INTRODUCTION
Shiga toxins (Stxs) are a family of genetically and functionally related cytotoxic proteins expressed by the enteric pathogens
Shigella dysenteriae serotype 1 and certain serotypes of
Escherichia coli. Antigenic similarity to Shiga toxin expressed by
S. dysenteriae serotype 1 was used to define Shiga toxin type 1 (Stx1) and Stx2 expressed by Shiga toxin-producing
E. coli (STEC) (
47). Cloning and sequencing of the toxin genes revealed that Stx1 differs from the prototypical Shiga toxin by 1 amino acid, while Stx2 shares 56% sequence homology at the deduced amino acid sequence level with Shiga toxin and Stx1 (
21,
46). Stxs are AB
5 toxins, consisting of a single A subunit in noncovalent association with 5 B subunits that form a pentameric ring. B subunits are responsible for binding to target cells, while the A subunit is responsible for protein synthesis inhibition (
43). The toxin receptor is the neutral globo series glycolipid globotriaosylceramide (Gb
3), although one Stx2 variant toxin (Stx2e) has been shown to be capable of binding globotetraosylceramide (Gb
4) (
9). Following internalization, the toxins undergo retrograde transport, which delivers the toxins to the endoplasmic reticulum (ER). A fragment of the A subunit is cleaved from the holotoxin by furin or a furin-like protease during retrograde transport. This fragment, termed the A
1 fragment, is translocated across the ER membrane using the Sec61 translocon and enters the cytosol, where it cleaves a single adenine residue from the 28S rRNA component of eukaryotic ribosomes (
22,
33,
44). Stx-induced depurination leads to protein synthesis inhibition by disrupting elongation factor-dependent aminoacyl-tRNA binding to nascent polypeptides (
36). Stxs have also been shown to activate host cell signaling pathways, including the ribotoxic stress response and ER stress pathways. Activation of these intracellular signaling cascades may be important for proinflammatory cytokine/chemokine production and apoptosis induction in some cell types (
7,
31,
45).
Ingestion of Stx-producing bacteria may lead to the development of bloody diarrhea and, in some cases, progression to acute renal failure, termed diarrhea-associated hemolytic uremic syndrome (D
+HUS) (
38). D
+HUS, a leading cause of pediatric acute renal failure, is characterized by rapid-onset oligouria or anuria, azotemia, microangiopathic hemolytic anemia with schistocytosis, and thrombocytopenia (
38,
49). Histopathological examination of D
+HUS renal tissues showed that glomerular microvascular endothelial cells were frequently swollen and detached from the basement membrane and glomerular capillary lumina were occluded with fibrin-rich microthrombi (
28,
40). Glomerular endothelial cells are not the only targets damaged by Stxs in the kidney. Immunohistochemical and immunofluorescence staining techniques used on murine, baboon, and human kidney sections showed that renal tubules were rich in Gb
3, and toxin overlay studies showed that Stxs bound to renal tubules (
32,
51,
52). Primary human proximal tubule cells express high levels of membrane Gb
3 and are highly sensitive to Stx cytotoxicity
in vitro (
17,
26,
27). Karpman et al. (
23) noted that cell damage in renal biopsy specimens from pediatric D
+HUS cases and in mice fed an Stx2-producing STEC strain was localized to the renal cortex, with pathological changes detected in both glomerular endothelial and tubular epithelial cells. Clinical studies using pediatric and geriatric renal biopsy specimens isolated from D
+HUS cases detected the presence of Stx1 and Stx2 within renal tubules (
6,
54). Finally, urinary excretion of markers of proximal tubular damage, such as
N-acetyl glucosaminidase and β
2-microglobulin, are elevated early in the course of D
+HUS, suggesting that Stx-mediated renal tubular damage may precede damage to glomerular endothelial cells (
48). Collectively, these data suggest that proximal tubules may be an important early target of the toxins and that damage to renal tubules may contribute to the progression of disease leading to glomerular damage and the signs and symptoms of D
+HUS.
Cell culture of primary and immortalized cells has been helpful in understanding the role single cell types play in renal function, pathology, and regeneration. Ryan et al. (
42) created an immortalized adult human proximal tubule epithelial cell line designated human kidney 2 (HK-2). This cell line, transformed using the human papillomavirus type 16 (HPV16) E6/E7 genes, maintains proximal tubule epithelial morphology. The goal of the present study was to characterize HK-2 cells as a suitable model to study Stx-mediated renal damage and the host response to Stx1 and Stx2. We show that HK-2 cells express high levels of membrane Gb
3 and are differentially susceptible to the cytotoxic action of Stx1 and Stx2. HK-2 cells traffic labeled Stx1 B subunits to both the lysosomal and ER compartments. Stx2 uniquely induced the expression of two chemokines, macrophage inflammatory protein 1α (MIP-1α/CCL3) and MIP-1β/CCL4. Stx1 and Stx2 differentially activate components of the ER stress response in HK-2 cells. Finally, Stxs appear to induce apoptosis in HK-2 cells by activating poly(ADP-ribose) polymerase (PARP) cleavage in a caspase 3-independent manner.
MATERIALS AND METHODS
Cell culture.
The HK-2 cell line and Vero cells, an African green monkey renal epithelial cell line, were purchased from the American Type Culture Collection (Manassas, VA). HK-2 cells were maintained in complete medium consisting of keratinocyte-serum-free medium (K-SFM) supplemented with bovine pituitary extract (BPE), human recombinant epidermal growth factor (EGF) (Invitrogen, Carlsbad, CA), penicillin (100 U/ml), and streptomycin (100 μg/ml) (Gibco-BRL, Grand Island, NY) at 37°C in 5% CO2 in a humidified incubator. The Vero cells were maintained in Dulbecco's modified Eagle's medium (DMEM) (Gibco-BRL, Grand Island, NY) supplemented with 10% fetal bovine serum (FBS) (HyClone Laboratories, Logan, UT), penicillin (100 U/ml), and streptomycin (100 μg/ml) at 37°C in humidified 5% CO2.
Toxins.
Stx1 was prepared as previously described (
51). Briefly, Stx1 was purified from cell lysates prepared from
E. coli DH5α(pCKS112) by sequential ion exchange and chromatofocusing chromatography. The purity of toxin preparations was assessed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) with silver staining and Western blot analysis with anti-Stx1 antibodies. Toxin preparations contained <0.1 ng of endotoxin per ml, as determined by the
Limulus amoebocyte lysate assay (Associates of Cape Cod, Falmouth, ME). Recombinant Stx2 was obtained through the NIAID, NIH Biodefense and Emerging Infections Research Resources Repository (BEI Resources) (Manassas, VA). Purified pentameric Stx1 B subunits were a kind gift from Cheleste Thorpe, Tufts University School of Medicine, Boston, MA.
Gb3 quantification.
HK-2 cells (5.0 × 105 cells) were placed in microcentrifuge tubes in 300 μl complete medium. Stxs bind to Gb3 at 4°C; therefore, all subsequent steps were done on ice to prevent receptor internalization. Stx1 B subunits (1.2 mg/ml) were added to the cells and incubated for 1 h with gentle shaking. The cells were then centrifuged and washed twice with cold PBS. The cells were resuspended in a 1:100 dilution of anti-Stx1 B subunit murine monoclonal antibody (13C4; Hycult Biotech Inc., Plymouth Meeting, PA) for 30 min. After incubation, the cells were centrifuged and washed twice with PBS. Finally, the cells were resuspended in a 1:50 dilution of fluorescein-conjugated horse anti-mouse IgG antibody (Vector Laboratories Inc., Burlingame, CA) for 30 min. The cells were washed in PBS and analyzed by flow cytometry (FACSAria; BD Bioscience, San Jose, CA). Fluorescence parameters were gated using stained and unstained untreated cells. At least 104 events were measured for each sample.
Cytotoxicity assay.
Vero or HK-2 cells (5.0 × 104 cells per well) were plated in 96-well plates and grown to 80% confluence at 37°C. The cells were exposed to various concentrations of Stx1 or Stx2, ranging from 100 fg/ml to 1.0 mg/ml. After incubation at 37°C in 5% CO2 for 24, 48, or 72 h, 25 μl of 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) solution (5.0 mg/ml) was added to each well and incubated for 2 h at 37°C in a 5% CO2 humidified incubator. After incubation, the plates were centrifuged for 5 min. The supernatants were removed, and 100 μl of lysis buffer (20% SDS, 50% 2-2-dimethylformamide [pH 4.7]) was added to each well. The plates were incubated at 37°C for 3 h. Optical densities (OD) were measured in an automated plate reader (Dynatech MR5000; Molecular Dynamics, Chantilly, VA). The percent cell survival was calculated as follows: [(average OD570 {OD at 570 nm} of treated cells − average OD570 of untreated control cells)/average OD570 of untreated control cells] × 100. Statistical significance was assessed using a t test in Graphpad Prism version 5 (Graphpad Software, La Jolla, CA).
Intracellular toxin trafficking.
Intracellular trafficking of Stxs into HK-2 cells was determined using purified Stx1 B subunits conjugated to a fluorescent tag (Stx1 B-Alexa 488). Fifty micrograms of purified Stx1 B subunits was labeled using Alexa Fluor-488 Microscale Kits (Molecular Probes Inc., Invitrogen, Eugene, OR) as described in the manufacturer's protocol. Briefly, HK-2 cells (1.0 × 105 cells/well) were seeded overnight in four-well Lab-Tek chambered borosilicate cover glass slides (Nalge-Nunc International, Rochester, NY). The cells were washed two times with complete medium before further staining for 30 min at 37°C with cell-permeable-lysosome- or endoplasmic-reticulum-specific dye (100 nM Lyso-Tracker or 60 nM ER-Tracker live-cell staining dye; Molecular Probes Inc., Invitrogen, Eugene, OR). Complete medium containing 100 ng/ml Stx1 B-Alexa 488 was added to the cell monolayers. The cells were washed extensively and then imaged over the next 5 to 90 min. Single confocal optical sections through the middle of the majority of cells in the field of view were taken simultaneously for the red, blue, and green emission channels using a Stallion Digital Imaging Station (Carl Zeiss Microscopes, Gottingen, Germany) and SlideBook 4.2 image software (Olympus America Inc., Center Valley, PA). The images are representative of two independent experiments. All data within each experiment were collected at identical settings.
Real-time reverse transcription (RT)-PCR.
HK-2 cells were exposed to either 75 pg/ml Stx1 or Stx2 for 15, 30, 60, 120, or 240 min. Total RNA was isolated using TRIzol Plus kits with an RNase-free DNase treatment (Invitrogen, Carlsbad, CA) according to the manufacturer's instructions. RNA was reverse transcribed to cDNA using a high-capacity cDNA reverse transcription kit (Applied Biosystems, Carlsbad, CA), and real-time PCR was performed on the resulting cDNAs using SYBR green I double-stranded DNA binding dye (Applied Biosystems, Carlsbad, CA). The following real-time primers were used: tumor necrosis factor alpha (TNF-α), F (5′-CCAGGCAGTCAGATCATCTTCTC-3′) and R (5′-AGCTGGTTATCTCTAGCTCCAC-3′); interleukin 1β (IL-1β), F (5′-TCCCCAGCCCTTTTGTTGA-3′) and R (5′-TTAGAACCAAATGTGGCCGTG-3′); IL-8, F (5′-AAGGAACCATCTCACTGTGTGTAAAC-3′) and R (5′-ATCAGGAAGGCTGCCAAGAG-3′); MIP-1α, F (5′-TTGTGATTGTTTGCTCTGAGAGTTC-3′) and R (5′-CGGTCGTCACCAGACACACT-5′); MIP-1β, F (5′-CCCTGGCCTTTCCTTTCAGT-3′) and R (5′-AGCTTCCTCGCGGTGTAAGA-3′); and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), F (5′-CAACGGATTTGGTCGTATTGG-3′) and R (5′-GGCAACAATATCCACTTTACCAGAGT-3′).
Real-time PCRs were carried out with 100 nM concentrations (each) of forward and reverse primers in a final volume of 25 μl. To control for the presence of contaminating DNA, reverse transcriptase-negative reaction mixtures were included. Nontemplate controls were run to test for DNA-contaminated primers. Real-time reactions were run and analyzed by using an ABI Prism 7500 sequence detection system (Applied Biosystems, Carlsbad, CA). Dissociation curves for PCR samples were made to guarantee amplification of the correct genes. The amount of mRNA, expressed as fold change, was determined from the change in threshold cycle (CT) values normalized for GAPDH expression and then normalized to the value derived from cells at time zero prior to medium change or treatment. Statistical analyses of real-time PCR data were performed using ΔCT values. Statistical significance was assessed at a P value of <0.05 by one-way analysis of variance (ANOVA) with Dunnett's posttest using Graphpad Prism version 5 (Graphpad Software, La Jolla, CA).
Measurement of cytokine and chemokine production.
HK-2 cells were treated with 75 pg/ml Stx1 or Stx2 for 15, 30, 60, 120, or 240 min. The cellular supernatants were collected and stored at −80°C until they were analyzed. The Bio-Plex Pro Human Cytokine Standard Group 1 27-Plex kit was purchased from Bio-Rad Laboratories (Hercules, CA) and used according to the manufacturer's instructions. A series of eight standards ranging in concentration from 1.95 to 32,000 pg/ml was included in each assay. Samples from three independent experiments were analyzed in triplicate and graphed using GraphPad Prism. The data are expressed as means of fold induction ± standard errors of the mean (SEM). Statistical significance was assessed at a P value of <0.05 by one-way ANOVA with Dunnett's posttest using Graphpad Prism version 5 (Graphpad Software, La Jolla, CA).
Western blot analysis.
HK-2 cells were treated with 75 pg/ml Stx1 or Stx2, 100 μM thapsigargin (ER stress positive control), or 100 μM doxorubicin HCl (apoptosis positive control) for various times. Cells were harvested and lysed at 4°C in modified radioimmunoprecipitation assay (RIPA) buffer (1.0% Nonidet P-40, 1.0% sodium deoxycholate, 150 nM NaCl, 50 nM Tris-HCl [pH 7.5], 0.25 mM sodium pyrophosphate, sodium vanadate, and sodium fluoride [2.0 mM each], 10 mg of aprotinin/ml, 1.0 mg of leupeptin and pepstatin/ml, and 200 mM phenylmethylsulfonyl fluoride). Extracts were collected and cleared by centrifugation at 15,000 × g for 10 min, and the protein concentration was determined using the DC Protein Assay kit (Bio-Rad). Equal amounts of protein (100 μg per lane) were separated by SDS-PAGE using 4 to 20% acrylamide gels and transferred to nitrocellulose membranes, which were blocked with 5% nonfat milk prepared in Tris-buffered saline (TBS)-Tween 20 (200 mM Tris [pH 7.6], 1.38 M NaCl, 0.1% Tween 20) and incubated overnight at 4°C with primary antibodies specific for phospho-IRE1 (Novus Biologicals, Littleton, CO), ATF-6, phospho-PERK, total PERK, BiP, CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP), PARP, caspase 3, caspase 8, and actin (Cell Signaling Technology, Inc., Danvers, MA) in 5% bovine serum albumin made with TBS-0.1% Tween 20. The membranes were then incubated with secondary antibodies (horseradish peroxidase-labeled anti-rabbit or anti-mouse antibodies; Cell Signaling Technology, Inc., Danvers, MA) for 1 h at room temperature. Bands were visualized using the Western Lightning Chemiluminescence System (NEN-Perkin Elmer, Boston, MA). The intensities of the protein bands captured on autoradiography film were quantified using Image J software (NIH, Bethesda, MD). The fold induction was calculated as treated protein band intensity values divided by untreated control protein band intensity values after normalizing for loading controls. The data shown are from at least three independent experiments. Statistical significance was assessed at a P value of <0.05 by one-way ANOVA with Dunnett's posttest using Graphpad Prism version 5 (Graphpad Software, La Jolla, CA).
DISCUSSION
Renal proximal epithelial cells express abundant Gb
3 in situ and have been implicated as targets of the action of Stxs. For example, Karpman et al. used human renal cortical epithelial cells and a renal carcinoma cell line to investigate the cytotoxic effects of Stxs (
23). Stxs induced apoptosis in both cell types, as evidenced by nuclear fragmentation, DNA laddering, and the presence of terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL)-positive cells. We showed that HK-2 cells maintain high levels of Gb
3 expression
in vitro and are sensitive to Stx1 and Stx2. Given epidemiologic studies suggesting that infections with Stx2-producing
E. coli strains are more likely to progress to D
+HUS (
3,
24,
37), one might predict that HK-2 cells would be more sensitive to Stx2. We found the converse to be the case. HK-2 cells were sensitive to all doses of Stx1 at all time points tested (CD
50 < 0.1 pg/ml). HK-2 cells were less sensitive to Stx2 and over time showed an upward trend in CD
50 values, suggesting that a subpopulation of cells may have survived Stx2 exposure. Vero cells are widely used as the prototypical cell type for defining Stx cytotoxicity, and we compared Vero and HK-2 cell sensitivities to the toxins. The data highlight the importance of establishing experimental parameters in determining CD
50s. At 24 h, HK-2 cells were significantly more sensitive to Stxs; by 48 h, the cells displayed similar dose-dependent cytotoxicity profiles; and at 72 h, Vero cell monolayers were completely destroyed while a subset of HK-2 cells appeared to have survived Stx2 challenge. Histopathologic examination of renal tissues from animals administered purified Stxs showed evidence of renal tubule regeneration (
51), and the HK-2 cells that survived Stx2 challenge may represent this subpopulation. Other cell types have displayed differential sensitivity to Stxs. For example, Bauwens et al. (
1) showed that a cell line derived from human umbilical veins was
∼10 times more sensitive to killing by Stx1 than by Stx2, while microvascular endothelial cells were more sensitive to killing by Stx2. There are a number of factors that may influence cell susceptibility to Stxs, including levels of Gb
3 expression, biochemical characteristics of Gb
3 (e.g., fatty acid chain length and degree of hydroxylation), Gb
3 presentation at the membrane (e.g., lipid raft-associated expression), and mechanisms of toxin internalization and intracellular routing. Fluorescently labeled Stx1 B subunits have been extensively employed to characterize Stx intracellular routing to different cellular compartments (
11,
14). We showed that B subunits were routed to ER and lysosomal compartments in HK-2 cells. Rapid transport to the ER, with maximal fluorescence detected 60 min after Stx1 B subunit exposure, correlates with the sensitivity of HK-2 cells to killing by Stxs. The data showing that HK-2 cells route Stxs to lysosomes suggest that the subpopulation of cells surviving toxin exposure may direct a portion of internalized toxins into the lysosomal degradation pathway.
Hughes et al. (
18) noted that primary human renal proximal tubule epithelial cells cultured
in vitro for 24 h expressed basal levels of soluble TNF-α and IL-1 (84.2 ± 17.4 and 9.0 ± 1.4 ng/mg, respectively). Incubation of the cells with sublethal concentrations (0.01 to 0.1 pg/ml) of Stx1 for 4 h failed to increase cytokine expression above basal levels, although exposure to Stx1 for 24 to 48 h resulted in 2- to 4-fold increases in TNF-α and IL-1 protein expression. Increases in cytokine protein expression were accompanied by greater increases in TNF-α and IL-1 transcripts. We also detected basal expression of soluble TNF-α (∼2.0 pg/ml) in supernatants collected from HK-2 cells. The treatment of HK-2 cells with Stx1 or Stx2 for up to 4 h increased TNF-α and IL-1β transcript levels without concomitant significant increases in protein expression. Thus, prior to the onset of extensive cell death, neither primary cells nor HK-2 cells appear to be significant producers of TNF-α or IL-1β when exposed to Stxs
in vitro. Stxs did not appear to induce the expression of the neutrophil chemoattractant IL-8. With the exception of a single time point following exposure to Stx2 for 30 min, we did not detect significant elevations in IL-8 mRNA levels, and IL-8 protein levels were reduced, albeit in a statistically nonsignificant manner, compared to basal IL-8 expression. However, HK-2 cells released the macrophage chemoattractants MIP-1α and MIP-1β in response to Stx2 treatment, but not in the presence of Stx1. Keepers et al. (
25) used a murine model of Stx-mediated renal damage to show that macrophages were recruited to the kidneys of mice injected with Stx2 with or without lipopolysaccharides (LPS), in association with increased renal production of chemoattractants, including MIP-1α. Neutralizing antibodies against MIP-1α decreased macrophage infiltration and fibrin deposition within the renal vasculature. These findings suggest a role for infiltrating macrophages in D
+HUS but do not clarify which cell type(s) secretes chemokines in response to Stxs. Our data suggest that Stx2 is uniquely capable of triggering the production and release of MIP-1α and MIP-1β, and localized upregulation of chemokine production may facilitate the recruitment of activated immune cells, such as macrophages, into sites of initial tissue damage. HK-2 cells express tissue factor constitutively, and Stx1 induces the expression of cell surface tissue factor in a dose- and time-dependent manner (
35). Toxin-mediated renal tubular damage may activate the coagulation system, leading to the formation of platelet-fibrin microthrombi. Macrophages and macrophage cell lines are known to produce tissue factor and cytokines in response to Stxs (
34). Thus, infiltrating macrophages may further exacerbate inflammation, thrombogenesis, and tissue damage.
The mechanism(s) by which Stxs induce apoptosis in HK-2 cells has not been well characterized. Wilson et al. (
56) showed that HK-2 cells were sensitive to Stx2, undergoing apoptosis, as assessed by PARP cleavage and nuclear fragmentation. The silencing of expression of the proapoptotic factor Bak decreased PARP cleavage and protected HK-2 cells from apoptosis, suggesting that Stxs trigger signaling through the intrinsic or mitochondrion-mediated apoptosis pathway. Stxs induce apoptosis in the THP-1 cell line through activation of the UPR and prolonged ER stress signaling (
31). The ER is the site of protein folding and posttranslational modification, trafficking of proteins to various locations, and intracellular Ca
2+ storage (
2,
41). Protein folding and processing are monitored by a series of “folding sensors” localized within the ER membrane: the transcriptional activator ATF6, the serine/threonine kinase PERK, and the kinase/endoribonuclease IRE1. The chaperone BiP dissociates from the sensors in the presence of unfolded or misfolded proteins, leading to kinase dimerization and activation and the transit of ATF6 to the Golgi apparatus for proteolysis and activation. The activated sensors initiate the UPR, a coordinated series of signaling events involving the attenuation of translation and the transcriptional activation of genes encoding proteins involved in protein folding and degradation. Thus, the UPR may result in decreased
de novo protein synthesis and clearance of unfolded proteins from the ER lumen (
41). However, prolonged UPR signaling leads to ER stress and the induction of apoptosis (
4,
41). Therefore, we examined the capacity of Stxs to induce ER stress in HK-2 cells. Stx1 and Stx2 activated different and nonoverlapping UPR sensors. Stx1 induced ATF6 cleavage from the inactive 90-kDa form to the active 50-kDa fragment. Stx2 triggered the phosphorylation of the sensors PERK and IRE1α. Signaling for increased expression of the chaperone BiP appeared to be correlated with ATF6 activation, as Stx1 transiently, but significantly, upregulated BiP expression. ER stress-induced apoptosis is mediated in large part by the transcriptional factor CHOP. The ability of CHOP to induce apoptosis is dependent on the duration and degree of ER stress (
19). CHOP is activated through PERK and ATF6 signaling and induces the expression of several proapoptotic factors. Stx1 induced a 9-fold increase in CHOP expression 1 h after intoxication compared to a 2.5-fold maximal increase in HK-2 cells exposed to Stx2. Thus, the increased sensitivity of HK-2 cells to Stx1 appears to be correlated with the effective activation of ATF6, leading to enhanced signaling through CHOP, while HK-2 cell survival following Stx2 challenge may be related to reduced ATF6 activation and the transient activation of ER stress. Additional studies will be required to explore the role of ER stress in HK-2 cell death and survival.
Despite differences in the ER stress response, both toxins triggered PARP cleavage. Caspase 3 is the major executioner caspase involved in PARP cleavage, and several studies have shown that Stxs activate caspase 3 in epithelial cells and cell lines (
50). Thus, our inability to detect procaspase 3 cleavage in HK-2 cells treated with Stxs was unexpected. Calpains may cleave PARP in the absence of caspase activation (
55), and preliminary data from our laboratory suggest that both Stx1 and Stx2 activate calpains in HK-2 cells. PARP may also be activated by proapoptotic mitochondrial intermembrane proteins released following the disruption of mitochondrial membranes, including apoptosis-inducing factor and endonuclease G (
8). Stxs activate apoptotic signaling through the rapid activation of caspase 8 in several different cell types (
13,
30). Our data suggest that Stx2 cleaves procaspase 8 in HK-2 cells while Stx1 fails to do so. Stx-induced activation of caspase 8 may lead to the activation of the Bcl-2 protein family member BID, which in turn translocates to the mitochondria to facilitate mitochondrial-membrane depolarization and the release of mitochondrial intermembrane constituents into the cytoplasm (
29). These events trigger the formation of the apoptosome and the activation of procaspase 9, and caspase 9 then activates caspase 3. We detected minimal caspase 3 generation in Stx-treated HK-2 cells. Precisely how caspase 3 activation is disrupted is not known, but Fujii et al. (
12) showed that Stx treatment of HeLa cells resulted in increased expression of XIAP, which blocks caspase 3 activation. Whether a similar pathway limiting apoptosis induction is operative in HK-2 cells requires additional scrutiny.
We used results gathered from
in vitro and
in vivo studies to derive a model for the role of renal epithelial cells in the pathogenesis of disease caused by STEC (
Fig. 9). Following adherence to colonic epithelial cells, the bacteria produce Stxs (
Fig. 9A). LPS from STEC (or from other intestinal flora) may gain access to the submucosa, as patients with STEC infections frequently possess elevated anti-STEC O-antigen antibody titers (
38). Once within the lamina propria, the toxins encounter resident tissue macrophages and neutrophils (
Fig. 9B). Toxin interaction with macrophages elicits the rapid expression and secretion of proinflammatory cytokines and chemokines (
15,
16). TNF-α and IL-1β secreted by macrophages may exacerbate Stx-induced damage to colonic capillaries, releasing blood into the lumen of the intestine and creating portals of entry for Stxs and LPS into the bloodstream (
Fig. 9C). Toxin binding to neutrophils may facilitate hematogenous spread and access to target organs (
Fig. 9B and D) (
5). The toxins may access the renal tubular epithelium via blood vessels supplying the tubules, although the precise mechanism is not known. Based on data presented here, Stx2 may selectively induce the expression of the chemokines MIP-1α and MIP-1β by proximal tubules of the human kidney (
Fig. 9E). These chemokines recruit macrophages to sites of injury. Infiltrating macrophages may produce cytokines and sensitize glomerular endothelial cells to Stxs by upregulating expression of membrane-bound Gb
3 (
Fig. 9F). Microvascular damage progresses to the formation of fibrin deposits, which bind circulating red blood cells and platelets, leading to hemolytic anemia and thrombocytopenia (
Fig. 9G).