Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
3.
Infect Immun ; 69(10): 6140-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553553

ABSTRACT

Exposure of humans to Shiga toxins (Stxs) is a risk factor for hemolytic-uremic syndrome (HUS). Because Stx-producing Escherichia coli (STEC) is a noninvasive enteric pathogen, the extent to which Stxs can cross the host intestinal epithelium may affect the risk of developing HUS. We have previously shown that Stxs can induce and superinduce IL-8 mRNA and protein in intestinal epithelial cells (IECs) in vitro via a ribotoxic stress response. We used cytokine expression arrays to determine the effect of Stx1 on various C-X-C chemokine genes in IECs. We observed that Stx1 induces multiple C-X-C chemokines at the mRNA level, including interleukin-8 (IL-8), GRO-alpha, GRO-beta, GRO-gamma, and ENA-78. Like that of IL-8, GRO-alpha and ENA-78 mRNAs are both induced and superinduced by Stx1. Furthermore, Stx1 induces both IL-8 and GRO-alpha protein in a dose-response fashion, despite an overall inhibition in host cell protein synthesis. Stx1 treatment stabilizes both IL-8 and GRO-alpha mRNA. We conclude that Stxs are able to increase mRNA and protein levels of multiple C-X-C chemokines in IECs, with increased mRNA stability at least one mechanism involved. We hypothesize that ribotoxic stress is a pathway by which Stxs can alter host signal transduction in IECs, resulting in the production of multiple chemokine mRNAs, leading to increased expression of specific proteins. Taken together, these data suggest that exposing IECs to Stxs may stimulate a proinflammatory response, resulting in influx of acute inflammatory cells and thus contributing to the intestinal tissue damage seen in STEC infection.


Subject(s)
Chemokines, CXC/genetics , Chemotactic Factors/genetics , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Interleukin-8/genetics , Shiga Toxin 1/immunology , Chemokine CXCL1 , Chemokine CXCL5 , Chemokines, CXC/biosynthesis , Chemotactic Factors/biosynthesis , Epithelial Cells/immunology , Gene Expression , Growth Substances/biosynthesis , Humans , Interleukin-8/analogs & derivatives , Interleukin-8/biosynthesis , Intestinal Mucosa/immunology , Oligonucleotide Array Sequence Analysis , RNA, Messenger , Tumor Cells, Cultured
4.
Infect Immun ; 69(10): 6148-55, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553554

ABSTRACT

Shiga toxin-producing E. coli (STEC) is a food-borne pathogen that causes serious illness, including hemolytic-uremic syndrome (HUS). STEC colonizes the lower intestine and produces Shiga toxins (Stxs). Stxs appear to translocate across intestinal epithelia and affect sensitive endothelial cell beds at various sites. We have previously shown that Stxs cross polarized intestinal epithelial cells (IECs) via a transcellular route and remain biologically active. Since acute inflammatory infiltration of the gut and fecal leukocytes is seen in many STEC-infected patients and since polymorphonuclear leukocyte (PMN) transmigration across polarized IECs diminishes the IEC barrier function in vitro, we hypothesized that PMN transmigration may enhance Stx movement across IECs. We found that basolateral-to-apical transmigration of neutrophils significantly increased the movement of Stx1 and Stx2 across polarized T84 IECs in the opposite direction. The amount of Stx crossing the T84 barrier was proportional to the degree of neutrophil transmigration, and the increase in Stx translocation appears to be due to increases in paracellular permeability caused by migrating PMNs. STEC clinical isolates applied apically induced PMN transmigration across and interleukin-8 (IL-8) secretion from T84 cells. Of the 10 STEC strains tested, three STEC strains lacking eae and espB (eae- and espB-negative STEC strains) induced significantly more neutrophil transmigration and significantly greater IL-8 secretion than eae- and espB-positive STEC or enteropathogenic E. coli. This study suggests that STEC interaction with intestinal epithelia induces neutrophil recruitment to the intestinal lumen, resulting in neutrophil extravasation across IECs, and that during this process Stxs may pass in greater amounts into underlying tissues, thereby increasing the risk of HUS.


Subject(s)
Chemotaxis, Leukocyte/immunology , Intestinal Mucosa/immunology , Neutrophils/immunology , Shiga Toxin 1/immunology , Shiga Toxin 2/immunology , Biological Transport , Epithelial Cells , Escherichia coli/immunology , Escherichia coli O157/immunology , Humans
5.
Antimicrob Agents Chemother ; 45(8): 2340-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11451694

ABSTRACT

GT160-246, a high-molecular-weight soluble anionic polymer, was tested in vitro and in vivo for neutralization of Clostridium difficile toxin A and B activities. Five milligrams of GT160-246 per ml neutralized toxin-mediated inhibition of protein synthesis in Vero cells induced by 5 ng of toxin A per ml or 1.25 ng of toxin B per ml. In ligated rat ileal loops, 1 mg of GT160-246 neutralized fluid accumulation caused by 5 microg of toxin A. At doses as high as 80 mg/loop, cholestyramine provided incomplete neutralization of fluid accumulation caused by 5 microg of toxin A. GT160-246 protected 80% of the hamsters from mortality caused by infection with C. difficile, whereas cholestyramine protected only 10% of animals. Treatment of C. difficile-infected hamsters with metronidazole initially protected 100% of the hamsters from mortality, but upon removal of treatment, 80% of the hamsters had relapses and died. In contrast, removal of GT160-246 treatment did not result in disease relapse in the hamsters. GT160-246 showed no antimicrobial activity in tests with a panel of 16 aerobic bacteria and yeast and 22 anaerobic bacteria and did not interfere with the in vitro activities of most antibiotics. GT160-246 offers a novel, nonantimicrobial treatment of C. difficile disease in humans.


Subject(s)
Bacterial Toxins/metabolism , Clostridioides difficile/physiology , Clostridium Infections/drug therapy , Colitis/drug therapy , Enterotoxins/metabolism , Ions/therapeutic use , Polymers/therapeutic use , Animals , Bacterial Proteins/antagonists & inhibitors , Chlorocebus aethiops , Cholestyramine Resin/therapeutic use , Clostridium Infections/metabolism , Clostridium Infections/microbiology , Colitis/metabolism , Colitis/microbiology , Cricetinae , Humans , In Vitro Techniques , Ions/metabolism , Ions/pharmacology , Lactams/pharmacology , Male , Metronidazole/therapeutic use , Microbial Sensitivity Tests , Polymers/metabolism , Polymers/pharmacology , Rats , Rats, Wistar , Sulfonic Acids , Survival Rate , Vero Cells/microbiology
6.
Appl Environ Microbiol ; 67(4): 1619-27, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11282614

ABSTRACT

Prevalence, antibiotic susceptibility, and genetic diversity were determined for Escherichia coli O157:H7 isolated over 11 months from four beef cattle feedlots in southwest Kansas. From the fecal pat (17,050) and environmental (7,134) samples collected, 57 isolates of E. coli O157:H7 were identified by use of bacterial culture and latex agglutination (C/LA). PCR showed that 26 isolates were eaeA gene positive. Escherichia coli O157:H7 was identified in at least one of the four feedlots in 14 of the 16 collections by C/LA and in 9 of 16 collections by PCR, but consecutive positive collections at a single feedlot were rare. Overall prevalence in fecal pat samples was low (0.26% by C/LA, and 0.08% by PCR). No detectable differences in prevalence or antibiotic resistance were found between isolates collected from home pens and those from hospital pens, where antibiotic use is high. Resistant isolates were found for six of the eight antibiotics that could be used to treat E. coli infections in food animals, but few isolates were multidrug resistant. The high diversity of isolates as measured by random amplification of polymorphic DNA and other characteristics indicates that the majority of isolates were unique and did not persist at a feedlot, but probably originated from incoming cattle. The most surprising finding was the low frequency of virulence markers among E. coli isolates identified initially by C/LA as E. coli O157:H7. These results demonstrate that better ways of screening and confirming E. coli O157:H7 isolates are required for accurate determination of prevalence.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cattle Diseases/epidemiology , Escherichia coli Infections/veterinary , Escherichia coli O157/drug effects , Genetic Variation/genetics , Animal Husbandry , Animals , Cattle , Cattle Diseases/microbiology , Drug Resistance, Microbial , Environmental Microbiology , Escherichia coli Infections/epidemiology , Escherichia coli O157/genetics , Escherichia coli O157/isolation & purification , Escherichia coli O157/pathogenicity , Feces/microbiology , Meat , Microbial Sensitivity Tests , Polymerase Chain Reaction/methods , Prevalence , Random Amplified Polymorphic DNA Technique , Virulence/genetics
8.
Infect Immun ; 69(3): 1934-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179378

ABSTRACT

The Shiga toxins (Stx) are critical virulence factors for Escherichia coli O157:H7 and other serotypes of enterohemorrhagic E. coli (EHEC). These potent toxins are encoded in the genomes of temperate lambdoid bacteriophages. We recently demonstrated that induction of the resident Stx2-encoding prophage in an O157:H7 clinical isolate is required for toxin production by this strain. Since several factors produced by human cells, including hydrogen peroxide (H2O2), are capable of inducing lambdoid prophages, we hypothesized that such molecules might also induce toxin production by EHEC. Here, we studied whether H2O2 and also human neutrophils, an important endogenous source of H2O2, induced Stx2 expression by an EHEC clinical isolate. Both H2O2 and neutrophils were found to augment Stx2 production, raising the possibility that these agents may lead to prophage induction in vivo and thereby contribute to EHEC pathogenesis.


Subject(s)
Diarrhea/microbiology , Escherichia coli O157/pathogenicity , Neutrophils/immunology , Shiga Toxin/biosynthesis , Escherichia coli O157/drug effects , Humans , Hydrogen Peroxide/pharmacology , Models, Immunological , Shiga Toxin 2/biosynthesis , Siphoviridae/genetics , Virus Activation
9.
J Bacteriol ; 183(6): 2081-5, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222608

ABSTRACT

Shiga toxins (Stxs), encoded by the stxA and stxB genes, are important contributors to the virulence of Escherichia coli O157:H7 and other Stx-producing E. coli (STEC) strains. The stxA and stxB genes in STEC strains are located on the genomes of resident prophages of the lambda family immediately downstream of the phage late promoters (p(R')). The phage-encoded Q proteins modify RNA polymerase initiating transcription at the cognate p(R') promoter which creates transcription complexes that transcend a transcription terminator immediately downstream of p(R') as well as terminator kilobases distal to p(R'). To test if this Q-directed processive transcription plays a role in stx(2)AB expression, we constructed a mutant prophage in an O157:H7 clinical isolate from which p(R') and part of Q were deleted but which has an intact pStx, the previously described stx(2)AB-associated promoter. We report that production of significant levels of Stx2 in this O157:H7 isolate depends on the p(R') promoter. Since transcription initiating at p(R') ultimately requires activation of the phage lytic cascade, expression of stx(2)AB in STEC depends primarily on prophage induction. By showing this central role for the prophage in stx(2)AB expression, our findings contradict the prevailing assumption that phages serve merely as agents for virulence gene transfer.


Subject(s)
Bacteriophage lambda/genetics , Escherichia coli O157/pathogenicity , Escherichia coli O157/virology , Promoter Regions, Genetic , Shiga Toxin 2/biosynthesis , Animals , Bacteriophage lambda/physiology , Escherichia coli Infections/virology , Escherichia coli O157/metabolism , Gene Expression Regulation, Bacterial , Humans , Mice , Shiga Toxin 2/genetics , Transcription, Genetic , Virulence/genetics , Virus Activation/genetics , Virus Activation/physiology
10.
Infect Immun ; 68(9): 5450-3, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10948182

ABSTRACT

Genetic studies of Campylobacter jejuni have been limited due to the lack of a transposon mutagenesis method. Here, we describe a novel technique for random transposon mutagenesis using a mariner-based transposon into C. jejuni strain 480. Insertions were random, as demonstrated by Southern blot analysis and insertional junction sequencing. We have demonstrated, for the first time, random in vivo transposon mutagenesis of C. jejuni.


Subject(s)
Campylobacter jejuni/genetics , DNA Transposable Elements , Mutagenesis
11.
J Food Prot ; 63(6): 819-21, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10852579

ABSTRACT

Escherichia coli O157:H7 is but one of a group of Shiga toxin-producing E. coli (STEC) that cause both intestinal disease such as bloody and nonbloody diarrhea and serious complications like hemolytic uremic syndrome (HUS). While E. coli O157: H7 is the most renowned STEC, over 200 different types of STEC have been documented in meat and animals, at least 60 of which have been linked with human disease. A number of studies have suggested that non-O157 STEC are associated with clinical disease, and non-O157 STEC are present in the food supply. Non-O157 STEC, such as O111 have caused large outbreaks and HUS in the United States and other countries. The current policy in the United States is to examine ground beef for O157:H7 only, but restricting the focus to O157 will miss other important human STEC pathogens.


Subject(s)
Escherichia coli O157/isolation & purification , Food Microbiology , Meat Products/microbiology , Animals , Humans , Meat Products/classification
13.
J Infect Dis ; 181(2): 664-70, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10669353

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) cause significant disease; treatment is supportive and antibiotic use is controversial. Ciprofloxacin but not fosfomycin causes Shiga toxin-encoding bacteriophage induction and enhanced Shiga toxin (Stx) production from E. coli O157:H7 in vitro. The potential clinical relevance of this was examined in mice colonized with E. coli O157:H7 and given either ciprofloxacin or fosfomycin. Both antibiotics caused a reduction in fecal STEC. However, animals treated with ciprofloxacin had a marked increase in free fecal Stx, associated with death in two-thirds of the mice, whereas fosfomycin did not. Experiments that used a kanamycin-marked Stx2 prophage demonstrated that ciprofloxacin, but not fosfomycin, caused enhanced intraintestinal transfer of Stx2 prophage from one E. coli to another. These observations suggest that treatment of human STEC infection with bacteriophage-inducing antibiotics, such as fluoroquinolones, may have significant adverse clinical consequences and that fluoroquinolone antibiotics may enhance the movement of virulence factors in vivo.


Subject(s)
Anti-Infective Agents/toxicity , Bacterial Toxins/biosynthesis , Ciprofloxacin/toxicity , Coliphages/drug effects , Escherichia coli Infections/microbiology , Escherichia coli O157/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Bacterial Toxins/genetics , Ciprofloxacin/pharmacology , Coliphages/genetics , Coliphages/physiology , Disease Models, Animal , Escherichia coli Infections/drug therapy , Escherichia coli Infections/mortality , Escherichia coli O157/metabolism , Escherichia coli O157/virology , Fosfomycin/pharmacology , Humans , Intestines/virology , Male , Mice , Shiga Toxins
14.
Infect Immun ; 67(12): 6670-7, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10569789

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) is an important food-borne pathogen that causes hemolytic-uremic syndrome. Following ingestion, STEC cells colonize the intestine and produce Shiga toxins (Stx), which appear to translocate across the intestinal epithelium and subsequently reach sensitive endothelial cell beds. STEC cells produce one or both of two major toxins, Stx1 and Stx2. Stx2-producing STEC is more often associated with disease for reasons as yet undetermined. In this study, we used polarized intestinal epithelial cells grown on permeable filters as a model to compare Stx1 and Stx2 movement across the intestinal epithelium. We have previously shown that biologically active Stx1 is able to translocate across cell monolayers in an energy-dependent, saturable manner. This study demonstrates that biologically active Stx2 is also capable of movement across the epithelium without affecting barrier function, but significantly less Stx2 crossed monolayers than Stx1. Chilling the monolayers to 4 degrees C reduced the amount of Stx1 and Stx2 movement by 200-fold and 20-fold respectively. Stx1 movement was clearly directional, favoring an apical-to-basolateral translocation, whereas Stx2 movement was not. Colchicine reduced Stx1, but not Stx2, translocation. Monensin reduced the translocation of both toxins, but the effect was more pronounced with Stx1. Brefeldin A had no effect on either toxin. Excess unlabeled Stx1 blocks the movement of (125)I-Stx1. Excess Stx2 failed to have any effect on Stx1 movement. Our data suggests that, despite the many common physical and biochemical properties of the two toxins, they appear to be crossing the epithelial cell barrier by different pathways.


Subject(s)
Bacterial Toxins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Animals , Biological Transport , Brefeldin A/pharmacology , Cell Line , Cell Polarity , Chlorocebus aethiops , Colchicine/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Monensin/pharmacology , Shiga Toxins , Vero Cells
15.
Infect Immun ; 67(12): 6710-4, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10569798

ABSTRACT

We produced isogenic Escherichia coli K-12 lysogens of seven different Shiga toxin 2 (Stx2)-encoding bacteriophages derived from clinical Shiga toxin-producing E. coli (STEC) isolates of serotypes O157:H7, O145, O111, and O83 to assess the variability among these phages and determine if there were phage-related differences in toxin production. Phage genomic restriction fragment length polymorphisms (RFLP) and superinfection resistance studies revealed significant differences among these phages and allowed the seven phages to be placed into five distinct groups. Experiments revealed striking differences in spontaneous phage and toxin production that were correlated with the groupings derived from the RFLP and resistance studies. These results suggest that the genotype of the Stx2 prophage can influence the level of phage release and toxin expression by host strains and thus may be relevant to STEC pathogenesis.


Subject(s)
Bacterial Toxins/biosynthesis , Coliphages/genetics , Escherichia coli/metabolism , Escherichia coli/virology , Lysogeny , Bacterial Toxins/genetics , Coliphages/physiology , DNA, Viral/analysis , DNA, Viral/genetics , Escherichia coli/classification , Escherichia coli Infections/microbiology , Escherichia coli O157/metabolism , Escherichia coli O157/virology , Humans , Polymorphism, Restriction Fragment Length , Shiga Toxins
16.
Infect Immun ; 67(11): 5985-93, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531258

ABSTRACT

In the 1980s, Shiga toxin (Stx)-producing Escherichia coli O157:H7 (STEC) was identified as a cause of hemorrhagic colitis in the United States and was found to be associated with hemolytic uremic syndrome (HUS), a microangiopathic hemolytic anemia characterized by thrombocytopenia and renal failure. The precise way that Stxs cause hemorrhagic colitis and HUS is unclear. Stxs have been thought to cause disease by killing or irreversibly harming sensitive cells through a nonspecific blockade of mRNA translation, eventually resulting in cytotoxicity by preventing synthesis of critical molecules needed to maintain cell integrity. Because STEC is noninvasive, we have been exploring the host-toxin response at the level of the gastrointestinal mucosa, where STEC infection begins. We have found that Stx is capable of interleukin-8 (IL-8) superinduction in a human colonic epithelial cell line. Despite a general blockade of mRNA translation, Stx treatment results in increased IL-8 mRNA as well as increased synthesis and secretion of IL-8 protein. Our data suggest that an active Stx A subunit is required for this activity. Ricin, which has the same enzymatic activity and trafficking pathway as Stx, has similar effects. Exploration of the effects of other protein synthesis inhibitors (cycloheximide, anisomycin) suggests a mechanism of gene regulation that is distinct from a general translational blockade. Use of the specific p38/RK inhibitor SB202190 showed that blocking of this pathway results in decreased Stx-mediated IL-8 secretion. Furthermore, Stxs induced mRNA of the primary response gene c-jun, which was subsequently partially blocked by SB202190. These data suggest a novel model of how Stxs contribute to disease, namely that Stxs may alter regulation of host cell processes in sensitive cells via activation of at least one member of the mitogen-activated protein kinase family in the p38/RK cascade and induction of c-jun mRNA. Stx-induced increases in chemokine synthesis from intestinal epithelial cells could be important in augmenting the host mucosal inflammatory response to STEC infection.


Subject(s)
Bacterial Toxins/toxicity , Interleukin-8/metabolism , Intestinal Mucosa/drug effects , Anisomycin/pharmacology , Cycloheximide/pharmacology , Genes, jun , Humans , Imidazoles/pharmacology , Interleukin-8/genetics , Intestinal Mucosa/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/physiology , Pyridines/pharmacology , RNA, Messenger/analysis , Shiga Toxins , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
18.
Scand J Gastroenterol ; 34(8): 777-83, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10499478

ABSTRACT

BACKGROUND: Shiga toxin causes net fluid secretion in rabbit jejunum by selectively targeting, and inhibiting protein synthesis in, absorptive villous cells. The effect of Shiga toxin on the colon, where it is presumably produced, is not known. This study was undertaken to investigate the effect of Shiga toxin on the rat distal colon. METHODS: Net absorption of water and Na was determined by in vivo perfusion of closed loops of rat colon pre-exposed to Shiga toxin or saline. Unidirectional and net fluxes of 22Na and 36Cl were measured in vitro under voltage-clamp conditions across rat distal colon mucosa pre-exposed to Shiga toxin. Shiga toxin binding to sections of rat distal colon was localized by immunohistochemistry. Protein synthesis was measured in surface and crypt colonocytes with 3H-leucine incorporation. RESULTS: In the in vivo perfusion studies net absorption of Na and water was increased in Shiga toxin-treated colon compared with controls (P < 0.01). In the studies carried out in vitro, J(net)Na and J9net)Cl across Shiga toxin-treated mucosa were found to be significantly higher than in control tissue (P < 0.001 and P < 0.01, respectively). Net absorption of Na or Cl did not increase further in the presence of 25 mM butyrate, indicating the absence of short-chain fatty acids (SCFA)-linked NaCl absorption in Shiga toxin-treated colon. Moreover, Shiga toxin-treated colon failed to respond to theophylline, which induced secretion in the normal colon. Immunohistochemistry showed Shiga toxin binding to crypt cells but not to surface cells in the distal colon. Shiga toxin inhibited protein synthesis (by 27.3%) in crypt cells but not in surface cells (P < 0.05). CONCLUSIONS: An unexpected increase in water and NaCl absorption was noted in Shiga toxin-treated rat distal colon, which appears to result from selective effects of the toxin on secretory crypt cells.


Subject(s)
Bacterial Toxins/pharmacology , Colon/drug effects , Exotoxins/pharmacology , Ion Transport/drug effects , Sodium Chloride/metabolism , Animals , Bacterial Toxins/metabolism , Colon/metabolism , Exotoxins/metabolism , Female , Immunohistochemistry , In Vitro Techniques , Intestinal Absorption , Male , Protein Binding , Radioisotopes , Rats , Rats, Wistar , Shiga Toxins , Shigella dysenteriae , Statistics, Nonparametric
19.
Infect Immun ; 67(3): 1439-44, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10024592

ABSTRACT

Endothelial damage is characteristic of infection with Shiga toxin (Stx)-producing Escherichia coli (STEC). Because Stx-mediated endothelial cell damage at the site of infection may lead to the characteristic hemorrhagic colitis of STEC infection, we compared the effects of Stx1 and Stx2 on primary and transformed human intestinal microvascular endothelial cells (HIMEC) to those on macrovascular endothelial cells from human saphenous vein (HSVEC). Adhesion molecule, interleukin-8 (IL-8), and Stx receptor expression, the effects of cytokine activation and Stx toxins on these responses, and Stx1 and Stx2 binding kinetics and bioactivity were measured. Adhesion molecule and IL-8 expression increased in activated HIMEC, but these responses were blunted in the presence of toxin, especially in the presence of Stx1. In contrast to HSVEC, unstimulated HIMEC constitutively expressed Stx receptor at high levels, bound large amounts of toxin, were highly sensitive to toxin, and were not further sensitized by cytokines. Although the binding capacities of HIMEC for Stx1 and Stx2 were comparable, the binding affinity of Stx1 to HIMEC was 50-fold greater than that of Stx2. Nonetheless, Stx2 was more toxic to HIMEC than an equivalent amount of Stx1. The decreased binding affinity and increased toxicity for HIMEC of Stx2 compared to those of Stx1 may be relevant to the preponderance of Stx2-producing STEC involved in the pathogenesis of hemorrhagic colitis and its systemic complications. The differences between primary and transformed HIMEC in these responses were negligible. We conclude that transformed HIMEC lines could represent a simple physiologically relevant model to study the role of Stx in the pathogenesis of hemorrhagic colitis.


Subject(s)
Bacterial Toxins/toxicity , Colitis/etiology , Endothelium, Vascular/drug effects , Gastrointestinal Hemorrhage/etiology , Endothelium, Vascular/cytology , Humans , Intercellular Adhesion Molecule-1/analysis , Interleukin-8/analysis , Shiga Toxins , Trihexosylceramides/biosynthesis , Vascular Cell Adhesion Molecule-1/analysis
20.
J Infect Dis ; 179(2): 503-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9878039

ABSTRACT

The role of inflammatory cells in the pathogenesis of hemolytic-uremic syndrome induced by Shiga toxin (Stx)-producing Escherichia coli remains unclear. The hypothesis that Stx has direct effects on polymorphonuclear cell (PMN) viability and function was examined by measuring apoptosis, necrosis, phagocytosis, and spontaneous and phorbol myristate acetate (PMA)-stimulated production of reactive oxygen intermediates. PMN from 6 healthy persons were exposed to medium, Stx1 (0.01-100 ng/mL), or heat-inactivated Stx1 or Stx1 B subunit (100 ng/mL). Stx1 induced a prominent dose-dependent respiratory burst from PMN at doses as low as 0.01 ng/mL; they were less responsive to PMA stimulation and had reduced ability for phagocytosis. This dysfunction was not due to cell death, as the magnitude of apoptosis and necrosis of PMN treated with Stx1 (100 ng/mL) for 20 h was identical to that of medium control. These results suggest that Stx has direct effects on PMN that could contribute to tissue injury early in the disease.


Subject(s)
Bacterial Toxins/toxicity , Neutrophils/drug effects , Phagocytosis/drug effects , Superoxides/metabolism , Apoptosis/drug effects , Carcinogens/toxicity , Cell Adhesion Molecules/metabolism , Cell Survival/drug effects , Humans , Neutrophils/metabolism , Phorbol Esters/toxicity , Shiga Toxins
SELECTION OF CITATIONS
SEARCH DETAIL