Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Antimicrob Agents Chemother ; 67(7): e0009023, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37278639

ABSTRACT

Mycobacterium abscessus infections are difficult to treat and are often considered untreatable without tissue resection. Due to the intrinsic drug-resistant nature of the bacteria, combination therapy of three or more antibiotics is recommended. A major challenge in treating M. abscessus infections is the absence of a universal combination therapy with satisfying clinical success rates, leaving clinicians to treat infections using antibiotics lacking efficacy data. We systematically measured drug combinations in M. abscessus to establish a resource of drug interaction data and identify patterns of synergy to help design optimized combination therapies. We measured 191 pairwise drug combination effects among 22 antibacterials and identified 71 synergistic pairs, 54 antagonistic pairs, and 66 potentiator-antibiotic pairs. We found that commonly used drug combinations in the clinic, such as azithromycin and amikacin, are antagonistic in the lab reference strain ATCC 19977, whereas novel combinations, such as azithromycin and rifampicin, are synergistic. Another challenge in developing universally effective multidrug therapies for M. abscessus is the significant variation in drug response between isolates. We measured drug interactions in a focused set of 36 drug pairs across a small panel of clinical isolates with rough and smooth morphotypes. We observed strain-dependent drug interactions that cannot be predicted from single-drug susceptibility profiles or known drug mechanisms of action. Our study demonstrates the immense potential to identify synergistic drug combinations in the vast drug combination space and emphasizes the importance of strain-specific combination measurements for designing improved therapeutic interventions.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Humans , Azithromycin/pharmacology , Azithromycin/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Amikacin/pharmacology , Amikacin/therapeutic use , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Drug Interactions , Microbial Sensitivity Tests
2.
Curr Opin Microbiol ; 65: 183-190, 2022 02.
Article in English | MEDLINE | ID: mdl-34929548

ABSTRACT

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.


Subject(s)
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
3.
Am J Physiol Gastrointest Liver Physiol ; 319(2): G227-G237, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32597706

ABSTRACT

Antibiotic treatment is a standard therapy for Clostridioides difficile infection, but dysbiosis of the gut microbiota due to antibiotic exposure is also a major risk factor for the disease. Following an initial episode of C. difficile infection, a relentless cycle of recurrence can occur, where persistent treatment-related dysbiosis predisposes the patient to subsequent relapse. This study uses a longitudinal study design to compare the effects of a narrow-spectrum (ridinilazole) or broad-spectrum antibiotic (vancomycin) on intestinal bile acid profiles and their associations with gut bacteria over the course of C. difficile infection treatment. At the end of treatment (day 10), subjects receiving vancomycin showed a nearly 100-fold increase in the ratio of conjugated to secondary bile acids in their stool compared with baseline, whereas subjects receiving ridinilazole maintained this ratio near baseline levels. Correlation analysis detected significant positive associations between secondary bile acids and several Bacteroidales and Clostridiales families. These families were depleted in the vancomycin group but preserved at near-baseline abundance in the ridinilazole group. Enterobacteriaceae, which expanded to a greater extent in the vancomycin group, correlated negatively and positively with secondary and conjugated primary bile acids, respectively. Bile acid ratios at the end of treatment were significantly different between those who recurred and those who did not. These results indicate that a narrow-spectrum antibiotic maintains an intestinal bile acid profile associated with a lowered risk of recurrence.NEW & NOTEWORTHY This is the first study to demonstrate in humans the relationships between Clostridioides difficile antibiotic treatment choice and bile acid metabolism both during therapy and after treatment cessation. The results show a microbiota- and metabolome-preserving property of a novel narrow-spectrum agent that correlates with the agent's favorable sustained clinical response rates compared with broad-spectrum antibiotic treatment.


Subject(s)
Anti-Bacterial Agents/pharmacology , Benzimidazoles/pharmacology , Bile Acids and Salts/chemistry , Clostridiales/drug effects , Gastrointestinal Microbiome/drug effects , Pyridines/pharmacology , Bile Acids and Salts/metabolism , Feces/chemistry , Gastrointestinal Microbiome/physiology , Humans
4.
PLoS One ; 13(8): e0199810, 2018.
Article in English | MEDLINE | ID: mdl-30071046

ABSTRACT

Ridinilazole, a novel targeted antibacterial being developed for the treatment of C. difficile infection (CDI) and prevention of recurrence, was shown in a recent Phase 2 study to be superior to vancomycin with regard to the primary efficacy measure, sustained clinical response (SCR), with the superiority being driven primarily by marked reductions in the rates of CDI recurrence within 30 days. Tolerability of ridinilazole was comparable to that of vancomycin. The current nested cohort study compared the effects of ridinilazole and vancomycin on fecal microbiota during and after treatment among participants in the Phase 2 study. Changes in the microbiota were assessed using qPCR and high-throughput sequencing on participants' stools collected at multiple time-points (baseline [Day 1], Day 5, end-of-treatment [EOT; Day 10], Day 25, end-of-study [EOS; Day 40], and at CDI recurrence). qPCR analyses showed profound losses of Bacteroides, C. coccoides, C. leptum, and Prevotella groups at EOT with vancomycin treatment, while ridinilazole-treated participants had a modest decrease in C. leptum group levels at EOT, with levels recovering by Day 25. Vancomycin-treated participants had a significant increase in the Enterobacteriaceae group, with this increase persisting beyond EOT. At EOT, alpha diversity decreased with both antibiotics, though to a significantly lesser extent with ridinilazole (p <0.0001). Beta diversity analysis showed a significantly larger weighted Unifrac distance from baseline-to-EOT with vancomycin. Taxonomically, ridinilazole had a markedly narrower impact, with modest reductions in relative abundance in Firmicutes taxa. Microbiota composition returned to baseline sooner with ridinilazole than with vancomycin. Vancomycin treatment resulted in microbiome-wide changes, with significant reductions in relative abundances of Firmicutes, Bacteroidetes, Actinobacteria, and a profound increase in abundance of Proteobacteria. These findings demonstrate that ridinilazole is significantly less disruptive to microbiota than vancomycin, which may contribute to the reduced CDI recurrence observed in the Phase 2 study.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Benzimidazoles/therapeutic use , Pyridines/therapeutic use , Vancomycin/therapeutic use , Actinobacteria/drug effects , Actinobacteria/isolation & purification , Anti-Bacterial Agents/pharmacology , Benzimidazoles/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Cohort Studies , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification , DNA, Bacterial/metabolism , Discriminant Analysis , Feces/microbiology , Firmicutes/drug effects , Firmicutes/isolation & purification , Gastrointestinal Microbiome/drug effects , Humans , Principal Component Analysis , Proteobacteria/drug effects , Proteobacteria/isolation & purification , Pyridines/pharmacology , Vancomycin/pharmacology
5.
J Antimicrob Chemother ; 73(8): 2078-2084, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29718329

ABSTRACT

Objectives: We evaluated the antimicrobial susceptibility and ribotypes of Clostridium difficile isolates from participants in a Phase 2 study of ridinilazole, a novel targeted-spectrum agent for treatment of C. difficile infection. Methods: Participants received ridinilazole (200 mg twice daily) or vancomycin (125 mg four times daily) for 10 days (ClinicalTrials.gov: NCT02092935). The MICs of ridinilazole and comparators for C. difficile isolates from stool samples were determined by agar dilution. Toxin gene profiling was performed by multiplex PCR and ribotype identification by capillary electrophoresis. Results: Eighty-nine isolates were recovered from 88/100 participants (one participant had two strains at baseline). The median colony count (cfu/g stool) was 1.9 × 104 (range: 2.5 × 102-7.0 × 106). Twelve participants (three received ridinilazole and nine received vancomycin) experienced recurrence, confirmed by immunoassays for free toxin in stool samples. The ribotype of eight out of nine isolates obtained at recurrence matched those of the initial isolates. All isolates, including those obtained at recurrence, were susceptible to ridinilazole within the expected range [median (range) MIC: 0.12 (0.06-0.5) mg/L]. The median (range) vancomycin MIC was 1 (0.5-4.0) mg/L. At baseline, 13.6% and 13.3% of samples in the ridinilazole and vancomycin groups were positive for VRE, increasing to 23.7% and 29.7% by day 40, respectively. Common ribotypes included 014-20 (14 isolates), 027 (13), 106 (7), 002 (7), 078-126 (4), 001 (4), 087 (3) and 198 (3). Toxin gene profiling of nearly all baseline isolates (98.9%) revealed a binary toxin gene (cdtA/cdtB) prevalence of 35%. Conclusions: Ridinilazole potently inhibited recovered C. difficile isolates. Recurrence was not associated with altered susceptibility.


Subject(s)
Benzimidazoles/pharmacology , Clostridioides difficile/classification , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Pyridines/pharmacology , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Double-Blind Method , Genes, Bacterial , Humans , Microbial Sensitivity Tests , Ribotyping
6.
Anaerobe ; 51: 68-72, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29709869

ABSTRACT

Extraintestinal infection with Clostridium difficile has been reported but remains uncommon. Treatment of this unusual complication is complex given the limitations of current therapeutic options. Here we report a novel case of recurrent extraintestinal C. difficile infection that occurred following fecal microbiota transplantation. Using whole genome sequencing, we confirmed recrudescence rather than reinfection was responsible. The patient ultimately responded to prolonged, targeted antimicrobial therapy informed by susceptibility testing.


Subject(s)
Clostridioides difficile/isolation & purification , Clostridium Infections/epidemiology , Fecal Microbiota Transplantation/adverse effects , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/classification , Clostridioides difficile/genetics , Humans , Male , Middle Aged , Recurrence , Treatment Outcome , Whole Genome Sequencing
8.
Toxins (Basel) ; 8(9)2016 09 02.
Article in English | MEDLINE | ID: mdl-27598200

ABSTRACT

Ricin activates the proinflammatory ribotoxic stress response through the mitogen activated protein 3 kinase (MAP3K) ZAK, resulting in activation of mitogen activated protein kinases (MAPKs) p38 and JNK1/2. We had a novel zak-/- mouse generated to study the role of ZAK signaling in vivo during ricin intoxication. To characterize this murine strain, we intoxicated zak-/- and zak+/+ bone marrow-derived murine macrophages with ricin, measured p38 and JNK1/2 activation by Western blot, and measured zak, c-jun, and cxcl-1 expression by qRT-PCR. To determine whether zak-/- mice differed from wild-type mice in their in vivo response to ricin, we performed oral ricin intoxication experiments with zak+/+ and zak-/- mice, using blinded histopathology scoring of duodenal tissue sections to determine differences in tissue damage. Unlike macrophages derived from zak+/+ mice, those derived from the novel zak-/- strain fail to activate p38 and JNK1/2 and have decreased c-jun and cxcl-1 expression following ricin intoxication. Furthermore, compared with zak+/+ mice, zak-/- mice have decreased duodenal damage following in vivo ricin challenge. zak-/- mice demonstrate a distinct ribotoxic stress-associated phenotype in response to ricin and therefore provide a new animal model for in vivo studies of ZAK signaling.


Subject(s)
Duodenum/drug effects , MAP Kinase Kinase Kinases/deficiency , Macrophages/drug effects , Ricin/toxicity , Stress, Physiological/drug effects , Animals , Cells, Cultured , Chemokine CXCL1/metabolism , Duodenum/enzymology , Duodenum/pathology , Enzyme Activation , Genotype , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Macrophages/enzymology , Macrophages/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Infect Immun ; 84(1): 138-48, 2016 01.
Article in English | MEDLINE | ID: mdl-26483408

ABSTRACT

Infection with enterohemorrhagic Escherichia coli (EHEC) can result in severe disease, including hemorrhagic colitis and the hemolytic uremic syndrome. Shiga toxins (Stx) are the key EHEC virulence determinant contributing to severe disease. Despite inhibiting protein synthesis, Shiga toxins paradoxically induce the expression of proinflammatory cytokines from various cell types in vitro, including intestinal epithelial cells (IECs). This effect is mediated in large part by the ribotoxic stress response (RSR). The Shiga toxin-induced RSR is known to involve the activation of the stress-activated protein kinases (SAPKs) p38 and JNK. In some cell types, Stx also can induce the classical mitogen-activated protein kinases (MAPKs) or ERK1/2, but the mechanism(s) by which this activation occurs is unknown. In this study, we investigated the mechanism by which Stx activates ERK1/2s in IECs and the contribution of ERK1/2 activation to interleukin-8 (IL-8) expression. We demonstrate that Stx1 activates ERK1/2 in a biphasic manner: the first phase occurs in response to StxB1 subunit, while the second phase requires StxA1 subunit activity. We show that the A subunit-dependent ERK1/2 activation is mediated through ZAK-dependent signaling, and inhibition of ERK1/2 activation via the MEK1/2 inhibitors U0126 and PD98059 results in decreased Stx1-mediated IL-8 mRNA. Finally, we demonstrate that ERK1/2 are activated in vivo in the colon of Stx2-intoxicated infant rabbits, a model in which Stx2 induces a primarily neutrophilic inflammatory response. Together, our data support a role for ERK1/2 activation in the development of Stx-mediated intestinal inflammation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Shiga Toxin 1/metabolism , Shiga Toxin 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Butadienes/pharmacology , Cell Line , Enterohemorrhagic Escherichia coli/pathogenicity , Enzyme Activation , Epithelial Cells/immunology , Escherichia coli Infections/microbiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , HEK293 Cells , Hemolytic-Uremic Syndrome/microbiology , Humans , Inflammation/immunology , Interleukin-8/biosynthesis , Interleukin-8/genetics , Intestinal Mucosa/immunology , MAP Kinase Kinase Kinases , Nitriles/pharmacology , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering , Rabbits
10.
Infect Immun ; 83(1): 28-38, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25312954

ABSTRACT

Shiga toxins (Stx) are a family of cytotoxic proteins that can cause hemolytic-uremic syndrome (HUS), a thrombotic microangiopathy, following infections by Shiga toxin-producing Escherichia coli (STEC). Renal failure is a key feature of HUS and a major cause of childhood renal failure worldwide. There are currently no specific therapies for STEC-associated HUS, and the mechanism of Stx-induced renal injury is not well understood primarily due to a lack of fully representative animal models and an inability to monitor disease progression on a molecular or cellular level in humans at early stages. Three-dimensional (3D) tissue models have been shown to be more in vivo-like in their phenotype and physiology than 2D cultures for numerous disease models, including cancer and polycystic kidney disease. It is unknown whether exposure of a 3D renal tissue model to Stx will yield a more in vivo-like response than 2D cell culture. In this study, we characterized Stx2-mediated cytotoxicity in a bioengineered 3D human renal tissue model previously shown to be a predictor of drug-induced nephrotoxicity and compared its response to Stx2 exposure in 2D cell culture. Our results demonstrate that although many mechanistic aspects of cytotoxicity were similar between 3D and 2D, treatment of the 3D tissues with Stx resulted in an elevated secretion of the kidney injury marker 1 (Kim-1) and the cytokine interleukin-8 compared to the 2D cell cultures. This study represents the first application of 3D tissues for the study of Stx-mediated kidney injury.


Subject(s)
Kidney/drug effects , Organoids/drug effects , Shiga Toxin 2/toxicity , Hepatitis A Virus Cellular Receptor 1 , Humans , Membrane Glycoproteins/analysis , Models, Biological , Organ Culture Techniques , Receptors, Virus/analysis
12.
Cancer Biol Ther ; 14(1): 56-63, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23114643

ABSTRACT

The adverse side effects of doxorubicin, including cardiotoxicity and cancer treatment-related fatigue, have been associated with inflammatory cytokines, many of which are regulated by mitogen-activated protein kinases (MAPKs). ZAK is an upstream kinase of the MAPK cascade. Using mouse primary macrophages cultured from ZAK-deficient mice, we demonstrated that ZAK is required for the activation of JNK and p38 MAPK by doxorubicin. Nilotinib, ponatinib and sorafenib strongly suppressed doxorubicin-mediated phosphorylation of JNK and p38 MAPK. In addition, these small molecule kinase inhibitors blocked the expression of IL-1ß, IL-6 and CXCL1 RNA and the production of these proteins. Co-administration of nilotinib and doxorubicin to mice decreased the expression of IL-1ß RNA in the liver and suppressed the level of IL-6 protein in the serum compared with mice that were injected with doxorubicin alone. Therefore, by reducing the production of inflammatory mediators, the inhibitors identified in the current study may be useful in minimizing the side effects of doxorubicin and potentially other chemotherapeutic drugs.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Inflammation/enzymology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Cells, Cultured , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Gene Expression/drug effects , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation Mediators/blood , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/blood , Interleukin-6/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrimidines/pharmacology
13.
Article in English | MEDLINE | ID: mdl-23162799

ABSTRACT

Shiga toxin producing Escherichia coli (STEC) are a major cause of food-borne illness worldwide. However, a consensus regarding the role Shiga toxins play in the onset of diarrhea and hemorrhagic colitis (HC) is lacking. One of the obstacles to understanding the role of Shiga toxins to STEC-mediated intestinal pathology is a deficit in small animal models that perfectly mimic human disease. Infant rabbits have been previously used to study STEC and/or Shiga toxin-mediated intestinal inflammation and diarrhea. We demonstrate using infant rabbits that Shiga toxin-mediated intestinal damage requires A-subunit activity, and like the human colon, that of the infant rabbit expresses the Shiga toxin receptor Gb(3). We also demonstrate that Shiga toxin treatment of the infant rabbit results in apoptosis and activation of p38 within colonic tissues. Finally we demonstrate that the infant rabbit model may be used to test candidate therapeutics against Shiga toxin-mediated intestinal damage. While the p38 inhibitor SB203580 and the ZAK inhibitor DHP-2 were ineffective at preventing Shiga toxin-mediated damage to the colon, pretreatment of infant rabbits with the drug imatinib resulted in a decrease of Shiga toxin-mediated heterophil infiltration of the colon. Therefore, we propose that this model may be useful in elucidating mechanisms by which Shiga toxins could contribute to intestinal damage in the human.


Subject(s)
Benzamides/metabolism , Intestines/drug effects , Intestines/pathology , Piperazines/metabolism , Protein Kinase Inhibitors/metabolism , Protein Kinases/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/metabolism , Shiga Toxin 2/toxicity , Animals , Animals, Newborn , Apoptosis , Imatinib Mesylate , MAP Kinase Kinase Kinases , Protein Subunits/toxicity , Rabbits , Shiga-Toxigenic Escherichia coli/pathogenicity , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Infect Immun ; 80(8): 2858-67, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22689816

ABSTRACT

Shiga-toxigenic Escherichia coli (STEC) O113:H21 strains that lack the locus of enterocyte effacement (LEE) efficiently invade eukaryotic cells in vitro, unlike LEE-positive O157:H7 strains. We used a fliC deletion mutant of the O113:H21 STEC strain 98NK2 (98NK2ΔfliC) to show that invasion of colonic epithelial (HCT-8) cells is heavily dependent on production of flagellin, even though adherence to the cells was actually enhanced in the mutant. Flagellin binds and signals through Toll-like receptor 5 (TLR5), but there was no evidence that either TLR5, the adaptor protein myeloid differentiation primary response gene 88 (MyD88), or the serine kinase interleukin-1 receptor-associated kinase (IRAK) were required for invasion of HCT-8 cells by strain 98NK2, as judged by transfection, RNA knockdown, or inhibitor studies. However, pretreatment of cells with anti-asialo-GM1 significantly decreased 98NK2 invasion (by 40.8%), while neuraminidase treatment (which cleaves terminal sialic acid residues, thus converting GM1 into asialo-GM1) significantly increased invasion (by 70.7%). Pretreatment of HCT-8 cells with either the cholesterol-depleting agent methyl-ß-cyclodextrin (MßCD) or the tyrosine kinase inhibitor genistein significantly decreased invasion by 98NK2, indicating a potential role for lipid rafts in the invasion mechanism. Confocal microscopy also showed invading 98NK2 colocalized with lipid raft markers caveolin-1 and GM1. Interestingly, anti-asialo-GM1, neuraminidase, MßCD, and genistein have similar effects on the vestigial level of STEC invasion seen for STEC strain 98NK2ΔfliC, indicating that lipid rafts mediate a common step in flagellin-dependent and flagellin-independent cellular invasion.


Subject(s)
Colon/cytology , Epithelial Cells/microbiology , Flagellin/metabolism , Membrane Microdomains/physiology , Shiga-Toxigenic Escherichia coli/metabolism , Cell Line, Tumor , Epithelial Cells/cytology , Flagellin/genetics , G(M1) Ganglioside/genetics , G(M1) Ganglioside/metabolism , Gene Expression Regulation, Bacterial/physiology , Humans , Protein Binding , RNA Interference , RNA, Small Interfering , Shiga-Toxigenic Escherichia coli/classification , Shiga-Toxigenic Escherichia coli/genetics , Toll-Like Receptor 5/metabolism
15.
Curr Top Microbiol Immunol ; 357: 41-65, 2012.
Article in English | MEDLINE | ID: mdl-22057792

ABSTRACT

Shiga toxins and ricin are potent inhibitors of protein synthesis. In addition to causing inhibition of protein synthesis, these toxins activate proinflammatory signaling cascades that may contribute to the severe diseases associated with toxin exposure. Treatment of cells with Shiga toxins and ricin have been shown to activate a number of signaling pathways including those associated with the ribotoxic stress response, Nuclear factor kappa B activation, inflammasome activation, the unfolded protein response, mTOR signaling, hemostasis, and retrograde trafficking. In this chapter, we review our current understanding of these signaling pathways as they pertain to intoxication by Shiga toxins and ricin.


Subject(s)
Ricin/pharmacology , Shiga Toxins/pharmacology , Signal Transduction/drug effects , Animals , Carrier Proteins/metabolism , Endoplasmic Reticulum Stress , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Peptide Chain Initiation, Translational , Protein Transport , Shiga Toxins/metabolism , Stress, Physiological
16.
J Infect Dis ; 202(9): 1415-23, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20874089

ABSTRACT

Subtilase cytotoxin (SubAB), produced by certain virulent Shiga toxigenic Escherichia coli strains, causes hemolytic uremic syndrome-like pathology in mice, including extensive microvascular thrombosis. SubAB acts by specifically cleaving the essential endoplasmic reticulum chaperone binding immunoglobulin protein (BiP). BiP has been reported to inhibit the activation of tissue factor (TF), the major initiator of extrinsic coagulation. We hypothesized that the apparent prothrombotic effect of SubAB in vivo may involve the stimulation of TF­dependent procoagulant activity. TF­dependent procoagulant activity, TF messenger RNA (mRNA) levels, and BiP cleavage were therefore examined in human macrophage cells and primary human umbilical vein endothelial cells exposed to SubAB. In both types of cells, SubAB significantly increased TF­dependent procoagulant activity, induced TF mRNA expression, and mediated BiP cleavage. No effects were seen when cells were treated with a nonproteolytic mutant toxin, SubAA272B. Our results suggest that the procoagulant effect of SubAB may be dependent on both the up­regulation of TF expression and the activation of TF by means of BiP cleavage.


Subject(s)
Escherichia coli Proteins/toxicity , Heat-Shock Proteins/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Subtilisins/toxicity , Thromboplastin/biosynthesis , Animals , Blood Coagulation/drug effects , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endothelial Cells/drug effects , Escherichia coli Proteins/metabolism , Gene Expression Profiling , Humans , Macrophages/drug effects , Mice , Subtilisins/metabolism , Thrombosis/chemically induced
17.
Infect Immun ; 78(7): 2984-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20439475

ABSTRACT

Shiga toxins expressed in the intestinal lumen during infection with Shiga-toxigenic Escherichia coli must translocate across the epithelium and enter the systemic circulation to cause systemic (pathological) effects, including hemolytic uremic syndrome. The transepithelial migration of polymorphonuclear leukocytes in response to chemokine expression by intestinal epithelial cells is thought to promote uptake of Stx from the intestinal lumen by compromising the epithelial barrier. In the present study, we investigated the hypothesis that flagellin acts in conjunction with Shiga toxin to augment this chemokine expression. We investigated the relative contributions of nuclear factor kappaB (NF-kappaB) and mitogen-activated protein kinase (MAPK) signaling to transcription and translation of interleukin-8. Using reporter gene constructs, we showed that flagellin-mediated interleukin-8 gene transcription is heavily dependent on both NF-kappaB and extracellular signal-regulated kinase 1 and 2 (ERK-1/2) activation. In contrast, inhibition of p38 has no detectable effect on interleukin-8 gene transcription, even though flagellin-mediated activation of host p38 is critical for maximal interleukin-8 protein expression. Inhibition of MAPK-interacting kinase 1 suggests that p38 signaling affects the posttranscriptional regulation of interleukin-8 protein expression induced by flagellin. Cotreatment with Stx2 and flagellin results in a synergistic upregulation of c-Jun N-terminal protein kinases (JNKs), p38 activation, and a superinduction of interleukin-8 mRNA. This synergism was also evident at the protein level, with increased interleukin-8 protein detectable following cotreatment with flagellin and Stx2. We propose that flagellin, in conjunction with Shiga toxin, synergistically upregulates stress-activated protein kinases, resulting in superinduction of interleukin-8 and, ultimately, absorption of Stx into the systemic circulation.


Subject(s)
Escherichia coli Infections/microbiology , Flagellin/metabolism , Interleukin-8/physiology , Shiga Toxin 2/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Cell Line , Cloning, Molecular , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Humans , Interleukin-8/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/physiology , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/physiology
20.
Cell Microbiol ; 10(9): 1775-86, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18433465

ABSTRACT

Subtilase cytotoxin (SubAB) is the prototype of a new family of AB(5) cytotoxins produced by Shiga toxigenic Escherichia coli. Its cytotoxic activity is due to its capacity to enter cells and specifically cleave the essential endoplasmic reticulum (ER) chaperone BiP (GRP78). In the present study, we have examined its capacity to trigger the three ER stress-signalling pathways in Vero cells. Activation of PKR-like ER kinase was demonstrated by phosphorylation of eIF2alpha, which occurred within 30 min of toxin treatment, and correlated with inhibition of global protein synthesis. Activation of inositol-requiring enzyme 1 was demonstrated by splicing of X-box-binding protein 1 mRNA, while activating transcription factor 6 activation was demonstrated by depletion of the 90 kDa uncleaved form, and appearance of the 50 kDa cleaved form. The rapidity with which ER stress-signalling responses are triggered by exposure of cells to SubAB is consistent with the hypothesis that cleavage by the toxin causes BiP to dissociate from the signalling molecules.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum/metabolism , Escherichia coli Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Subtilisins/metabolism , eIF-2 Kinase/metabolism , Animals , Chlorocebus aethiops , Signal Transduction , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...