Apoptosis STX

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

178

Induction of Apoptosis in Normal Human Renal Tubular Epithelial Cells by


Escherichia coli Shiga Toxins 1 and 2
Nobutaka Kiyokawa, Tomoko Taguchi, Tetsuya Mori, Department of Pathology, and Department of Infectious Diseases
Hiroshi Uchida, Norihide Sato, Tae Takeda, Research, National Children’s Medical Research Center, Tokyo, Japan
and Junichiro Fujimoto

The cytotoxicity of Shiga toxin (Stx) 1 and Stx2 produced by Escherichia coli to human renal
cortical epithelial cells (HRCEC) in primary culture was investigated. HRCEC express CD24, the
marker of renal distal tubules, as well as globotriaosyl ceramide/CD77, the receptor for Stxs. Binding
of Stxs to HRCEC was confirmed by positive staining with specific antibodies to Stxs. Treatment
of HRCEC with Stxs induced rapid cell death, which was reversed in the presence of neutralizing
antibody specific for Stx. DNA fragmentation was found to be accompanied by Stx-mediated cell
death in HRCEC, indicating that apoptosis was part of the process. These data and previous reports

Downloaded from http://jid.oxfordjournals.org/ at UNIVERSITY OF ARIZONA on May 30, 2015


indicate that a variety of renal cell types, including tubular epithelial cells as well as glomerular
capillary endothelial cells, may be targets for Stx-mediated apoptosis, which could contribute to the
pathogenesis of hemolytic-uremic syndrome caused by Stx-producing E. coli infection.

Recent worrisome increases in the incidences of infections a related disorder, thrombotic thrombocytopenic purpura, may
due to Shiga toxin (Stx) – producing strains of Escherichia coli support this idea [9 – 11].
(STEC) throughout the world have attracted the attention of On the other hand, several studies demonstrated that renal
numerous public health workers. The major outbreaks in the tubular epithelial cells are another possible primary target for
western part of the United States, Germany, and Japan are the Stxs [12 – 18]. First, in a murine model, injection of Stxs and
latest cases in point. The major symptom of STEC infection oral administration of STEC have been reported to damage the
is bloody diarrhea. However, a more serious problem associated cortical tubular epithelium while glomeruli remain unaffected
with this infection is the frequent development of a fatal sys- [12 – 14]. Second, the expression of the functional receptor for
temic complication called hemolytic-uremic syndrome (HUS) Stxs, globotriaosyl ceramide (Gb3)/CD77 [19 – 21], and the
[1]. binding of Stxs has been demonstrated on renal tubular epithe-
HUS, characterized by the triad of hemolytic anemia, acute lial cells [15, 16]. Third, we have observed marked elevation
renal failure, and thrombocytopenia, remains the leading cause of urinary N-acetyl glucosaminidase and b2-microglobulin,
of acute renal failure in childhood [2]. Although Stxs have both of which are specific markers of tubular function, in the
been postulated since the early 1980s to be the substance re- acute stage of HUS associated with STEC infection [17].
sponsible for HUS development [1], in vivo kinetics of Stxs In connection with the above reports, we previously demon-
as well as the pathogenesis of HUS are still unclear. However, strated that Stxs are cytotoxic to the renal carcinoma line
on the basis of histopathologic findings [3] and in vitro experi- ACHN, an in vitro model of human renal tubular epithelial
ments [4 – 7], endothelial damage in the glomeruli and arterioles cells, which is 106- to 107-fold more sensitive to Stxs than are
of the kidney induced by Stx is believed to play a crucial HUVEC [17]. More interestingly, we have recently shown that
role in the pathogenesis of Stx-mediated HUS [4, 8]. A few apoptosis is involved in Stx-mediated cytotoxicity against
observations on the correlation between apoptosis of glomeru- ACHN cells [18].
lar endothelial cells and sporadic (non – Stx-mediated) HUS or To address whether these observations in ACHN represent
the nature of normal tubular epithelial cells in the kidney, we
examined the effect of Stxs on normal human renal cortical
epithelial cells (HRCEC) by using a primary culture system.
Received 1 December 1997; revised 10 February 1998. We used HRCEC as an in vitro model of distal tubular epithelial
Financial support: Ministry of Health and Welfare (Health Sciences Research
Grants, Grant for Pediatric Research [9C-04, 9C-05], Grant-in Aid for Cancer
cells in the kidney and investigated the mechanism of the cyto-
Research [5-24, 9-10]; Entrustment of Research Programme of the Foundation toxicity of Stx1 and Stx2.
for Promotion of Cancer Research in Japan; Program for Promotion of Funda-
mental Studies in Health Sciences of the Organization for Drug ADR Relief,
R & D Promotion and Product Review of Japan; Foundation for Promotion of Materials and Methods
Cancer Research (Research Resident Fellowship to N.S.).
Reprints or correspondence: Dr. Nobutaka Kiyokawa, Department of Pathol- Shiga toxins, cells, antibodies, and cytokines. Stx1 and Stx2
ogy, National Children’s Medical Research Center, 3-35-31, Taishido, Seta-
gaya-ku, Tokyo 154-8509, Japan (nkiyokawa@nch.go.jp).
were prepared as described previously [22, 23]. Normal HRCEC
in primary culture, derived from a 24-year-old volunteer, were
The Journal of Infectious Diseases 1998;178:178–84
q 1998 by the Infectious Diseases Society of America. All rights reserved. purchased from Clonetics (Walkersville, MD) and maintained at
0022–1899/98/7801–0022$02.00 377C in a humidified 5% CO2 atmosphere. The monoclonal anti-

/ 9d4a$$jy16 05-27-98 00:20:51 jinfa UC: J Infect


JID 1998;178 (July) Shiga Toxins Induce Apoptosis in Renal Tubules 179

bodies used in this study were CD10 (IF-6 [24]), CD24 (L30 [25]),
CD34 (QBEnd10; Coulter/Immunotec, Westbrook, MA), Apo2.7
(Coulter), CD77 (38.13, Coulter; and 1A4, gift of Reiji Kannagi,
Laboratory of Experimental Pathology, Research Institute, Aichi
Cancer Center, Nagoya, Japan), anti-Stx1 (13C4, ATCC CRL 1794
[26]), and anti-Stx2 (11E10, ATCC CRL 1907; and 11F11, ATCC
CRL 1908 [27]). Fluorescein-conjugated and enzyme-conjugated
secondary antibodies were purchased from Jackson Laboratory
(West Grove, PA).
Immunofluorescence study. Cell suspensions of HRCEC were
produced by treating the cells with a non-enzymatic cell dissocia-
tion solution (Sigma-Aldrich, St. Louis) for 5 min, followed by
pipetting. The cells were then stained with monoclonal antibodies
and analyzed by flow cytometry (EPICS Profile and EPICS-XL;
Coulter) as described previously [24]. To detect Stx binding, 106

Downloaded from http://jid.oxfordjournals.org/ at UNIVERSITY OF ARIZONA on May 30, 2015


cells in suspension were incubated with and without 100 ng/mL
Stx1 or Stx2 for 1 h at 47C followed by intensive washing before
staining. To detect 7A6 antigen, which is located in the cytoplasm,
Stx-treated cells were harvested and fixed, followed by permeabili-
zation before staining with phycoerythrin-Cy5–conjugated
APO2.7 antibody, as described previously [28].
Immunohistochemical analysis. For immunohistochemical
analysis, cells grown on tissue culture chamber slides (Lab-Tek;
Miles Laboratories, Naperville, IL) were washed with PBS and air
dried, followed by fixation with 100% acetone for 15 min at 47C.
Immunoperoxidase staining was done as described previously [29].
To detect binding of Stxs, cells fixed on glass slides were incubated
with 100 ng/mL Stx1 or Stx2 for 1 h at room temperature before
staining with specific antibody.
MTT assay. To assess growth and viability, cells were plated
on 96-well plates (Corning, Corning, NY) at a concentration of
104 cells in 100 mL of complete medium/well, with and without
Stxs. After incubation for the indicated periods, MTT assays were
done as described previously [30]. Each experiment was done in
triplicate, and averages were calculated.
DNA fragmentation assay. Cells were assayed for DNA ladder
formation by gel electrophoresis. After treatment with and without
Stxs, DNAs were extracted from the cells, separated in 1% agarose
gels by electrophoresis, and examined under UV light as described
previously [31].
DNA fragmentation was also investigated in situ by the TUNEL
method using a in situ apoptosis detection kit (TACS 2 TdT[TBL]; Figure 1. Analysis of antigen expression on surfaces of HRCEC.
Trevigen, Gaithersburg, MD). Experiments were done according A, HRCEC in suspension were stained with specific monoclonal anti-
to the manufacturer’s protocol. bodies and analyzed by flow cytometry. Histogram obtained was
To evaluate relative DNA contents, cells were stained with pro- superimposed on that of negative control (CNT; cells stained with
pidium iodide (Sigma), then analyzed by flow cytometry as de- isotype-matched control immunoglobulin). x axis, fluorescence inten-
scribed previously [32]. sity; y axis, relative cell number. B, HRCEC were grown on chamber
slides. After fixation with 100% cold acetone, cells were immunohis-
tochemically stained with specific monoclonal antibodies.

Results

Characterization of cell surface antigens expressed on strongly positive for CD24 on flow cytometry but negative for
HRCEC in primary culture. It has been reported that the CD10 and CD34. HRCEC were also found to express low
epithelial components of renal tubules can be classified ac- levels of Gb3/CD77, the functional receptor for Stxs. The pat-
cording to the expression pattern of hematopoietic cell surface tern of the histograms, which depict rather wide peaks, is indic-
antigens [29, 33, 34]. Thus, we first characterized cell surface ative of a less-than-homogeneous cell population.
antigens expressed on HRCEC by using antibodies against CD The expressions of CD24 and CD77 were also confirmed
leukocyte antigens. As shown in figure 1A, HRCEC were by immunohistochemistry (figure 1B). On the basis of the dis-

/ 9d4a$$jy16 05-27-98 00:20:51 jinfa UC: J Infect


180 Kiyokawa et al. JID 1998;178 (July)

Cytotoxic effect of Stxs on HRCEC. Next, we investigated


whether Stxs affect the growth and survival of HRCEC. When
HRCEC were incubated with Stx1, their growth as assessed
by MTT assay was markedly reduced in a dose-dependent
manner (figure 3A), showing HRCEC to be very sensitive to the
cytotoxic effect of Stx1. The reduced cell growth in response to
treatment with Stx1 was due to cell death, as confirmed by
trypan blue staining (data not shown). Similar results were
obtained when HRCEC were incubated with Stx2 (figure 3B),
indicating that HRCEC are susceptible to both Stx1 and Stx2.
There was no significant difference in the effects of Stx1 and
Stx2 on HRCEC. Another population of HRCEC, derived from
a different volunteer (age 19 years), was similarly examined,
and identical results were obtained (data not shown).

Downloaded from http://jid.oxfordjournals.org/ at UNIVERSITY OF ARIZONA on May 30, 2015


Figure 2. Detection of Stx binding to HRCEC. A, HRCEC were
grown on chamber slides. After fixation with 100% cold acetone,
cells were incubated with 100 ng/mL Stx1 for 1 h at 47C. After
intensive washing, Stx1 bound to HRCEC was immunohistochemi-
cally stained with anti-Stx1 monoclonal antibody 13C4 (right). As
control, isotype-matched control immunoglobulin was also used (left).
B, HRCEC in suspension were incubated with (right) and without
(left) 100 ng/mL Stx2 for 1 h at 47C. After intensive washing, cells
were stained with anti-Stx2 monoclonal antibody 11E10 and analyzed
by flow cytometry. Histogram obtained was superimposed on that of
negative control (CNT; cells stained with isotype-matched control
immunoglobulin).

tribution of these molecules in normal renal tissue [29, 33, 34],


HRCEC in primary culture were thought to correspond to the
epithelia of distal tubules.
Stx binding to HRCEC. Since HRCEC were found to ex-
press CD77 on their cell surfaces, we investigated whether Stxs
bind to these cells. As shown in figure 2A, anti-Stx1 mono-
clonal antibody labeling of HRCEC pretreated with Stx1 was
demonstrated immunohistochemically. HRCEC not treated
with Stx1, on the other hand, failed to react with anti-Stx1
antibody (data not shown). Since the specificity of this antibody
was confirmed by immunostaining and ELISA (data not
shown), the positive staining of Stx1-pretreated HRCEC with
anti-Stx1 antibody was considered to reflect specific binding
of Stx1 to HRCEC.
The specific binding of Stx to HRCEC was also confirmed
by flow cytometry. As shown in figure 2B, antibody against Figure 3. Cytotoxic effects of Stx1 (A) and Stx2 (B) on HRCEC.
HRCEC (104 cells/100 mL of complete medium/well) were incubated
Stx2 clearly stained HRCEC pretreated with Stx2, whereas no with different concentrations of Stx1 or Stx2. After incubation, viable
staining was obtained when the antibody was reacted with cell numbers were estimated by MTT assay and are presented as ratio
untreated HRCEC. against non – Stx-treated cells.

/ 9d4a$$jy16 05-27-98 00:20:51 jinfa UC: J Infect


JID 1998;178 (July) Shiga Toxins Induce Apoptosis in Renal Tubules 181

Figure 4. Detection of DNA frag-


mentation in HRCEC treated with
Stx. A, HRCEC at 50% confluence
were cultured with (lanes 3 and 4)
or without (lane 2) 100 pg/mL Stx1.
After incubation, DNAs were ex-
tracted, and 1.5 mg of DNA from
each sample was electrophoresed in
1% agarose gel. As molecular
weight control, mixture of HindIII-
digested l DNA and HaeIII-digested

Downloaded from http://jid.oxfordjournals.org/ at UNIVERSITY OF ARIZONA on May 30, 2015


f DNA was also loaded on same
gel (lane 1). B, HRCEC treated with
(lower) and without (upper) 100 pg/
mL Stx2 were tested by TUNEL
method. Typical apoptotic cells
(dark color) are indicated by arrows.
C, After treatment with Stx1 (left) or
Stx2 (right) or without Stxs (center),
HRCEC were stained with propid-
ium iodide, and DNA contents were
examined. Each experiment was
done in triplicate, and averages of
ratio of subploid cells are shown.

To assess whether the cytotoxic effect described above is cating the occurrence of DNA fragmentation induced by Stx2.
specifically mediated by Stx, we examined the effects of anti- The TUNEL assay – positive cells were already detected at an
bodies neutralizing Stxs. When 13C4 antibody, which neutral- earlier time point when HRCEC were still trypan blue – nega-
izes Stx1, was added to the culture, the cytotoxic effect of tive (data not shown).
Stx1 against HRCEC was markedly reduced (data not shown). The cleavage of nuclear DNA in HRCEC treated with Stxs
Similar results were obtained with a combination of Stx2 and was further confirmed by the detection of subploid cells with
the anti-Stx2 monoclonal antibody 11F11 on HRCEC (data not propidium iodide staining (figure 4C). Again, no significant
shown), thereby establishing that the cytotoxic effects were difference was observed between the effect of Stx1 and that
directly mediated by Stxs. of Stx2 on HRCEC in this experiment.
Detection of apoptosis in the Stx-induced cell death of A 38-kDa protein, 7A6, detected by APO2.7 antibody was
HRCEC. As we have previously observed that Stxs induce recently identified as an early-stage marker of the apoptotic
apoptosis on ACHN cells derived from human renal tubular process in various cells [28]. APO2.7 stained only a fraction
epithelial cancer cells [18], we examined the possibility that the of untreated HRCEC, but the proportion of APO2.7-positive
cytotoxic effect of Stxs on HRCEC involves an apoptotic path- HRCEC increased after treatment with Stx2 in a time-depen-
way. First, we tested whether cleavage of nuclear DNA occurs dent manner (figure 5). Equivalent results were obtained with
during the Stx-induced death of HRCEC. As shown in figure Stx1 treatment (data not shown). These findings demonstrate
4A, DNA prepared from HRCEC pretreated with Stx1 exhibited clearly that apoptosis had indeed occurred in HRCEC following
DNA ladder formation on agarose gel electrophoresis. treatment with Stxs.
Cleavage of the nuclear DNA in HRCEC treated with Stx
was also confirmed by in situ detection by the TUNEL method, Discussion
as shown in figure 4B. After being treated with Stx2, a portion Although the capillary microangiopathy and endothelial
of HRCEC clearly incorporated biotinylated nucleotides, indi- damage mediated by Stxs are generally believed to be the major

/ 9d4a$$jy16 05-27-98 00:20:51 jinfa UC: J Infect


182 Kiyokawa et al. JID 1998;178 (July)

Figure 5. Induction of apoptotic antigen


7A6. After incubation with 100 pg/mL Stx2,
HRCEC were fixed and stained with either
APO2.7 monoclonal antibody (lower panels)
or isotype matched control immunoglobulin
(upper panels), then analyzed by flow cytome-
try. To identify positive cells more clearly,
fluorescence intensity was displayed as 2-di-
mensional histogram against side light scatter.
x axis, fluorescence intensity; y axis, side light
scatter.

Downloaded from http://jid.oxfordjournals.org/ at UNIVERSITY OF ARIZONA on May 30, 2015


events triggering Stx-mediated HUS [3 – 8], several lines of intestinal epithelium of rabbit, ACHN renal adenocarcinoma,
evidence have been reported that imply that renal tubular im- Burkitt’s lymphoma, Vero, and MDCK cells [18, 31, 40 – 44].
pairment also contributes to the pathogenesis of HUS [12 – 18]. Consistent with these observations, we have demonstrated
More recently, we have made several observations support- herein that Stx also mediate apoptosis in normal renal cells in
ing this idea. First, using dual-color immunohistochemistry, primary culture. Fragmentation of genomic DNA of Stx-treated
we have demonstrated that only tubular epithelial cells ex- HRCEC clearly indicates the presence of nucleosomal DNA
pressing CD24, a surface marker of renal epithelium repre- breakage, a phenomenon typical of apoptosis. In addition, the
senting distal tubules, can bind to Stxs (Mori T, et al., unpub- expression of an apoptosis marker, the 7A6 molecule [28],
lished data). Second, we had the opportunity to examine the provides further evidence that apoptosis is indeed involved in
frozen renal autopsy tissue from a child who died of HUS the mechanism of Stx-mediated cell death of normal human
after STEC infection and found clear deposition of Stxs on a tubular epithelial cells.
significant portion of distal tubules as well as on endothelial Under our experimental condition, Stxs caused apoptosis in
cells (Uchida H, et al., unpublished data). On the basis of a portion of HRCEC. However, considering the several findings
these findings, we hypothesized that renal tubular epithelium that suggest the synergism between cytokines and Stxs in the
might be another primary target of Stxs in vivo and should induction of apoptosis [6, 18, 45, 46], the apoptosis-inducing
be viewed as a major site of the renal dysfunction that occurs effect of Stxs could be enhanced in vivo during STEC infection,
in the early phase of HUS. in which elevated plasma and urine concentrations of cytokines
In fact, the data presented herein show clearly that HRCEC have been observed [47 – 49]. In addition, previous reports de-
are highly susceptible to the cytotoxic effect of Stxs. Based on scribed a possible involvement of apoptosis in the process of
the distribution of various molecules in normal renal tissue [29, Stx-induced damage of other renal cells, such as glomerular
33], HRCEC are assumed to correspond to renal epithelia of endothelia [4, 6, 42 – 45, 50]. These observations, together with
the distal tubules. Collectively, our data indicate that distal the results described herein, further emphasize the likelihood
tubular epithelia, expressing CD24 and Gb3/CD77, are highly of a pathologic role of Stx-mediated apoptosis of various renal
susceptible to Stxs and can be directly damaged. cells in HUS.
Stx consists of A and B subunits [35]. The B subunit works The difference in sensitivity to Stx-mediated apoptosis ob-
as a receptor-binding site and facilitates the entry of the holo- served among HRCEC could be due to heterogeneity in the
toxin into susceptible cells [36]. It is generally accepted that cell population. The factors or mechanisms determining the
the cytotoxic effect of Stxs is mediated by the A subunit, which sensitivity of HRCEC to apoptosis mediated by Stx remain to
has RNA N-glycohydrolase activity and cleaves a specific ade- be elucidated.
nine residue on the 28S ribosomal subunit, resulting in inhibi- Although it has been clarified that Stxs are capable of inducing
tion of protein synthesis [36 – 39]. However, it has been re- apoptosis of various cell types from several animal species,
ported that Stxs induce apoptosis in various cells, such as nothing is known about the mechanism of apoptotic signal trans-

/ 9d4a$$jy16 05-27-98 00:20:51 jinfa UC: J Infect


JID 1998;178 (July) Shiga Toxins Induce Apoptosis in Renal Tubules 183

duction. Gb3/CD77 is known to serve as a functional receptor 8. Kaplan BS, Cleary TG, Obrig TG. Recent advances in understanding the
pathogenesis of the hemolytic uremic syndromes. Pediatr Nephrol 1990;
for Stxs [19– 21], but its natural ligand and physiologic function
4:276 – 83.
are also unknown. In the case of Burkitt’s lymphoma cells, since 9. Arends MJ, Harrison DJ. Novel histopathologic findings in a surviving
the B subunit alone was found to be sufficient to induce case of hemolytic uremic syndrome after bone marrow transplantation.
apoptosis, the presence of a signaling pathway mediating Hum Pathol 1989; 20:89 – 91.
apoptosis downstream from Gb3/CD77, which is independent 10. Laurence J, Mitra D, Steiner M, Staiano-Coico L, Jaffe E. Plasma from
of the function of the A subunit, has been postulated [31, 51]. patients with idiopathic and human immunodeficiency virus – associated
thrombotic thrombocytopenic purpura induces apoptosis in microvascu-
On the other hand, in the case of Vero cells, the B subunit
lar endothelial cells. Blood 1996; 87:3245 – 54.
itself does not participate in cell killing, suggesting the involve- 11. Mitra D, Jaffe EA, Weksler B, Hajjar KA, Soderland C, Laurence J.
ment of the A subunit in apoptosis induction [44]. That several Thrombotic thrombocytopenic purpura and sporadic hemolytic-uremic
other members of the ribosome-inactivating protein family also syndrome plasmas induce apoptosis in restricted lineages of human
cause apoptosis in epithelial cells suggests the requirement of microvascular endothelial cells. Blood 1997; 89:1224 – 34.
12. Wadolkowski EA, Sung LM, Burris JA, Samuel JE, O’Brien AD. Acute
protein synthesis inhibition for the apoptosis caused by these
renal tubular necrosis and death of mice orally infected with Escherichia
agents [42, 44]. This apparent inconsistency may indicate the coli strains that produce Shiga-like toxin type II. Infect Immun 1990;

Downloaded from http://jid.oxfordjournals.org/ at UNIVERSITY OF ARIZONA on May 30, 2015


presence of multiple mechanisms in Stx-mediated apoptosis. 58:3959 – 65.
The mechanism transducing the apoptotic signal mediated by 13. Lindgren SW, Melton AR, O’Brien AD. Virulence of enterohemorrhagic
Stx binding to Gb3/CD77 on HRCEC will be investigated in Escherichia coli O91:H21 clinical isolates in an orally infected mouse
a future study. model. Infect Immun 1993; 61:3832 – 42.
14. Tesh VL, Burris JA, Owens JW, et al. Comparison of the relative toxicities
In conclusion, the present as well as our past studies indicate
of Shiga-like toxins type I and type II for mice. Infect Immun 1993;
the involvement of renal tubular epithelial cell damage caused 61:3392 – 402.
by Stxs in the development of HUS. Although additional basic 15. Oosterwijk E, Kalisiak A, Wakka JC, Scheinberg DA, Old LJ. Monoclonal
studies must be done, the possibility of Stx-mediated apoptosis antibodies against Gala1-4Galb1-4Glc(Pk, CD77) produced with a syn-
in renal tubular epithelium, as described above, should provide thetic glycoconjugate as immunogen: reactivity with carbohydrates,
with fresh frozen human tissues and hematopoietic tumors. Int J Cancer
new approaches to understanding the pathogenesis of HUS and
1991; 48:848 – 54.
to its prevention. 16. Lingwood CA: Verotoxin-binding in human renal sections. Nephron 1994;
66:21 – 8.
17. Takeda T, Dohi S, Igarashi T, Yamanaka T, Yoshiya K, Kobayashi N.
Impairment by verotoxin of tubular function contributes to the renal
Acknowledgments
damage seen in haemolytic uremic syndrome. J Infect 1993; 27:339 –
41.
We thank R. Kannagi for the gift of CD77 monoclonal antibody 18. Taguchi T, Uchida H, Kiyokawa N, et al. Verotoxins induce apoptosis in
1A4 and M. Sone for excellent secretarial work. human renal tubular epithelium derived cells. Kidney Int 1998; 53(6).
19. Jacewicz M, Clausen H, Nudelman E, Donohue-Rolfe A, Keusch GT.
Pathogenesis of Shigella diarrhea. XI. Isolation of a Shigella toxin –
binding glycolipid from rabbit jejunum and HeLa cells and its identifi-
References cation as globotriaosylceramide. J Exp Med 1986; 163:1391 – 404.
1. Karmali MA, Petric M, Lim C, Fleming DC, Arbus GS, Lior H. The 20. Lindberg AA, Brown JE, Stromberg N, Westling-Ryd M, Schultz JE,
association between idiopathic hemolytic uremic syndrome and infec- Karlsson KA. Identification of the carbohydrate receptor for Shiga toxin
tion by verotoxin producing Escherichia coli. J Infect Dis 1985; 151: produced by Shigella dysenteriae type 1. J Biol Chem 1987; 262:1779 –
775 – 82. 85.
2. Fong JS, De Chadarevian JP, Kaplan BS. Hemolytic uremic syndrome. 21. Lingwood CA, Law H, Richardson S, et al. Glycolipid binding of purified
Current concepts and management. Pediatr Clin North Am 1982; 29: and recombinant Escherichia coli produced verotoxin in vitro. J Biol
835 – 56. Chem 1987; 262:8834 – 9.
3. Richardson SE, Karmali MA, Becker LE, Smith CR. The histopathology 22. Noda M, Yutsudo T, Nakabayashi N, Hirayama T, Takeda Y. Purification
of the hemolytic uremic syndrome associated with verocytotoxin-pro- and some properties of Shiga-like toxin from Escherichia coli O157:
ducing Escherichia coli infections. Hum Pathol 1988; 19:1102 – 8. H7 that is immunologically identical to Shiga toxin. Microb Pathog
4. Obrig TG, Del-Vecchio PJ, Brown JE, et al. Direct cytotoxic action of 1987; 2:339 – 49.
Shiga toxin on human vascular endothelial cells. Infect Immun 1988; 23. Oku Y, Yutsudo T, Hirayama T, O’Brien AD, Takeda Y. Purification and
56:2373 – 8. some properties of a Vero toxin from a human strain of Escherichia
5. Tesh VL, Samuel JE, Perera LP, Sharefkin JB, O’Brien AD. Evaluation coli that is immunologically related to Shiga-like toxin II (VT2). Microb
of the role of Shiga and Shiga-like toxins in mediating direct damage Pathog 1989; 6:113 – 22.
to human vascular endothelial cells. J Infect Dis 1991; 164:344 – 52. 24. Fujimoto J, Ishimoto K, Kiyokawa N, Tanaka S, Ishii E, Hata J. Immuno-
6. Kaye SA, Louise CB, Boyd B, Lingwood CA, Obrig TG. Shiga toxin – cytological and immunochemical analysis on the common acute lym-
associated hemolytic uremic syndrome: interleukin-1b enhancement of phoblastic leukemia antigen (CALLA): evidence that CALLA on ALL
Shiga toxin cytotoxicity toward human vascular endothelial cells in cells and granulocytes are structurally related. Hybridoma 1988; 7:
vitro. Infect Immun 1993; 61:3886 – 91. 227 – 36.
7. Obrig TG, Louise CB, Lingwood CA, Boyd B, Barley-Maloney L, Daniel 25. Kokai Y, Ishii Y, Kikuchi K. Characterization of two distinct antigens
TO. Endothelial heterogeneity in Shiga toxin receptors and responses. expressed on either resting or activated human B cells as defined by
J Biol Chem 1993; 268:15484 – 8. monoclonal antibodies. Clin Exp Immunol 1986; 64:382 – 91.

/ 9d4a$$jy16 05-27-98 00:20:51 jinfa UC: J Infect


184 Kiyokawa et al. JID 1998;178 (July)

26. Strockbine NA, Marques LR, Holmes RK, O’Brien AD. Characterization Shiga toxin on eukaryotic ribosomes. RNA N-glycosidase activity of
of monoclonal antibodies against Shiga-like toxin from Escherichia the toxins. Eur J Biochem 1988; 171:45 – 50.
coli. Infect Immun 1985; 50:695 – 700. 39. Saxena SK, O’Brien AD, Ackerman EJ. Shiga toxin, Shiga-like toxin II
27. Perera LP, Marques LR, O’Brien AD. Isolation and characterization of variant, and ricin are all single-site RNA N-glycosidases of 28 S RNA
monoclonal antibodies to Shiga-like toxin II of enterohemorrhagic Esch- when microinjected into Xenopus oocytes. J Biol Chem 1989; 264:596 –
erichia coli and use of the monoclonal antibodies in a colony enzyme- 601.
linked immunosorbent assay. J Clin Microbiol 1988; 26:2127 – 31. 40. Keenan KP, Sharpnack DD, Collins H, Formal SB, O’Brien AD. Morpho-
28. Zhang C, Ao Z, Seth A, Schlossman SF. A mitochondrial membrane logic evaluation of the effects of Shiga toxin and E. coli Shiga-like
protein defined by a novel monoclonal antibody is preferentially de- toxin on the rabbit intestine. Am J Pathol 1986; 125:69 – 80.
tected in apoptotic cells. J Immunol 1996; 157:3980 – 7. 41. Pai CH, Kelly JK, Meyers GL. Experimental infection of infant rabbits with
29. Ishii E, Fujimoto J, Tanaka S, Hata J. Immunohistochemical analysis on verotoxin-producing Escherichia coli. Infect Immun 1986;51:16–23.
normal nephrogenesis and Wilms’ tumor using monoclonal antibodies 42. Sandvig K, van Deurs B. Toxin-induced cell lysis: protection by 3-methyl-
reactive with lymphohaemopoietic antigens. Virchows Arch A 1987; adenine and cycloheximide. Exp Cell Res 1992; 200:253 – 62.
411:315 – 22. 43. Inward CD, Williams J, Chant I, et al. Verocytotoxin-1 induces apoptosis
30. Hansen MB, Nielsen SE, Berg K. Re-examination and further development in vero cells. J Infect 1995; 30:213 – 8.
of a precise and rapid dye method for measuring cell growth/cell kill. 44. Williams JM, Lea N, Lord JM, Roberts LM, Milford DV, Taylor CM.
J Immunol Methods 1989; 119:203 – 10. Comparison of ribosome-inactivating proteins in the induction of

Downloaded from http://jid.oxfordjournals.org/ at UNIVERSITY OF ARIZONA on May 30, 2015


31. Mangeney M, Lingwood CA, Taga S, Caillou B, Tursz T, Wiels J. apoptosis. Toxicol Lett 1997; 91:121 – 7.
45. Polunovsky VA, Wendt CH, Ingbar DH, Peterson MS, Bitterman PB.
Apoptosis induced in Burkitt’s lymphoma cells via Gb3/CD77, a glyco-
Induction of endothelial cell apoptosis by TNF alpha: modulation by
lipid antigen. Cancer Res 1993; 53:5314 – 9.
inhibitors of protein synthesis. Exp Cell Res 1994; 214:584 – 94.
32. Gong J, Traganos F, Darzynkiewicz Z. A selective procedure for DNA
46. Eissner G, Kohlhuber F, Grell M, et al. Critical involvement of transmem-
extraction from apoptotic cells applicable for gel electrophoresis and
brane tumor necrosis factor-alpha in endothelial programmed cell death
flow cytometry. Anal Biochem 1994; 218:314 – 9.
mediated by ionizing radiation and bacterial endotoxin. Blood 1995; 86:
33. Platt JL, LeBien TW, Michael AF. Stages of renal ontogenesis identified by
4184 – 93.
monoclonal antibodies reactive with lymphohemopoietic differentiation
47. Fitzpatrick MM, Shah V, Trompeter RS, Dillon MJ, Barratt TM. Interleu-
antigens. J Exp Med 1983; 157:155 – 72.
kin-8 and polymorphoneutrophil leucocyte activation in hemolytic ure-
34. Fina L, Molgaard HV, Robertson D, et al. Expression of the CD34 gene
mic syndrome of childhood. Kidney Int 1992; 42:951 – 6.
in vascular endothelial cells. Blood 1990; 75:2417 – 26.
48. van de Kar NC, Sauerwein RW, Demacker PN, Grau GE, van Hinsbergh
35. Lingwood CA. Verotoxins and their glycolipid receptors. Adv Lipid Res VW, Monnens LA. Plasma cytokine levels in hemolytic uremic syn-
1993; 25:189 – 212. drome. Nephron 1995; 71:309 – 13.
36. Donohue-Rolfe A, Keusch GT, Edson C, Thorley-Lawson D, Jacewicz 49. Karpman D, Andreasson A, Thysell H, Kaplan BS, Svanborg C. Cytokines
M. Pathogenesis of Shigella diarrhea. IX. Simplified high yield purifica- in childhood hemolytic uremic syndrome and thrombotic thrombocyto-
tion of Shigella toxin and characterization of subunit composition and penic purpura. Pediatr Nephrol 1995; 9:694 – 9.
function by the use of subunit-specific monoclonal and polyclonal anti- 50. Mahan JD, McAllister C, Karmali M. Verocytotoxin-1 (VT-1) induction
bodies. J Exp Med 1984; 160:1767 – 81. of apoptosis in human glomerular capillary endothelial cells (GCEC) in
37. Obrig TG, Moran TP, Brown JE. The mode of action of Shiga toxin on vitro is dependent on cytokines, cell confluence, and cell cycle [abstract
peptide elongation of eukaryotic protein synthesis. Biochem J 1987; A2067]. J Am Soc Nephrol 1996; 7:1661.
244:287 – 94. 51. Taga S, Carlier K, Mishal Z, et al. Intracellular signaling events in
38. Endo Y, Tsurugi K, Yutsudo T, Takeda Y, Ogasawara T, Igarashi K. Site CD77-mediated apoptosis of Burkitt’s lymphoma cells. Blood 1997;
of action of a Vero toxin (VT2) from Escherichia coli O157:H7 and of 90:2757 – 67.

/ 9d4a$$jy16 05-27-98 00:20:51 jinfa UC: J Infect

You might also like