Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
Nat Commun ; 15(1): 4462, 2024 May 25.
Article En | MEDLINE | ID: mdl-38796512

Virulence and metabolism are often interlinked to control the expression of essential colonisation factors in response to host-associated signals. Here, we identified an uncharacterised transporter of the dietary monosaccharide ʟ-arabinose that is widely encoded by the zoonotic pathogen enterohaemorrhagic Escherichia coli (EHEC), required for full competitive fitness in the mouse gut and highly expressed during human infection. Discovery of this transporter suggested that EHEC strains have an enhanced ability to scavenge ʟ-arabinose and therefore prompted us to investigate the impact of this nutrient on pathogenesis. Accordingly, we discovered that ʟ-arabinose enhances expression of the EHEC type 3 secretion system, increasing its ability to colonise host cells, and that the underlying mechanism is dependent on products of its catabolism rather than the sensing of ʟ-arabinose as a signal. Furthermore, using the murine pathogen Citrobacter rodentium, we show that ʟ-arabinose metabolism provides a fitness benefit during infection via virulence factor regulation, as opposed to supporting pathogen growth. Finally, we show that this mechanism is not restricted to ʟ-arabinose and extends to other pentose sugars with a similar metabolic fate. This work highlights the importance integrating central metabolism with virulence regulation in order to maximise competitive fitness of enteric pathogens within the host-niche.


Arabinose , Citrobacter rodentium , Enterohemorrhagic Escherichia coli , Arabinose/metabolism , Animals , Mice , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/metabolism , Citrobacter rodentium/genetics , Humans , Virulence , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/metabolism , Enterohemorrhagic Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Virulence Factors/metabolism , Virulence Factors/genetics , Enterobacteriaceae Infections/microbiology , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Escherichia coli Infections/microbiology , Female
2.
Nature ; 628(8006): 180-185, 2024 Apr.
Article En | MEDLINE | ID: mdl-38480886

The gut microbiome has major roles in modulating host physiology. One such function is colonization resistance, or the ability of the microbial collective to protect the host against enteric pathogens1-3, including enterohaemorrhagic Escherichia coli (EHEC) serotype O157:H7, an attaching and effacing (AE) food-borne pathogen that causes severe gastroenteritis, enterocolitis, bloody diarrhea and acute renal failure4,5 (haemolytic uremic syndrome). Although gut microorganisms can provide colonization resistance by outcompeting some pathogens or modulating host defence provided by the gut barrier and intestinal immune cells6,7, this phenomenon remains poorly understood. Here, we show that activation of the neurotransmitter receptor dopamine receptor D2 (DRD2) in the intestinal epithelium by gut microbial metabolites produced upon dietary supplementation with the essential amino acid L-tryptophan protects the host against Citrobacter rodentium, a mouse AE pathogen that is widely used as a model for EHEC infection8,9. We further find that DRD2 activation by these tryptophan-derived metabolites decreases expression of a host actin regulatory protein involved in C. rodentium and EHEC attachment to the gut epithelium via formation of actin pedestals. Our results reveal a noncanonical colonization resistance pathway against AE pathogens that features an unconventional role for DRD2 outside the nervous system in controlling actin cytoskeletal organization in the gut epithelium. Our findings may inspire prophylactic and therapeutic approaches targeting DRD2 with dietary or pharmacological interventions to improve gut health and treat gastrointestinal infections, which afflict millions globally.


Citrobacter rodentium , Intestinal Mucosa , Receptors, Dopamine D2 , Tryptophan , Animals , Female , Humans , Male , Mice , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Bacterial Load/drug effects , Citrobacter rodentium/growth & development , Citrobacter rodentium/metabolism , Citrobacter rodentium/pathogenicity , Dietary Supplements , Disease Models, Animal , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/prevention & control , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli O157/pathogenicity , Escherichia coli O157/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Receptors, Dopamine D2/metabolism , Tryptophan/administration & dosage , Tryptophan/metabolism , Tryptophan/pharmacology
3.
Biochem Biophys Res Commun ; 669: 103-112, 2023 08 20.
Article En | MEDLINE | ID: mdl-37269592

Tumor necrosis factor receptor-associated factor 6 (TRAF6) plays a pivotal role in the induction of inflammatory responses not only in innate immune cells but also in non-immune cells, leading to the activation of adaptive immunity. Signal transduction mediated by TRAF6, along with its upstream molecule MyD88 in intestinal epithelial cells (IECs) is crucial for the maintenance of mucosal homeostasis following inflammatory insult. The IEC-specific TRAF6-deficient (TRAF6ΔIEC) and MyD88-deficient (MyD88ΔIEC) mice exhibit increased susceptibility to DSS-induced colitis, emphasizing the critical role of this pathway. Moreover, MyD88 also plays a protective role in Citrobacter rodentium (C. rodentium) infection-induced colitis. However, its pathological role of TRAF6 in infectious colitis remains unclear. To investigate the site-specific roles of TRAF6 in response to enteric bacterial pathogens, we infected TRAF6ΔIEC and dendritic cell (DC)-specific TRAF6-deficient (TRAF6ΔDC) mice with C. rodentium and found that the pathology of infectious colitis was exacerbated with significantly decreased survival rates in TRAF6ΔDC mice, but not in TRAF6ΔIEC mice, compared to those in control mice. TRAF6ΔDC mice showed increased bacterial burdens, marked disruption of epithelial and mucosal structures with increased infiltration of neutrophils and macrophages, and elevated cytokine levels in the colon at the late stages of infection. The frequencies of IFN-γ producing Th1 cells and IL-17A producing Th17 cells in the colonic lamina propria were significantly reduced in TRAF6ΔDC mice. Finally, we demonstrated that TRAF6-deficient DCs failed to produce IL-12 and IL-23 in response to C. rodentium stimulation, and to induce both Th1 and Th17 cells in vitro. Thus, TRAF6 signaling in DCs, but not in IECs, protects against colitis induced by C. rodentium infection by producing IL-12 and IL-23 that induce Th1 and Th17 responses in the gut.


Citrobacter rodentium , Colitis , Animals , Mice , Citrobacter rodentium/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Th17 Cells , Colitis/pathology , Signal Transduction , Intestinal Mucosa/metabolism , Colon/pathology , Adaptor Proteins, Signal Transducing/metabolism , Dendritic Cells/metabolism , Interleukin-12/metabolism , Interleukin-23/metabolism , Mice, Inbred C57BL , Th1 Cells/metabolism
4.
Cell Death Dis ; 14(4): 282, 2023 04 21.
Article En | MEDLINE | ID: mdl-37080966

Citrobacter rodentium is an enteropathogen that causes intestinal inflammatory responses in mice reminiscent of the pathology provoked by enteropathogenic and enterohemorrhagic Escherichia coli infections in humans. C. rodentium expresses various virulence factors that target specific signaling proteins involved in executing apoptotic, necroptotic and pyroptotic cell death, suggesting that each of these distinct cell death modes performs essential host defense functions that the pathogen aims to disturb. However, the relative contributions of apoptosis, necroptosis and pyroptosis in protecting the host against C. rodentium have not been elucidated. Here we used mice with single or combined deficiencies in essential signaling proteins controlling apoptotic, necroptotic or pyroptotic cell death to reveal the roles of these cell death modes in host defense against C. rodentium. Gastrointestinal C. rodentium infections in mice lacking GSDMD and/or MLKL showed that both pyroptosis and necroptosis were dispensable for pathogen clearance. In contrast, while RIPK3-deficient mice showed normal C. rodentium clearance, mice with combined caspase-8 and RIPK3 deficiencies failed to clear intestinal pathogen loads. Although this demonstrated a crucial role for caspase-8 signaling in establishing intestinal host defense, Casp8-/-Ripk3-/- mice remained capable of preventing systemic pathogen persistence. This systemic host defense relied on inflammasome signaling, as Casp8-/-Ripk3-/- mice with combined caspase-1 and -11 deletion succumbed to C. rodentium infection. Interestingly, although it is known that C. rodentium can activate the non-canonical caspase-11 inflammasome, selectively disabling canonical inflammasome signaling by single caspase-1 deletion sufficed to render Casp8-/-Ripk3-/- mice vulnerable to C. rodentium-induced lethality. Moreover, Casp8-/-Ripk3-/- mice lacking GSDMD survived a C. rodentium infection, suggesting that pyroptosis was not crucial for the protective functions of canonical inflammasomes in these mice. Taken together, our mouse genetic experiments revealed an essential cooperation between caspase-8 signaling and GSDMD-independent canonical inflammasome signaling to establish intestinal and systemic host defense against gastrointestinal C. rodentium infection.


Citrobacter rodentium , Inflammasomes , Animals , Humans , Mice , Caspase 1/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspases/metabolism , Citrobacter rodentium/metabolism , Gasdermins , Inflammasomes/metabolism , Mice, Inbred C57BL
5.
J Microbiol Biotechnol ; 33(1): 35-42, 2023 Jan 28.
Article En | MEDLINE | ID: mdl-36457188

This study aimed to identify the therapeutic ability of a novel toll-like receptor (TLR) 5 agonist, KMRC011, on ulcerative colitis induced by Citrobacter rodentium and dextran sulfate sodium in a C57BL/6N mouse model. Ulcerative colitis was induced in the mice by the oral administration of 1% dextran sulfate sodium in sterile drinking water for seven days ad libitum, followed by C. rodentium infection on the seventh day by intra-gastric administration (DSS-CT group). KMRC011 was administered intramuscularly at both 24 h and 15 min before (Treatment 1 group), and at both 15 min and 24 h after (Treatment 2 group) the C. rodentium infection. The length of the large intestine and histopathological counts were significantly greater and mucosal thickness was significantly thinner in the Treatment 1 group compared to the DSS-CT and Treatment 2 groups. Il-6 and Il-10 mRNA expression levels were upregulated, while Ifn-γ and Tnf-α mRNA expression levels were significantly downregulated in the Treatment 1 group, compared to the DSS-CT group. NF-κB p65 expression level was elevated due to ulcerative colitis in the DSS-CT group, but was significantly downregulated in the Treatment 1 group. Overall, KMRC011 showed protective effects against murine colitis by inhibiting NF-κB signaling.


Colitis, Ulcerative , Colitis , Mice , Animals , Colitis, Ulcerative/chemically induced , NF-kappa B/metabolism , Citrobacter rodentium/metabolism , Toll-Like Receptor 5/metabolism , Toll-Like Receptor 5/therapeutic use , Dextran Sulfate/adverse effects , Colon/pathology , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , RNA, Messenger/metabolism , Disease Models, Animal
6.
Curr Opin Microbiol ; 65: 183-190, 2022 02.
Article En | MEDLINE | ID: mdl-34929548

The formation of attaching and effacing (A/E) lesions on intestinal epithelium, combined with Shiga toxin production, are hallmarks of enterohemorrhagic Escherichia coli (EHEC) infection that can lead to lethal hemolytic uremic syndrome. Although an animal infection model that fully recapitulates human disease remains elusive, mice orally infected with Citrobacter rodentium(ϕStx2dact), a natural murine pathogen lysogenized with an EHEC-derived Shiga toxin 2-producing bacteriophage, develop intestinal A/E lesions and toxin-dependent systemic disease. This model has facilitated investigation of how: (A) phage gene expression and prophage induction contribute to disease and are potentially triggered by antibiotic treatment; (B) virulence gene expression is altered by microbiota and the colonic metabolomic milieu; and (C) innate immune signaling is affected by Stx. Thus, the model provides a unique tool for accessing diverse aspects of EHEC pathogenesis.


Bacteriophages , Enterohemorrhagic Escherichia coli , Escherichia coli Infections , Hemolytic-Uremic Syndrome , Animals , Bacteriophages/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Disease Models, Animal , Enterohemorrhagic Escherichia coli/metabolism , Female , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/pathology , Humans , Intestinal Mucosa/metabolism , Male , Mice
7.
Toxins (Basel) ; 13(8)2021 07 29.
Article En | MEDLINE | ID: mdl-34437405

Stx2 is the major virulence factor of EHEC and is associated with an increased risk for HUS in infected patients. The conditions influencing its expression in the intestinal tract are largely unknown. For optimal management and treatment of infected patients, the identification of environmental conditions modulating Stx2 levels in the human gut is of central importance. In this study, we established a set of chromosomal stx2 reporter assays. One system is based on superfolder GFP (sfGFP) using a T7 polymerase/T7 promoter-based amplification loop. This reporter can be used to analyze stx2 expression at the single-cell level using FACSs and fluorescence microscopy. The other system is based on the cytosolic release of the Gaussia princeps luciferase (gluc). This latter reporter proves to be a highly sensitive and scalable reporter assay that can be used to quantify reporter protein in the culture supernatant. We envision that this new set of reporter tools will be highly useful to comprehensively analyze the influence of environmental and host factors, including drugs, small metabolites and the microbiota, on Stx2 release and thereby serve the identification of risk factors and new therapies in Stx-mediated pathologies.


Biological Assay , Shiga Toxin 2/genetics , Animals , Chlorocebus aethiops , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , Vero Cells
8.
Curr Opin Microbiol ; 63: 238-243, 2021 10.
Article En | MEDLINE | ID: mdl-34450388

To establish infection, enteric pathogens integrate environmental cues to navigate the gastrointestinal tract and precisely control expression of virulence determinants. Emerging data indicate that post-transcriptional and post-translational gene regulation plays a key role in virulence regulation and pathogen adaptation to the host environment. Here, we highlight recent studies that reveal how physiologically relevant signals initiate post-transcriptional and post-translational regulatory circuits and the impact on virulence gene expression in the attaching and effacing pathogens, enteropathogenic Escherichia coli, enterohemorrhagic E. coli O157:H7, and Citrobacter rodentium.


Enterohemorrhagic Escherichia coli , Escherichia coli Proteins , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Cues , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression , Gene Expression Regulation, Bacterial
9.
Methods Mol Biol ; 2291: 381-397, 2021.
Article En | MEDLINE | ID: mdl-33704765

Shiga toxin-producing E. coli (STEC) is a common foodborne pathogen in developed countries. STEC generates "attaching and effacing" (AE) lesions on colonic epithelium, characterized by effacement of microvilli and the formation of actin "pedestals" beneath intimately attached bacteria. In addition, STEC are lysogenized with a phage that, upon induction, can produce potent Shiga toxins (Stx), potentially leading to both hemorrhagic colitis and hemolytic uremic syndrome. Investigation of the pathogenesis of this disease has been challenging because STEC does not readily colonize conventional mice.Citrobacter rodentium (CR) is a related mouse pathogen that also generates AE lesions. Whereas CR does not produce Stx, a murine model for STEC utilizes CR lysogenized with an E. coli-derived Stx phage, generating CR(Φstx), which both colonizes conventional mice and readily gives rise to systemic disease. We present here key methods for the use of CR(Φstx) infection as a highly predictable murine model for infection and disease by STEC. Importantly, we detail CR(Φstx) inoculation by feeding, determination of pathogen colonization, production of phage and toxin, and assessment of intestinal and renal pathology. These methods provide a framework for studying STEC-mediated systemic disease that may aid in the development of efficacious therapeutics.


Bacteriophages , Citrobacter rodentium , Colitis , Gastrointestinal Hemorrhage , Hemolytic-Uremic Syndrome , Intestinal Mucosa , Lysogeny , Shiga Toxins , Shiga-Toxigenic Escherichia coli , Animals , Bacteriophages/genetics , Bacteriophages/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/virology , Colitis/genetics , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Gastrointestinal Hemorrhage/genetics , Gastrointestinal Hemorrhage/metabolism , Gastrointestinal Hemorrhage/microbiology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Shiga Toxins/biosynthesis , Shiga Toxins/genetics
10.
Clin Exp Immunol ; 204(3): 361-372, 2021 06.
Article En | MEDLINE | ID: mdl-33662140

Dedicator of cytokinesis 2 (Dock2), an atypical guanine exchange factor, is specifically expressed on immune cells and mediates cell adhesion and migration by activating Rac and regulates actin cytoskeleton remodeling. It plays a crucial role in the migration, formation of immune synapses, cell proliferation, activation of T and B lymphocytes and chemotaxis of pDCs and neutrophils. However, in-vivo physiological functions of Dock2 have been relatively seldom studied. Our previous studies showed that Dock2-/- mice were highly susceptible to colitis induced by Citrobacter rodentium infection, and in early infection, Dock2-/- mice had defects in macrophage migration. However, the specific roles of Dock2 in the migration and functions of macrophages are not clear. In this study, we found that the expression of chemokines such as chemokine (C-C motif) ligand (CCL)4 and CCL5 and chemokine receptors such as chemokine (C-C motif) receptor (CCR)4 and CCR5 in bone marrow-derived macrophages (BMDM) of Dock2-/- mice decreased after infection, which were supported by the in-vivo infection experimental results; the Transwell experiment results showed that Dock2-/- BMDM had a defect in chemotaxis. The bacterial phagocytic and bactericidal experiment results also showed that Dock2-/- BMDM had the defects of bacterial phagocytosis and killing. Furthermore, the adoptive transfer of wild-type BMDM alleviated the susceptibility of Dock2-/- mice to C. rodentium infection. Our results show that Dock2 affects migration and phagocytic and bactericidal ability of macrophages by regulating the expression of chemokines, chemokine receptors and their responses to chemokine stimulation, thus playing an essential role in the host defense against enteric bacterial infection.


Cell Movement/physiology , Citrobacter rodentium/metabolism , Enterobacteriaceae Infections/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Macrophages/metabolism , Animals , B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Cell Adhesion/physiology , Chemokines/metabolism , Chemotaxis/physiology , Dendritic Cells/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/metabolism
11.
Dig Dis Sci ; 66(1): 88-104, 2021 01.
Article En | MEDLINE | ID: mdl-32034605

BACKGROUND: Environmental enteropathy (EE) is associated with stunting, impairment of responses to oral vaccines, and other adverse health consequences in young children throughout the developing world. EE is characterized by chronic low-grade intestinal inflammation and disrupted epithelial barrier integrity, partly resulting from dysregulation of tight junction proteins, observed in other enteropathies such as celiac disease. During EE, this dysregulation of tight junction expression amplifies translocation of pathogenic bacteria across the intestinal mucosa. AIMS: The aim was to determine whether enteropathogen-mediated epithelial barrier failure can be ameliorated using contra-pathogenicity therapies. METHODS: Intestinal epithelial barrier damage was assessed in Caco-2 cells incubated with three important enteropathogens identified in EE patients: Enteropathogenic Escherichia coli (EPEC), Citrobacter rodentium (C. rodentium), and Cryptosporidium parvum (C. parvum). Potential therapeutic molecules were tested to detect effects on transepithelial resistance (TER), bacterial translocation (BT), claudin-4 expression, and regulation of the inflammatory cytokine response. RESULTS: All three enteropathogens compared to uninfected cells, reduced TER (EPEC; p < 0.0001, C. rodentium; p < 0.0001, C. parvum; p < 0.0007), reduced claudin-4 expression, and permitted BT (EPEC; p < 0.0001, C. rodentium; p < 0.0001, C. parvum; p < 0.0003) through the monolayer. Zinc, colostrum, epidermal growth factor, trefoil factor 3, resistin-like molecule-ß, hydrocortisone, and the myosin light chain kinase inhibitor ML7 (Hexahydro-1-[(5-iodo-1-naphthalenyl)sulfonyl]-1H-1,4-diazepine hydrochloride); ML7) improved TER (up to 70%) and decreased BT (as much as 96%). Only zinc demonstrated modest antimicrobial activity. CONCLUSION: The enteropathogens impaired intestinal-epithelial barrier integrity with dysregulation of claudin-4 and increased bacterial translocation. Enteropathogen-mediated damage was reduced using contra-pathogenicity agents which mitigated the effects of pathogens without direct antimicrobial activity.


Bacterial Translocation/physiology , Citrobacter rodentium/metabolism , Cryptosporidium parvum/metabolism , Enteropathogenic Escherichia coli/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Bacterial Translocation/drug effects , Caco-2 Cells , Citrobacter rodentium/drug effects , Cryptosporidium parvum/drug effects , Enteropathogenic Escherichia coli/drug effects , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/therapeutic use , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Humans , Hydrocortisone/pharmacology , Hydrocortisone/therapeutic use , Intestinal Diseases/drug therapy , Intestinal Diseases/metabolism , Intestinal Diseases/microbiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/physiology
12.
Cell Host Microbe ; 28(6): 789-797.e5, 2020 12 09.
Article En | MEDLINE | ID: mdl-33301718

The colonic microbiota exhibits cross-sectional heterogeneity, but the mechanisms that govern its spatial organization remain incompletely understood. Here we used Citrobacter rodentium, a pathogen that colonizes the colonic surface, to identify microbial traits that license growth and survival in this spatial niche. Previous work showed that during colonic crypt hyperplasia, type III secretion system (T3SS)-mediated intimate epithelial attachment provides C. rodentium with oxygen for aerobic respiration. However, we find that prior to the development of colonic crypt hyperplasia, T3SS-mediated intimate attachment is not required for aerobic respiration but for hydrogen peroxide (H2O2) respiration using cytochrome c peroxidase (Ccp). The epithelial NADPH oxidase NOX1 is the primary source of luminal H2O2 early after C. rodentium infection and is required for Ccp-dependent growth. Our results suggest that NOX1-derived H2O2 is a resource that governs bacterial growth and survival in close proximity to the mucosal surface during gut homeostasis.


Citrobacter rodentium/growth & development , Citrobacter rodentium/metabolism , Cytochrome-c Peroxidase/physiology , Hydrogen Peroxide/metabolism , NADPH Oxidase 1/physiology , Anaerobiosis , Animals , Colon/microbiology , DNA, Bacterial , Feces/microbiology , Female , Germ-Free Life , Homeostasis , Host-Pathogen Interactions , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Ribosomal, 16S , Specific Pathogen-Free Organisms , Type III Secretion Systems/physiology
13.
Proc Natl Acad Sci U S A ; 117(22): 12387-12393, 2020 06 02.
Article En | MEDLINE | ID: mdl-32409599

Microbiota, host and dietary metabolites/signals compose the rich gut chemical environment, which profoundly impacts virulence of enteric pathogens. Enterohemorrhagic Escherichia coli (EHEC) engages a syringe-like machinery named type-III secretion system (T3SS) to inject effectors within host cells that lead to intestinal colonization and disease. We previously conducted a high-throughput screen to identify metabolic pathways that affect T3SS expression. Here we show that in the presence of arginine, the arginine sensor ArgR, identified through this screen, directly activates expression of the genes encoding the T3SS. Exogenously added arginine induces EHEC virulence gene expression in vitro. Congruently, a mutant deficient in arginine transport (ΔartP) had decreased virulence gene expression. ArgR also augments murine disease caused by Citrobacter rodentium, which is a murine pathogen extensively employed as a surrogate animal model for EHEC. The source of arginine sensed by C. rodentium is not dietary. At the peak of C. rodentium infection, increased arginine concentration in the colon correlated with down-regulation of the host SLC7A2 transporter. This increase in the concentration of colonic arginine promotes virulence gene expression in C. rodentium Arginine is an important modulator of the host immune response to pathogens. Here we add that arginine also directly impacts bacterial virulence. These findings suggest that a delicate balance between host and pathogen responses to arginine occur during disease progression.


Citrobacter rodentium/metabolism , Enterobacteriaceae Infections/microbiology , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Gene Expression Regulation, Bacterial , Animals , Arginine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/pathogenicity , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Humans , Mice , Mice, Inbred C3H , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
14.
Article En | MEDLINE | ID: mdl-32015030

Infections with enterohemorrhagic Escherichia coli (EHEC) cause disease ranging from mild diarrhea to hemolytic-uremic syndrome (HUS) and are the most common cause of renal failure in children in high-income countries. The severity of the disease derives from the release of Shiga toxins (Stx). The use of antibiotics to treat EHEC infections is generally avoided, as it can result in increased stx expression. Here, we systematically tested different classes of antibiotics and found that their influence on stx expression and release varies significantly. We assessed a selection of these antibiotics in vivo using the Citrobacter rodentium ϕstx2dact mouse model and show that stx2d-inducing antibiotics resulted in weight loss and kidney damage despite clearance of the infection. However, several non-Stx-inducing antibiotics cleared bacterial infection without causing Stx-mediated pathology. Our results suggest that these antibiotics might be useful in the treatment of EHEC-infected human patients and decrease the risk of HUS development.


Acute Kidney Injury/prevention & control , Anti-Bacterial Agents/therapeutic use , Enterohemorrhagic Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Shiga Toxin 2/metabolism , Acute Kidney Injury/microbiology , Animals , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Disease Models, Animal , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/microbiology , Mice , Mice, Inbred C57BL , Shiga Toxin 2/genetics , Shiga Toxin 2/toxicity
15.
Sci Rep ; 10(1): 1073, 2020 01 23.
Article En | MEDLINE | ID: mdl-31974499

Many Gram-negative bacterial pathogens interact with mammalian cells by using type III secretion systems (T3SS) to inject virulence proteins into host cells. A subset of these injected protein 'effectors' are enzymes that inhibit the function of host proteins by catalyzing the addition of unusual post-translational modifications. The E. coli and Citrobacter rodentium NleB effectors, as well as the Salmonella enterica SseK effectors are glycosyltransferases that modify host protein substrates with N-acetyl glucosamine (GlcNAc) on arginine residues. This post-translational modification disrupts the normal functioning of host immune response proteins. T3SS effectors are thought to be inactive within the bacterium and fold into their active conformations after they are injected, due to the activity of chaperones that keep the effectors in a structural state permissive for secretion. While performing mass spectrometry experiments to identify glycosylation substrates of NleB orthologs, we unexpectedly observed that the bacterial glutathione synthetase (GshB) is glycosylated by NleB on arginine residue R256. NleB-mediated glycosylation of GshB resulted in enhanced GshB activity, leading to an increase in glutathione production, and promoted C. rodentium survival in oxidative stress conditions. These data represent, to our knowledge, the first intra-bacterial activity for a T3SS effector and show that arginine-GlcNAcylation, once thought to be restricted to host cell compartments, also plays an important role in regulating bacterial physiology.


Bacterial Proteins/metabolism , Citrobacter rodentium/metabolism , Salmonella enterica/metabolism , Type III Secretion Systems/metabolism , Bacterial Proteins/genetics , Citrobacter rodentium/genetics , Glycosylation , Salmonella enterica/genetics , Type III Secretion Systems/genetics
16.
Nat Microbiol ; 5(2): 368-378, 2020 02.
Article En | MEDLINE | ID: mdl-31873206

Enteric pathogens sense the complex chemistry within the gastrointestinal tract to efficiently compete with the resident microbiota and establish a colonization niche. Here, we show that enterohaemorrhagic Escherichia coli and Citrobacter rodentium, its surrogate in a mouse infection model, sense galacturonic acid to initiate a multi-layered program towards successful mammalian infection. Galacturonic acid utilization as a carbon source aids the initial pathogen expansion. The main source of galacturonic acid is dietary pectin, which is converted to galacturonic acid by the prominent member of the microbiota, Bacteroides thetaiotamicron. This is regulated by the ExuR transcription factor. However, galacturonic acid is also sensed as a signal through ExuR to modulate the expression of the genes encoding a molecular syringe known as a type III secretion system, leading to infectious colitis and inflammation. Galacturonic acid acts as both a nutrient and a signal directing the exquisite microbiota-pathogen relationships within the gastrointestinal tract. This work highlights that differential dietary sugar availability influences the relationship between the microbiota and enteric pathogens, as well as disease outcomes.


Citrobacter rodentium/pathogenicity , Enterohemorrhagic Escherichia coli/pathogenicity , Gastrointestinal Microbiome/physiology , Hexuronic Acids/metabolism , Animals , Bacteroides thetaiotaomicron/metabolism , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Diet , Disease Models, Animal , Enterobacteriaceae Infections/etiology , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Infections/etiology , Female , Genes, Bacterial , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Pectins/metabolism , Virulence/genetics , Virulence/physiology
17.
Nat Microbiol ; 5(1): 116-125, 2020 01.
Article En | MEDLINE | ID: mdl-31686025

Metabolic reprogramming is associated with the adaptation of host cells to the disease environment, such as inflammation and cancer. However, little is known about microbial metabolic reprogramming or the role it plays in regulating the fitness of commensal and pathogenic bacteria in the gut. Here, we report that intestinal inflammation reprograms the metabolic pathways of Enterobacteriaceae, such as Escherichia coli LF82, in the gut to adapt to the inflammatory environment. We found that E. coli LF82 shifts its metabolism to catabolize L-serine in the inflamed gut in order to maximize its growth potential. However, L-serine catabolism has a minimal effect on its fitness in the healthy gut. In fact, the absence of genes involved in L-serine utilization reduces the competitive fitness of E. coli LF82 and Citrobacter rodentium only during inflammation. The concentration of luminal L-serine is largely dependent on dietary intake. Accordingly, withholding amino acids from the diet markedly reduces their availability in the gut lumen. Hence, inflammation-induced blooms of E. coli LF82 are significantly blunted when amino acids-particularly L-serine-are removed from the diet. Thus, the ability to catabolize L-serine increases bacterial fitness and provides Enterobacteriaceae with a growth advantage against competitors in the inflamed gut.


Diet , Enterobacteriaceae/physiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Serine/metabolism , Animals , Citrobacter rodentium/genetics , Citrobacter rodentium/growth & development , Citrobacter rodentium/metabolism , Citrobacter rodentium/physiology , Colitis/microbiology , Colitis/pathology , Diet/adverse effects , Enterobacteriaceae/genetics , Enterobacteriaceae/growth & development , Enterobacteriaceae/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli/physiology , Gene Expression Regulation, Bacterial , Intestinal Mucosa/metabolism , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Microbial Interactions , Serine/deficiency , Specific Pathogen-Free Organisms
18.
Nat Rev Microbiol ; 17(11): 701-715, 2019 11.
Article En | MEDLINE | ID: mdl-31541196

Citrobacter rodentium is an extracellular enteric mouse-specific pathogen used to model infections with human pathogenic Escherichia coli and inflammatory bowel disease. C. rodentium injects type III secretion system effectors into intestinal epithelial cells (IECs) to target inflammatory, metabolic and cell survival pathways and establish infection. While the host responds to infection by activating innate and adaptive immune signalling, required for clearance, the IECs respond by rapidly shifting bioenergetics to aerobic glycolysis, which leads to oxygenation of the epithelium, an instant expansion of mucosal-associated commensal Enterobacteriaceae and a decline of obligate anaerobes. Moreover, infected IECs reprogramme intracellular metabolic pathways, characterized by simultaneous activation of cholesterol biogenesis, import and efflux, leading to increased serum and faecal cholesterol levels. In this Review we summarize recent advances highlighting the intimate relationship between C. rodentium pathogenesis, metabolism and the gut microbiota.


Citrobacter rodentium/growth & development , Citrobacter rodentium/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Host Microbial Interactions , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Adaptive Immunity , Aerobiosis , Animals , Citrobacter rodentium/metabolism , Energy Metabolism , Epithelial Cells/immunology , Epithelial Cells/pathology , Glycolysis , Immunity, Innate , Mice , Type III Secretion Systems/metabolism , Virulence Factors/metabolism
19.
Braz J Microbiol ; 50(4): 881-886, 2019 Oct.
Article En | MEDLINE | ID: mdl-31456170

Enterohemorrhagic (EHEC) and enteropathogenic Escherichia coli (EPEC) are human intestinal pathogens of clinical importance and their mechanism of pathogenicity is widely studied. However, both EHEC and EPEC poorly infect mice, whereas they do not develop important characteristics of the disease, hindering studies about mechanisms of virulence in vivo. Citrobacter rodentium exhibits high similarity of its genes with these human pathogens, including the island of pathogenicity Locus of Enterocyte Effacement (LEE). Therefore, C. rodentium becomes an alternative in vivo model for microorganisms that harbor LEE. The QseC directly regulates LEE as well as virulence mechanisms on these pathogens. Here, we report a novel surface motility in C. rodentium QseC-mediated in this non-flagellated bacterium. Moreover, we show norepinephrine and ethanolamine act as environmental signals in this movement. Hence, this study clarifies a novel role of the sensor QseC in completely unreported motility process of C. rodentium.


Bacterial Proteins/metabolism , Citrobacter rodentium/cytology , Citrobacter rodentium/metabolism , Ethanolamine/metabolism , Norepinephrine/metabolism , Bacterial Proteins/genetics , Citrobacter rodentium/genetics , Citrobacter rodentium/pathogenicity , Enterobacteriaceae Infections/microbiology , Genomic Islands , Humans , Virulence
...