Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
bioRxiv ; 2024 May 06.
Article in English | MEDLINE | ID: mdl-38766229

ABSTRACT

Campylobacter infections are a leading cause of bacterial-derived gastroenteritis worldwide with particularly profound impacts on pediatric patients in low-and-middle income countries. It remains unclear how Campylobacter impacts these hosts, though it is becoming increasingly evident that it is a multifactorial process that depends on the host immune response, the gastrointestinal microbiota, various bacterial factors, and host nutritional status. Since these factors likely vary between adult and pediatric patients in different regions of the world, it is important that studies define these attributes in well characterized clinical cohorts in diverse settings. In this study, we analyzed the fecal microbiota and the metabolomic and micronutrient profiles of asymptomatic and symptomatic pediatric patients in Colombia that were either infected or uninfected with Campylobacter during a case-controlled study on acute diarrheal disease. Here, we report that the microbiome of Campylobacter- infected children only changed in their abundance of Campylobacter spp. despite the inclusion of children with or without diarrhea. In addition to increased Campylobacter, computational models were used to identify fecal metabolites that were associated with Campylobacter infection and found that glucose-6-phosphate and homovanillic acid were the strongest predictors of infection in these pediatric patients, which suggest that colonocyte metabolism are impacted during infection. Despite changes to the fecal metabolome, the concentrations of intestinal minerals and trace elements were not significantly impacted by Campylobacter infection, but were elevated in uninfected children with diarrhea. Importance: Gastrointestinal infection with pathogenic Campylobacter species has long been recognized as a significant cause of human morbidity. Recently, it has been observed that pediatric populations in low-and-middle income countries are uniquely impacted by these organisms in that infected children can be persistently colonized, develop enteric dysfunction, and exhibit reduced development and growth. While the association of Campylobacter species with these long-term effects continues to emerge, the impact of infection on the gastrointestinal environment of these children remains uncharacterized. To address this knowledge gap, our group leveraged clinical samples collected during a previous study on gastrointestinal infections in pediatric patients to examine the fecal microbiota, metabolome, and micronutrient profiles of those infected with Campylobacter species, and found that the metabolome was impacted in a way that suggests gastrointestinal cell metabolism is affected during infection, which is some of the first data indicating how gastrointestinal health in these patients may be affected.

2.
Proc Natl Acad Sci U S A ; 121(2): e2316540120, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38170751

ABSTRACT

How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using 6-wk-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni, ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within 2 to 3 d of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP, which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter (lctP) led to identification of a putative thiol-based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides better insights into the pathogenicity of C. jejuni.


Subject(s)
Campylobacter Infections , Campylobacter jejuni , Animals , Humans , Mice , Lactic Acid/metabolism , Campylobacter jejuni/genetics , Ferrets , Monocarboxylic Acid Transporters
3.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37873437

ABSTRACT

How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using six-week-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni , ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within two-three days of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP , which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter ( lctP ) led to discovery of a putative thiol based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides new insights into the pathogenicity of C. jejuni . Significance: There is a gap in knowledge about the mechanisms by which C. jejuni populations expand during infection. Using an animal model which accurately reflects human infection without the need to alter the host microbiome or the immune system prior to infection, we explored pathophysiological alterations of the gut after C. jejuni infection. Our study identified the gut metabolite L-lactate as playing an important role as a growth substrate for C. jejuni during acute infection. We identified a DNA binding protein, LctR, that binds to the lctP promoter and may repress lctP expression, resulting in decreased lactate transport under low oxygen levels. This work provides new insights about C. jejuni pathogenicity.

4.
Sci Adv ; 9(32): eade2693, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37566649

ABSTRACT

Histone modifications control numerous processes in eukaryotes, including inflammation. Some bacterial pathogens alter the activity or expression of host-derived factors, including sirtuins, to modify histones and induce responses that promote infection. In this study, we identified a deacetylase encoded by Campylobacter jejuni which has sirtuin activities and contributes to activation of human neutrophils by the pathogen. This sirtuin is secreted from the bacterium into neutrophils, where it associates with and deacetylates host histones to promote neutrophil activation and extracellular trap production. Using the murine model of campylobacteriosis, we found that a mutant of this bacterial sirtuin efficiently colonized the gastrointestinal tract but was unable to induce cytokine production, gastrointestinal inflammation, and tissue pathology. In conclusion, these results suggest that secreted bacterial sirtuins represent a previously unreported class of bacterial effector and that bacterial-mediated modification of host histones is responsible for the inflammation and pathology that occurs during campylobacteriosis.


Subject(s)
Campylobacter Infections , Campylobacter jejuni , Mice , Humans , Animals , Campylobacter jejuni/physiology , Histones , Campylobacter Infections/microbiology , Campylobacter Infections/pathology , Neutrophil Activation , Inflammation
5.
J Leukoc Biol ; 112(6): 1457-1470, 2022 12.
Article in English | MEDLINE | ID: mdl-35866361

ABSTRACT

The discovery of neutrophil subtypes has expanded what is known about neutrophil functions, yet there is still much to learn about the role of these subtypes during bacterial infection. We investigated whether Campylobacter jejuni induced differentiation of human neutrophils into the hypersegmented, CD16hi /CD62Llo subtype. In addition, we investigated whether C. jejuni-dependent differentiation of this neutrophil subtype induced cancer-promoting activities of human T cells and colonocytes, which were observed in other studies of hypersegmented, CD16hi /CD62Llo neutrophils. We found that C. jejuni causes a significant shift in human neutrophil populations to the hypersegmented, CD16hi /CD62Llo subtype and that those populations exhibit delayed apoptosis, elevated arginase-1 expression, and increased reactive oxygen species production. Furthermore, incubation of C. jejuni-infected neutrophils with human T cells resulted in decreased expression of the ζ-chain of the TCR, which was restored upon supplementation with exogenous l-arginine. In addition, incubation of C. jejuni-infected neutrophils with human colonocytes resulted in increased HIF-1α stabilization and NF-κB activation in those colonocytes, which may result in the up-regulation of protumorigenic genes.


Subject(s)
Campylobacter jejuni , Leukocyte Disorders , Humans , Neutrophils/metabolism , Leukocyte Disorders/metabolism , Signal Transduction , Up-Regulation
6.
Curr Protoc ; 1(12): e293, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34875141

ABSTRACT

Nucleases are ubiquitous in pathogens and allow bacteria to acquire nucleotide nutrients, take up foreign DNA, induce tissue damage, degrade neutrophil extracellular traps, and modulate the host inflammatory response. Furthermore, nucleases can modulate numerous bacterial virulence factors, promoting bacterial growth and disease. To understand how bacteria can produce nucleases, an unbiased approach is needed to identify these systems. Campylobacter jejuni is the leading cause of bacterial-derived gastroenteritis and utilizes numerous systems to damage host DNA. Therefore, it is imperative to identify C. jejuni nucleases to understand the molecular mechanism of both infection and pathology. Detailed protocols for a transposon insertion sequencing-based DNase agar screen, a quantitative PCR nuclease screen, and PCR transposon insertion confirmation are included in this article. © 2021 Wiley Periodicals LLC. Basic Protocol 1: DNase agar colony screen of Campylobacter jejuni transposon insertion sequencing library isolates Basic Protocol 2: Quantitative PCR nuclease screen of transposon insertion sequencing library isolates Basic Protocol 3: PCR transposon insertion confirmation.


Subject(s)
Campylobacter jejuni , Extracellular Traps , Campylobacter jejuni/genetics , Deoxyribonucleases , Polymerase Chain Reaction
7.
Curr Protoc ; 1(11): e294, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34807525

ABSTRACT

Campylobacter jejuni is the leading cause of bacterial-derived gastroenteritis worldwide, infecting 96 million individuals annually. During infection, inflammation and tissue pathology occur in the lower gastrointestinal tract, including the recruitment of leukocytes. Neutrophils are the most abundant leukocyte in humans, and recruitment is associated with bacterial infections and the development of various inflammatory disorders, including inflammatory bowel disease. Neutrophils possess three main antibacterial functions: phagocytosis and degradation of microbes, degranulation to release antimicrobial proteins, and extrusion of neutrophil extracellular traps (NETs). Because neutrophils are recruited to the site of C. jejuni infection and they are associated with damaging inflammation in other diseases, it is imperative to understand the immunopathology that occurs during C. jejuni infection and thoroughly study the neutrophil response to the pathogen. Detailed protocols for human and ferret neutrophil isolations, neutrophil gentamicin protection assay, neutrophil activation flow cytometry assay, NET induction and quantification, and neutrophil western blot analysis are included in this article. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Isolation of human and ferret neutrophils Basic Protocol 2: Neutrophil gentamicin protection assay Basic Protocol 3: Neutrophil activation flow cytometry analyses Basic Protocol 4: Neutrophil extracellular trap induction and quantification Basic Protocol 5: Western blot detection of neutrophil-derived antimicrobial proteins.


Subject(s)
Campylobacter jejuni , Extracellular Traps , Animals , Ferrets , Humans , Neutrophil Activation , Neutrophils
8.
Infect Immun ; 89(8): e0011621, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34031129

ABSTRACT

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


Subject(s)
Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Campylobacter/immunology , Disease Susceptibility/immunology , Host-Pathogen Interactions/immunology , Immunity, Cellular , Animals , Campylobacter Infections/complications , Gastrointestinal Diseases/complications , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Humans
9.
J Bacteriol ; 203(15): e0016421, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34001558

ABSTRACT

A previously identified transcriptional regulator in Campylobacter jejuni, termed HeuR, was found to positively regulate heme utilization. Additionally, transcriptomic work demonstrated that the putative operons CJJ81176_1390 to CJJ81176_1394 (CJJ81176_1390-1394) and CJJ81176_1214-1217 were upregulated in a HeuR mutant, suggesting that HeuR negatively regulates expression of these genes. Because genes within these clusters include a cystathionine ß-lyase (metC) and a methionine synthase (metE), it appeared HeuR negatively regulates C. jejuni methionine biosynthesis. To address this, we confirmed mutation of HeuR reproducibly results in metC overexpression under nutrient-replete conditions but did not affect expression of metE, while metC expression in the wild type increased to heuR mutant levels during iron limitation. We subsequently determined that both gene clusters are operonic and demonstrated the direct interaction of HeuR with the predicted promoter regions of these operons. Using DNase footprinting assays, we were able to show that HeuR specifically binds within the predicted -35 region of the CJJ81176_1390-1394 operon. As predicted based on transcriptional results, the HeuR mutant was able to grow and remain viable in a defined medium with and without methionine, but we identified significant impacts on growth and viability in metC and metE mutants. Additionally, we observed decreased adherence, invasion, and persistence of metC and metE mutants when incubated with human colonocytes, while the heuR mutant exhibited increased invasion. Taken together, these results suggest that HeuR regulates methionine biosynthesis in an iron-responsive manner and that the ability to produce methionine is an important factor for adhering to and invading the gastrointestinal tract of a susceptible host. IMPORTANCE As the leading cause of bacterium-derived gastroenteritis worldwide, Campylobacter jejuni has a significant impact on human health. Investigating colonization factors that allow C. jejuni to successfully infect a host furthers our understanding of genes and regulatory elements necessary for virulence. In this study, we have begun to characterize the role of the transcriptional regulatory protein, HeuR, on methionine biosynthesis in C. jejuni. When the ability to synthesize methionine is impaired, detrimental impacts on growth and viability are observed during growth in limited media lacking methionine and/or iron. Additionally, mutations in the methionine biosynthetic pathway result in decreased adhesion, invasion, and intracellular survival of C. jejuni when incubated with human colonocytes, indicating the importance of regulating methionine biosynthesis.


Subject(s)
Bacterial Proteins/genetics , Campylobacter Infections/microbiology , Campylobacter jejuni/enzymology , Colon/microbiology , Gene Expression Regulation, Bacterial , Lyases/genetics , Methionine/biosynthesis , Bacterial Proteins/metabolism , Campylobacter jejuni/genetics , HCT116 Cells , Humans , Lyases/metabolism , Multigene Family , Operon
10.
Metallomics ; 13(1)2021 01 16.
Article in English | MEDLINE | ID: mdl-33570133

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.


Subject(s)
Campylobacter jejuni/metabolism , Helicobacter pylori/metabolism , Homeostasis , Metals, Heavy/metabolism , Biological Transport
11.
Front Microbiol ; 11: 571064, 2020.
Article in English | MEDLINE | ID: mdl-33224113

ABSTRACT

As a leading cause of bacterial-derived gastroenteritis worldwide, Campylobacter jejuni has a significant impact on human health in both the developed and developing worlds. Despite its prevalence as a human pathogen, the source of these infections remains poorly understood due to the mutation frequency of the organism and past limitations of whole genome analysis. Recent advances in both whole genome sequencing and computational methods have allowed for the high-resolution analysis of intraspecies diversity, leading multiple groups to postulate that these approaches may be used to identify the sources of Campylobacter jejuni infection. To address this hypothesis, our group conducted a regionally and temporally restricted sampling of agricultural and environmental Campylobacter sources and compared isolated C. jejuni genomes to those that caused human infections in the same region during the same time period. Through a network analysis comparing genomes from various sources, we found that human C. jejuni isolates clustered with those isolated from cattle and chickens, indicating these as potential sources of human infection in the region.

12.
Cell Microbiol ; 22(8): e13210, 2020 08.
Article in English | MEDLINE | ID: mdl-32329205

ABSTRACT

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


Subject(s)
Campylobacter jejuni/immunology , Campylobacter jejuni/physiology , Extracellular Traps/immunology , Extracellular Traps/microbiology , Feces/chemistry , Host Microbial Interactions/immunology , Neutrophils/immunology , Animals , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Cells, Cultured , Colon/cytology , Colon/microbiology , Colon/pathology , Ferrets , Humans , Inflammation , Leukocyte Elastase/metabolism , Male , Neutrophils/chemistry , Neutrophils/microbiology , Phagocytosis
13.
Gastroenterol Res Pract ; 2020: 2868373, 2020.
Article in English | MEDLINE | ID: mdl-32184815

ABSTRACT

Calgranulin proteins are an important class of molecules involved in innate immunity. These members of the S100 class of the EF-hand family of calcium-binding proteins have numerous cellular and antimicrobial functions. One protein in particular, S100A12 (also called EN-RAGE or calgranulin C), is highly abundant in neutrophils during acute inflammation and has been implicated in immune regulation. Structure-function analyses reveal that S100A12 has the capacity to bind calcium, zinc, and copper, processes that contribute to nutritional immunity against invading microbial pathogens. S100A12 is a ligand for the receptor for advanced glycation end products (RAGE), toll-like receptor 4 (TLR4), and CD36, which promote cellular and immunological pathways to alter inflammation. We conducted a scoping review of the existing literature to define what is known about the association of S100A12 with digestive disease and health. Results suggest that S100A12 is implicated in gastroenteritis, necrotizing enterocolitis, gastritis, gastric cancer, Crohn's disease, irritable bowel syndrome, inflammatory bowel disease, and digestive tract cancers. Together, these results reveal S100A12 is an important molecule broadly associated with the pathogenesis of digestive diseases.

14.
mBio ; 10(4)2019 08 06.
Article in English | MEDLINE | ID: mdl-31387912

ABSTRACT

The stator units of the flagellum supply power to the flagellar motor via ion transport across the cytoplasmic membrane and generate torque on the rotor for rotation. Flagellar motors across bacterial species have evolved adaptations that impact and enhance stator function to meet the demands of each species, including producing stator units using different fuel types or various stator units for different motility modalities. Campylobacter jejuni produces one of the most complex and powerful flagellar motors by positioning 17 stator units at a greater radial distance than in most other bacteria to increase power and torque for high velocity of motility. We report another evolutionary adaptation impacting flagellar stators by identifying FlgX as a chaperone for C. jejuni stator units to ensure sufficient power and torque for flagellar rotation and motility. We discovered that FlgX maintains MotA and MotB stator protein integrity likely through a direct interaction with MotA that prevents their degradation. Suppressor analysis suggested that the physiology of C. jejuni drives the requirement for FlgX to protect stator units from proteolysis by the FtsH protease complex. C. jejuni ΔflgX was strongly attenuated for colonization of the natural avian host, but colonization capacity was greatly restored by a single mutation in MotA. These findings suggest that the likely sole function of FlgX is to preserve stator unit integrity for the motility required for host interactions. Our findings demonstrate another evolved adaptation in motile bacteria to ensure the equipment of the flagellar motor with sufficient power to generate torque for motility.IMPORTANCE The bacterial flagellum is a reversible rotating motor powered by ion transport through stator units, which also exert torque on the rotor component to turn the flagellum for motility. Species-specific adaptations to flagellar motors impact stator function to meet the demands of each species to sufficiently power flagellar rotation. We identified another evolutionary adaptation by discovering that FlgX of Campylobacter jejuni preserves the integrity of stator units by functioning as a chaperone to protect stator proteins from degradation by the FtsH protease complex due to the physiology of the bacterium. FlgX is required to maintain a level of stator units sufficient to power the naturally high-torque flagellar motor of C. jejuni for motility in intestinal mucosal layers to colonize hosts. Our work continues to identify an increasing number of adaptations to flagellar motors across bacterial species that provide the mechanics necessary for producing an effective rotating nanomachine for motility.


Subject(s)
Bacterial Proteins/metabolism , Campylobacter jejuni/metabolism , Flagella/metabolism , Molecular Chaperones/metabolism , Bacterial Proteins/genetics , Campylobacter jejuni/genetics , Flagella/genetics , Molecular Chaperones/genetics , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism
15.
Article in English | MEDLINE | ID: mdl-30984629

ABSTRACT

Iron is a transition metal utilized by nearly all forms of life for essential cellular processes, such as DNA synthesis and cellular respiration. During infection by bacterial pathogens, the host utilizes various strategies to sequester iron in a process termed, nutritional immunity. To circumvent these defenses, Gram-negative pathogens have evolved numerous mechanisms to obtain iron from heme. In this review we outline the systems that exist in several Gram-negative pathogens that are associated with heme transport and utilization, beginning with hemolysis and concluding with heme degradation. In addition, Gram-negative pathogens must also closely regulate the intracellular concentrations of iron and heme, since high levels of iron can lead to the generation of toxic reactive oxygen species. As such, we also provide several examples of regulatory pathways that control heme utilization, showing that co-regulation with other cellular processes is complex and often not completely understood.


Subject(s)
Gram-Negative Bacteria/metabolism , Heme/metabolism , Biological Transport , Biotransformation , Gene Expression Regulation, Bacterial , Hemolysis , Iron/metabolism , Metabolic Networks and Pathways
16.
Infect Immun ; 86(6)2018 06.
Article in English | MEDLINE | ID: mdl-29610259

ABSTRACT

Campylobacter jejuni is a leading cause of bacterially derived gastroenteritis worldwide. Campylobacter is most commonly acquired through the consumption of undercooked poultry meat or through drinking contaminated water. Following ingestion, Campylobacter adheres to the intestinal epithelium and mucus layer, causing toxin-mediated inflammation and inhibition of fluid reabsorption. Currently, the human response to infection is relatively unknown, and animal hosts that model these responses are rare. As such, we examined patient fecal samples for the accumulation of the neutrophil protein calgranulin C during infection with Campylobacter jejuni In response to infection, calgranulin C was significantly increased in the feces of humans. To determine whether calgranulin C accumulation occurs in an animal model, we examined disease in ferrets. Ferrets were effectively infected by C. jejuni, with peak fecal loads observed at day 3 postinfection and full resolution by day 12. Serum levels of interleukin-10 (IL-10) and tumor necrosis factor alpha (TNF-α) significantly increased in response to infection, which resulted in leukocyte trafficking to the colon. As a result, calgranulin C increased in the feces of ferrets at the time when C. jejuni loads decreased. Further, the addition of purified calgranulin C to C. jejuni cultures was found to inhibit growth in a zinc-dependent manner. These results suggest that upon infection with C. jejuni, leukocytes trafficked to the intestine release calgranulin C as a mechanism for inhibiting C. jejuni growth.


Subject(s)
Campylobacter jejuni/growth & development , S100A12 Protein/metabolism , Zinc/metabolism , Animals , Campylobacter jejuni/drug effects , Colon/cytology , Colon/microbiology , Female , Ferrets , Humans , Leukocytes , Male , Microbial Sensitivity Tests , S100A12 Protein/genetics
17.
Front Microbiol ; 8: 487, 2017.
Article in English | MEDLINE | ID: mdl-28386253

ABSTRACT

Campylobacter jejuni is the leading cause of bacteria-derived gastroenteritis worldwide. In the developed world, Campylobacter is usually acquired by consuming under-cooked poultry, while in the developing world it is often obtained through drinking contaminated water. Once consumed, the bacteria adhere to the intestinal epithelium or mucus layer, causing toxin-mediated inhibition of fluid reabsorption from the intestine and invasion-induced inflammation and diarrhea. Traditionally, severe or prolonged cases of campylobacteriosis have been treated with antibiotics; however, overuse of these antibiotics has led to the emergence of antibiotic-resistant strains. As the incidence of antibiotic resistance, emergence of post-infectious diseases, and economic burden associated with Campylobacter increases, it is becoming urgent that novel treatments are developed to reduce Campylobacter numbers in commercial poultry and campylobacteriosis in humans. The purpose of this review is to provide the current status of present and proposed treatments to combat Campylobacter infection in humans and colonization in animal reservoirs. These treatments include anti-Campylobacter compounds, probiotics, bacteriophage, vaccines, and anti-Campylobacter bacteriocins, all of which may be successful at reducing the incidence of campylobacteriosis in humans and/or colonization loads in poultry. In addition to reviewing treatments, we will also address several proposed targets that may be used in future development of novel anti-Campylobacter treatments.

18.
Methods Mol Biol ; 1512: 257-272, 2017.
Article in English | MEDLINE | ID: mdl-27885613

ABSTRACT

The advent of next-generation sequencing technology has enabled experimental approaches to characterize large, complex populations of DNA molecules with high resolution. Included among these are methods to assess populations of transposon insertion libraries for the fitness cost of any particular mutant allele after applying selection to a population. These approaches have proven invaluable for identifying genetic factors that influence survival of bacterial pathogens within different environments, including animal hosts. One such method, termed insertion-site sequencing (INSeq), was designed to generate a 16 bp fragment of transposon-flanking genomic DNA captured during the protocol, which then serves as the substrate for massively parallel sequencing. Here we describe the generation of a transposon mutant library of Campylobacter jejuni amenable to INSeq and its use in identifying colonization determinants in a day-of-hatch chicken colonization model.


Subject(s)
Bacterial Proteins/genetics , Campylobacter jejuni/pathogenicity , DNA Transposable Elements , DNA, Bacterial/genetics , Gene Expression Regulation, Bacterial , Gene Library , Repressor Proteins/genetics , Sequence Analysis, DNA/methods , Alleles , Animals , Bacterial Proteins/metabolism , Base Sequence , Campylobacter Infections/microbiology , Campylobacter Infections/pathology , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Chickens , Genetic Fitness , High-Throughput Nucleotide Sequencing , Mutagenesis, Insertional , Repressor Proteins/metabolism , Selection, Genetic , Virulence
19.
mBio ; 7(6)2016 11 15.
Article in English | MEDLINE | ID: mdl-27935836

ABSTRACT

Campylobacter jejuni is a leading cause of bacterially derived gastroenteritis. A previous mutant screen demonstrated that the heme uptake system (Chu) is required for full colonization of the chicken gastrointestinal tract. Subsequent work identified a PAS domain-containing regulator, termed HeuR, as being required for chicken colonization. Here we confirm that both the heme uptake system and HeuR are required for full chicken gastrointestinal tract colonization, with the heuR mutant being particularly affected during competition with wild-type C. jejuni Transcriptomic analysis identified the chu genes-and those encoding other iron uptake systems-as regulatory targets of HeuR. Purified HeuR bound the chuZA promoter region in electrophoretic mobility shift assays. Consistent with a role for HeuR in chu expression, heuR mutants were unable to efficiently use heme as a source of iron under iron-limiting conditions, and mutants exhibited decreased levels of cell-associated iron by mass spectrometry. Finally, we demonstrate that an heuR mutant of C. jejuni is resistant to hydrogen peroxide and that this resistance correlates to elevated levels of catalase activity. These results indicate that HeuR directly and positively regulates iron acquisition from heme and negatively impacts catalase activity by an as yet unidentified mechanism in C. jejuni IMPORTANCE: Annually, Campylobacter jejuni causes millions of gastrointestinal infections in the United States, due primarily to its ability to reside within the gastrointestinal tracts of poultry, where it can be released during processing and contaminate meat. In the developing world, humans are often infected by consuming contaminated water or by direct contact with livestock. Following consumption of contaminated food or water, humans develop disease that is characterized by mild to severe diarrhea. There is a need to understand both colonization of chickens, to make food safer, and colonization of humans, to better understand disease. Here we demonstrate that to efficiently colonize a host, C. jejuni requires iron from heme, which is regulated by the protein HeuR. Understanding how HeuR functions, we can develop ways to inhibit its function and reduce iron acquisition during colonization, potentially reducing C. jejuni in the avian host, which would make food safer, or limiting human colonization.


Subject(s)
Bacterial Proteins/metabolism , Campylobacter jejuni/metabolism , Gene Expression Regulation, Bacterial , Heme/metabolism , Iron/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Campylobacter jejuni/enzymology , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Catalase/metabolism , Chickens , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Gene Expression Profiling , Heme/genetics , Humans , Hydrogen Peroxide/metabolism , Mutation
20.
J Biol Chem ; 291(43): 22686-22702, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27474744

ABSTRACT

Campylobacter jejuni is a leading cause of bacterial gastroenteritis in the developed world. Despite its prevalence, its mechanisms of pathogenesis are poorly understood. Peptidoglycan (PG) is important for helical shape, colonization, and host-pathogen interactions in C. jejuni Therefore, changes in PG greatly impact the physiology of this organism. O-acetylation of peptidoglycan (OAP) is a bacterial phenomenon proposed to be important for proper cell growth, characterized by acetylation of the C6 hydroxyl group of N-acetylmuramic acid in the PG glycan backbone. The OAP gene cluster consists of a PG O-acetyltransferase A (patA) for translocation of acetate into the periplasm, a PG O-acetyltransferase B (patB) for O-acetylation, and an O-acetylpeptidoglycan esterase (ape1) for de-O-acetylation. In this study, reduced OAP in ΔpatA and ΔpatB had minimal impact on C. jejuni growth and fitness under the conditions tested. However, accumulation of OAP in Δape1 resulted in marked differences in PG biochemistry, including O-acetylation, anhydromuropeptide levels, and changes not expected to result directly from Ape1 activity. This suggests that OAP may be a form of substrate level regulation in PG biosynthesis. Ape1 acetylesterase activity was confirmed in vitro using p-nitrophenyl acetate and O-acetylated PG as substrates. In addition, Δape1 exhibited defects in pathogenesis-associated phenotypes, including cell shape, motility, biofilm formation, cell surface hydrophobicity, and sodium deoxycholate sensitivity. Δape1 was also impaired for chick colonization and adhesion, invasion, intracellular survival, and induction of IL-8 production in INT407 cells in vitro The importance of Ape1 in C. jejuni biology makes it a good candidate as an antimicrobial target.


Subject(s)
Campylobacter jejuni/metabolism , Campylobacter jejuni/pathogenicity , Cell Wall/metabolism , Peptidoglycan/metabolism , Virulence Factors/metabolism , Acetylation , Acetyltransferases/genetics , Acetyltransferases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Campylobacter jejuni/genetics , Cell Wall/genetics , Peptidoglycan/genetics , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL