: 21692368 DOI: 10.1002/9780470920541.ch3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Affiliation(s)
- Mathew D Routh
- Molecular, Cellular and Developmental Biology Interdepartmental Graduate Program, Iowa State University, Ames, Iowa, USA
| | | | | | | | | | | |
Collapse
|
42
|
Scott NE, Parker BL, Connolly AM, Paulech J, Edwards AVG, Crossett B, Falconer L, Kolarich D, Djordjevic SP, Højrup P, Packer NH, Larsen MR, Cordwell SJ. Simultaneous glycan-peptide characterization using hydrophilic interaction chromatography and parallel fragmentation by CID, higher energy collisional dissociation, and electron transfer dissociation MS applied to the N-linked glycoproteome of Campylobacter jejuni. Mol Cell Proteomics 2011; 10:M000031-MCP201. [PMID: 20360033 PMCID: PMC3033663 DOI: 10.1074/mcp.m000031-mcp201] [Citation(s) in RCA: 237] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 03/29/2010] [Indexed: 11/06/2022] Open
Abstract
Campylobacter jejuni is a gastrointestinal pathogen that is able to modify membrane and periplasmic proteins by the N-linked addition of a 7-residue glycan at the strict attachment motif (D/E)XNX(S/T). Strategies for a comprehensive analysis of the targets of glycosylation, however, are hampered by the resistance of the glycan-peptide bond to enzymatic digestion or β-elimination and have previously concentrated on soluble glycoproteins compatible with lectin affinity and gel-based approaches. We developed strategies for enriching C. jejuni HB93-13 glycopeptides using zwitterionic hydrophilic interaction chromatography and examined novel fragmentation, including collision-induced dissociation (CID) and higher energy collisional (C-trap) dissociation (HCD) as well as CID/electron transfer dissociation (ETD) mass spectrometry. CID/HCD enabled the identification of glycan structure and peptide backbone, allowing glycopeptide identification, whereas CID/ETD enabled the elucidation of glycosylation sites by maintaining the glycan-peptide linkage. A total of 130 glycopeptides, representing 75 glycosylation sites, were identified from LC-MS/MS using zwitterionic hydrophilic interaction chromatography coupled to CID/HCD and CID/ETD. CID/HCD provided the majority of the identifications (73 sites) compared with ETD (26 sites). We also examined soluble glycoproteins by soybean agglutinin affinity and two-dimensional electrophoresis and identified a further six glycosylation sites. This study more than doubles the number of confirmed N-linked glycosylation sites in C. jejuni and is the first to utilize HCD fragmentation for glycopeptide identification with intact glycan. We also show that hydrophobic integral membrane proteins are significant targets of glycosylation in this organism. Our data demonstrate that peptide-centric approaches coupled to novel mass spectrometric fragmentation techniques may be suitable for application to eukaryotic glycoproteins for simultaneous elucidation of glycan structures and peptide sequence.
Collapse
Affiliation(s)
| | - Benjamin L. Parker
- ¶Discipline of Pathology, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | | | - Jana Paulech
- From the ‡School of Molecular and Microbial Biosciences and
| | - Alistair V. G. Edwards
- ¶Discipline of Pathology, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Ben Crossett
- From the ‡School of Molecular and Microbial Biosciences and
| | - Linda Falconer
- **New South Wales Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Camden 2570, Australia
| | - Daniel Kolarich
- ‡‡Department of Chemistry and Biomolecular Sciences, Macquarie University, 2109, Australia
| | - Steven P. Djordjevic
- **New South Wales Department of Primary Industries, Elizabeth Macarthur Agricultural Institute, Camden 2570, Australia
- §§Institute for the Biotechnology of Infectious Diseases, University of Technology, Sydney 2007, Australia, and
| | - Peter Højrup
- ¶¶Protein Research Group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5000 Odense, Denmark
| | - Nicolle H. Packer
- ‡‡Department of Chemistry and Biomolecular Sciences, Macquarie University, 2109, Australia
| | - Martin R. Larsen
- ¶¶Protein Research Group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5000 Odense, Denmark
| | - Stuart J. Cordwell
- From the ‡School of Molecular and Microbial Biosciences and
- ¶Discipline of Pathology, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
- ¶¶Protein Research Group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
43
|
Standing genetic variation in contingency loci drives the rapid adaptation of Campylobacter jejuni to a novel host. PLoS One 2011; 6:e16399. [PMID: 21283682 PMCID: PMC3025981 DOI: 10.1371/journal.pone.0016399] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 12/15/2010] [Indexed: 12/02/2022] Open
Abstract
The genome of the food-borne pathogen Campylobacter jejuni contains multiple highly mutable sites, or contingency loci. It has been suggested that standing variation at these loci is a mechanism for rapid adaptation to a novel environment, but this phenomenon has not been shown experimentally. In previous work we showed that the virulence of C. jejuni NCTC11168 increased after serial passage through a C57BL/6 IL-10-/- mouse model of campylobacteriosis. Here we sought to determine the genetic basis of this adaptation during passage. Re-sequencing of the 1.64Mb genome to 200-500X coverage allowed us to define variation in 23 contingency loci to an unprecedented depth both before and after in vivo adaptation. Mutations in the mouse-adapted C. jejuni were largely restricted to the homopolymeric tracts of thirteen contingency loci. These changes cause significant alterations in open reading frames of genes in surface structure biosynthesis loci and in genes with only putative functions. Several loci with open reading frame changes also had altered transcript abundance. The increase in specific phases of contingency loci during in vivo passage of C. jejuni, coupled with the observed virulence increase and the lack of other types of genetic changes, is the first experimental evidence that these variable regions play a significant role in C. jejuni adaptation and virulence in a novel host.
Collapse
|
44
|
Line J, Hiett K, Guard J, Seal B. Temperature affects sole carbon utilization patterns of Campylobacter coli 49941. Curr Microbiol 2010; 62:821-5. [PMID: 20981547 DOI: 10.1007/s00284-010-9785-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 06/27/2010] [Indexed: 11/26/2022]
Abstract
Campylobacter spp. are small, asaccharolytic bacteria exhibiting unique nutritional and environmental requirements. Campylobacter spp. exist as commensal organisms in some animal species, yet are estimated to be the most common causative agents of foodborne illness in humans. C. jejuni is most often associated with poultry, while C. coli are more frequently associated with swine. Temperature has been suggested to trigger potential colonization or virulence factors in C. jejuni, and recent studies have demonstrated temperature-dependent genes are important to colonization. It is possible that temperature-dependent colonization factors are in part responsible for the species-specific colonization characteristics of C. coli also. We determined utilization of 190 different sole carbon substrates by C. coli ATCC 49941 at 37 and 42°C using phenotype microarray (PM) technology. Temperature did affect amino acid utilization. L-asparagine and L-serine allowed significantly (P = 0.004) more respiration by C. coli ATCC 49941 at the lower temperature of 37°C as compared to 42°C. Conversely, L-glutamine was utilized to a significantly greater extent (P = 0.015) at the higher temperature of 42°C. Other organic substrates exhibited temperature-dependent utilization including succinate, D,L-malate, and propionate which all supported active respiration by C. coli to a significantly greater extent at 42°C. Further investigation is needed to determine the basis for the temperature-dependent utilization of substrates by Campylobacter spp. and their possible role in species-specific colonization.
Collapse
|
45
|
Abstract
Like for all microbes, the goal of every pathogen is to survive and replicate. However, to overcome the formidable defenses of their hosts, pathogens are also endowed with traits commonly associated with virulence, such as surface attachment, cell or tissue invasion, and transmission. Numerous pathogens couple their specific virulence pathways with more general adaptations, like stress resistance, by integrating dedicated regulators with global signaling networks. In particular, many of nature's most dreaded bacteria rely on nucleotide alarmones to cue metabolic disturbances and coordinate survival and virulence programs. Here we discuss how components of the stringent response contribute to the virulence of a wide variety of pathogenic bacteria.
Collapse
Affiliation(s)
- Zachary D. Dalebroux
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah L. Svensson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erin C. Gaynor
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michele S. Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
46
|
Haddad N, Marce C, Magras C, Cappelier JM. An overview of methods used to clarify pathogenesis mechanisms of Campylobacter jejuni. J Food Prot 2010; 73:786-802. [PMID: 20377972 DOI: 10.4315/0362-028x-73.4.786] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Thermotolerant campylobacters are the most frequent cause of bacterial infection of the lower intestine worldwide. The mechanism of pathogenesis of Campylobacter jejuni comprises four main stages: adhesion to intestinal cells, colonization of the digestive tract, invasion of targeted cells, and toxin production. In response to the high number of cases of human campylobacteriosis, various virulence study models are available according to the virulence stage being analyzed. The aim of this review is to compare the different study models used to look at human disease. Molecular biology tools used to identify genes or proteins involved in virulence mechanisms are also summarized. Despite high cost and limited availability, animal models are frequently used to study digestive disease, in particular to analyze the colonization stage. Eukaryotic cell cultures have been developed because of fewer restrictions on their use and the lower cost of these cultures compared with animal models, and this ex vivo approach makes it possible to mimic the bacterial cell-host interactions observed in natural disease cases. Models are complemented by molecular biology tools, especially mutagenesis and DNA microarray methods to identify putative virulence genes or proteins and permit their characterization. No current model seems to be ideal for studying the complete range of C. jejuni virulence. However, the models available deal with different aspects of the complex pathogenic mechanisms particular to this bacterium.
Collapse
Affiliation(s)
- N Haddad
- Unit INRA 1014 SECALIM, National Veterinary School of Nantes, Route de Gachet, Nantes cedex 3, France
| | | | | | | |
Collapse
|
47
|
Ang CW, Dijkstra JR, de Klerk MA, Endtz HP, van Doorn PA, Jacobs BC, Jeurissen SHM, Wagenaar JA. Host factors determine anti-GM1 response following oral challenge of chickens with Guillain-Barré syndrome derived Campylobacter jejuni strain GB11. PLoS One 2010; 5:e9820. [PMID: 20339556 PMCID: PMC2842441 DOI: 10.1371/journal.pone.0009820] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 02/19/2010] [Indexed: 11/18/2022] Open
Abstract
Background Anti-ganglioside antibodies with a pathogenic potential are present in C. jejuni-associated Guillain-Barré syndrome (GBS) patients and are probably induced by molecular mimicry. Immunization studies in rabbits and mice have demonstrated that these anti-ganglioside antibodies can be induced using purified lipo-oligosaccharides (LOS) from C. jejuni in a strong adjuvant. Methodology/Principal Findings To investigate whether natural colonization of chickens with a ganglioside-mimicking C. jejuni strain induces an anti-ganglioside response, and to investigate the diversity in anti-ganglioside response between and within genetically different chicken lines, we orally challenged chickens with different C. jejuni strains. Oral challenge of chickens with a C. jejuni strain from a GBS patient, containing a LOS that mimics ganglioside GM1, induced specific IgM and IgG anti-LOS and anti-GM1 antibodies. Inoculation of chickens with the Penner HS:3 serostrain, without a GM1-like structure, induced anti-LOS but no anti-ganglioside antibodies. We observed different patterns of anti-LOS/ganglioside response between and within the five strains of chickens. Conclusions Natural infection of chickens with C. jejuni induces anti-ganglioside antibodies. The production of antibodies is governed by both microbial and host factors.
Collapse
Affiliation(s)
- C Wim Ang
- Department of Medical Microbiology and Infection Control, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
van Putten JPM, van Alphen LB, Wösten MMSM, de Zoete MR. Molecular mechanisms of campylobacter infection. Curr Top Microbiol Immunol 2010; 337:197-229. [PMID: 19812984 DOI: 10.1007/978-3-642-01846-6_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Campylobacter jejuni is the principal bacterial foodborne pathogen. A major challenge still is to identify the virulence strategies exploited by C. jejuni. Recent genomics, proteomics, and metabolomics approaches indicate that C. jejuni displays extensive inter- and intrastrain variation. The diverse behavior enables bacterial adaptation to different environmental conditions and directs interactions with the gut mucosa. Here, we report recent progress in understanding the molecular mechanisms and functional consequences of the phenotype diversity. The results suggest that C. jejuni actively penetrates the intestinal mucus layer, secretes proteins mainly via its flagellar apparatus, is engulfed by intestinal cells, and can disrupt the integrity of the epithelial lining. C. jejuni stimulates the proinflammatory pathway and the production of a large repertoire of cytokines, chemokines, and innate effector molecules. Novel experimental infection models suggest that the activation of the innate immune response is important for the development of intestinal pathology.
Collapse
Affiliation(s)
- Jos P M van Putten
- Department of Infectious Diseases & Immunology, Utrecht University, Yalelaan 1, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
49
|
Basic concepts of microarrays and potential applications in clinical microbiology. Clin Microbiol Rev 2010; 22:611-33. [PMID: 19822891 DOI: 10.1128/cmr.00019-09] [Citation(s) in RCA: 201] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The introduction of in vitro nucleic acid amplification techniques, led by real-time PCR, into the clinical microbiology laboratory has transformed the laboratory detection of viruses and select bacterial pathogens. However, the progression of the molecular diagnostic revolution currently relies on the ability to efficiently and accurately offer multiplex detection and characterization for a variety of infectious disease pathogens. Microarray analysis has the capability to offer robust multiplex detection but has just started to enter the diagnostic microbiology laboratory. Multiple microarray platforms exist, including printed double-stranded DNA and oligonucleotide arrays, in situ-synthesized arrays, high-density bead arrays, electronic microarrays, and suspension bead arrays. One aim of this paper is to review microarray technology, highlighting technical differences between them and each platform's advantages and disadvantages. Although the use of microarrays to generate gene expression data has become routine, applications pertinent to clinical microbiology continue to rapidly expand. This review highlights uses of microarray technology that impact diagnostic microbiology, including the detection and identification of pathogens, determination of antimicrobial resistance, epidemiological strain typing, and analysis of microbial infections using host genomic expression and polymorphism profiles.
Collapse
|
50
|
Zeng X, Xu F, Lin J. Development and Evaluation of CmeC Subunit Vaccine against Campylobacter jejuni. ACTA ACUST UNITED AC 2010; 1. [PMID: 22140651 DOI: 10.4172/2157-7560.1000112] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Campylobacter jejuni is the leading bacterial cause of human enteritis in many industrialized countries. There is no commercial vaccine against C. jejuni available to date. CmeC is an essential outer membrane component of CmeABC multidrug efflux pump that plays a critical role in antibiotic resistance and in vivo colonization of C. jejuni. CmeC is prevalent in C. jejuni strains and is dramatically induced and immunogenic in vivo. In this study, we analyzed CmeC sequence homology, examined in vitro immune protection of CmeC peptide antibodies, and produced full-length recombinant CmeC (rCmeC) for evaluating immunogenicity and protective efficacy of the CmeC subunit vaccine against C. jejuni using chicken model system. Amino acid sequences of CmeC from 24 diverse C. jejuni strains were determined and subjected to alignment, which revealed that CmeC is highly conserved in C. jejuni with a identity ranging from 97.3% to 100%. CmeC peptide antibodies inhibited the function of CmeABC efflux pump and enhanced susceptibility of C. jejuni to bile salts, the natural antimicrobial present in the intestine. Two full-length rCmeC proteins with N- or C-terminal His tag were produced in E. coli; the N-terminal His-tagged rCmeC with high purity and yield was obtained by single step affinity purification. The purified rCmeC was used in two vaccination trials using a chicken model of C. jejuni infection. Stimulation of CmeC-specific serum IgG responses via oral vaccination required immunization with higher doses of rCmeC (200μg) together with 70μg of mucosal adjuvant mLT (modified E. coli heat-labile enterotoxin). Subcutaneous vaccination of chickens with rCmeC remarkably stimulated both serum IgG and IgA responses. However, CmeC-specific intestinal secretory IgA response was not significantly stimulated regardless of vaccination regimen and the rCmeC vaccination did not confer protection against C. jejuni infection. Together, these findings provide further compelling evidence that CmeC is a promising subunit vaccine candidate against C. jejuni infection. However, the CmeC vaccination regimen should be optimized to enhance CmeC-specific mucosal immune response in for protection against C. jejuni.
Collapse
Affiliation(s)
- Ximin Zeng
- Department of Animal Science, The University of Tennessee, Knoxville, TN 37996, USA
| | | | | |
Collapse
|