The American Journal of Pathology, Vol. 181, No. 10, October 2012
Copyright © 2012 American Society for Investigative Pathology.
Published by Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.ajpath.2012.06.016
Gastrointestinal, Hepatobiliary, and Pancreatic Pathology
p53 Mediates TNF-Induced Epithelial Cell Apoptosis
in IBD
Tatiana Goretsky,* Ramanarao Dirisina,*
Preetika Sinh,* Navdha Mittal,*
Elizabeth Managlia,* David B. Williams,*
Daniela Posca,*† Hyunji Ryu,*
Rebecca B. Katzman,* and Terrence A. Barrett*
From the Division of Gastroenterology,* Department of Medicine,
Northwestern University Feinberg School of Medicine, Chicago,
Illinois; and the Department of Experimental and Clinical
Medicine,† Magna Graecia, University of Catanzaro, Catanzaro,
Italy
Chronic ulcerative colitis (CUC) is characterized by
increased intestinal epithelial cell (IEC) apoptosis associated with elevated tumor necrosis factor (TNF),
inducible nitric oxide synthase (iNOS), and p53. We
previously showed that p53 is increased in crypt IECs
in human colitis and is needed for IEC apoptosis in
chronic dextran sulfate sodium-colitis. Herein, we examined the roles of TNF and iNOS in regulating p53induced IEC apoptosis in CUC. The IEC TUNEL staining, caspases 3, 8, and 9, and p53 protein levels,
induced by anti-CD3 monoclonal antibody (mAb) activation of T cells, were markedly reduced in TNF
receptor 1 and 2 gene knockout mice. Induction of
IEC apoptosis correlated with increased p53, which
was attenuated in iNOSⴚ/ⴚ mice. IEC p53 levels and
apoptosis were reduced in IL-10ⴚ/ⴚ colitic mice
treated with neutralizing TNF mAb and the iNOS inhibitor, aminoguanidine, further suggesting that TNF
and iNOS are upstream of p53 during colitis-induced
IEC apoptosis. IEC apoptosis and p53 levels were assessed in control versus untreated or anti-TNF–treated
CUC patients with equivalent levels of inflammation. Data indicated that IEC apoptosis and p53 levels were clearly higher in untreated CUC but markedly reduced in patients treated with anti-TNF mAb.
Therefore, TNF-induced iNOS activates a p53-dependent pathway of IEC apoptosis in CUC. The inhibition of IEC apoptosis may be an important mechanism for mucosal healing in anti-TNF–treated CUC
patients. (Am J Pathol 2012, 181:1306–1315; http://dx.doi.
org/10.1016/j.ajpath.2012.06.016)
1306
Human inflammatory bowel diseases (IBDs) are characterized by excessive crypt epithelial apoptosis, surface
ulceration, distorted crypt architecture, diarrhea, and
bleeding. Barrier disruption is linked to epithelial apoptosis caused by aberrant activation of innate and adaptive
immune responses.1–3 A hallmark of severe IBD is the
overproduction of tumor necrosis factor (TNF) in mucosal
tissue.2,4 The importance of TNF in disease pathogenesis
is underlined by the pronounced clinical improvement
induced when anti-TNF antibodies reduce diarrhea,
weight loss, and bleeding.4,5 At the mucosal level, antiTNF antibody treatment enhances mucosal healing with
rapid re-epithelialization of ulcerated surfaces. Studies
indicate some (eg, infliximab and adalimumab), but not
all (eg, certolizumab), anti-TNF agents induce apoptosis
of lamina propria cells, despite all three being able to
enhance mucosal healing.6,7 However, it remains unclear
whether the effect of anti-TNF on mucosal healing is
related to reduced epithelial apoptosis and, if so, through
what mechanism.
Overproduction of TNF in IBD has potent effects on
mucosal adaptive and innate immune responses.8,9 TNF
participates in macrophage activation by enhancing antimicrobial functions.10 In response to TNF, macrophages
increase production of reactive nitrosative species, such
as nitric oxide (NO⫺) and its metabolite, peroxynitrite
(ONOO⫺).11 Inducible NO synthase (iNOS) blockade inhibits disease severity and epithelial apoptosis in animal
models of IBD.12,13 Data from human IBD studies suggest that NO⫺ and ONOO⫺ stabilize p53 and activate
response pathways.14,15 During tumorigenesis, NO⫺-induced mutations of p53 inactivate tumor suppressor
function, with loss of protective effects.16 Thus, TNF-mediated activation of iNOS may be an important pathway
Supported by grants from the NIH (R01DK-054778 and R01AI-6171702 to
T.A.B.).
Accepted for publication June 27, 2012.
Supplemental material for this article can be found at http://ajp.
amjpathol.org or at http://dx.doi.org/10.1016/j.ajpath.2012.06.016.
Address reprint requests to Terrence A. Barrett, M.D., Division of Gastroenterology, Department of Medicine, Northwestern University Feinberg
School of Medicine, 676 N St Clair, Suite 1400, Chicago, IL 60611. E-mail:
[email protected].
Epithelial Cell Apoptosis in IBD
1307
AJP October 2012, Vol. 181, No. 10
for regulating epithelial cell apoptosis during colitis and
colitis-induced dysplasia.
Understanding TNF receptor signaling is complex and
difficult to apply to in vivo systems. TNF receptor 1
(TNFR1) associates with the TNF receptor–associated
death domain, which activates the extrinsic, caspase
8 –linked pathway of apoptosis.17,18 However, in some
systems examined, TNF receptor–associated death domain is dispensable for TNF-induced apoptosis, and
cross activation of TNFRl and TNFR2 converges unto
common downstream signaling events, resulting in apoptosis mediated by intrinsic (mitochondrial) pathways.17,19
The proliferative zone for intestinal epithelial cells (IECs)
resides in lower crypt regions. Cellular proliferation requires enhanced mitochondrial function. Given that epithelial apoptosis in IBD occurs in proliferative crypt epithelial cells, we suspected that pathways involving
induction of mitochondrial pathways were used. In addition, a comprehensive understanding of the role of TNF
receptor signaling within the mucosal microenvironment
requires that receptor deficiency be restricted to distinct
populations participating in mucosal immune responses.
Increased epithelial crypt cell apoptosis commonly occurs in ulcerative colitis (UC) and Crohn’s disease.20,21
Numerous in vitro and in vivo model systems have studied
this phenomenon, suggesting that TNF-mediated pathways play key roles in inducing programmed cell death in
epithelial crypts. To model these pathways, a well-characterized model of T-cell activation was used that induces transient stem/progenitor cell activation, crypt IEC
proliferation, and TNF-mediated diarrhea reminiscent of
human IBD.22,23 The reproducible kinetics of the model
permitted identification of the events in immune-mediated
apoptosis and allowed application to relevant gene
knockout models. We recently reported that p53 is the
major mediator of colonic crypt IEC apoptosis in colitis.24
This article examines the upstream events leading up to
p53 activation and IEC apoptosis. Results suggest a
mechanism by which TNF signals, through both TNFR1
and TNFR2, stimulate iNOS-mediated p53-dependent
apoptosis of crypt IECs. Studies in the IL-10⫺/⫺ murine
model of colitis confirmed that TNF-induced iNOS led to
activation of p53 and induced IEC apoptosis. Finally, we
confirm that TNF-induced p53-mediated apoptosis also
occurs in vivo during human UC. Overall, the findings
suggest that T-cell activation causes TNF and iNOS-mediated stabilization of p53, followed by p53-mediated
crypt cell apoptosis in IBD. These data have direct relevance to mechanisms of barrier disruption, ulceration,
and initiation of dysplasia seen in p53 mutant crypts.
Materials and Methods
Mice and Treatments
C57BL/6, TNFR1-knockout (TNFR1⫺/⫺), TNFR2-knockout
(TNFR2⫺/⫺), combined TNFR1/2-knockout (TNFR1/2⫺/⫺),
iNOS-knockout (iNOS⫺/⫺), p53-knockout (p53⫺/⫺), and
IL-10 – knockout (IL-10⫺/⫺) mice on the C57BL/6 background (⬎10 generations) were obtained from the Jack-
son Laboratory (Bar Harbor, ME) and screened for the
absence of wild-type (WT) gene before use. Mice were
maintained in barrier housing in the Northwestern University Center for Comparative Medicine (Chicago, IL), in
accordance with guidelines of the Northwestern University Animal Care and Usage Committee. To model acute
inflammation, mice were given i.p. injections of 0.2 mg
hamster anti-CD3 monoclonal antibody (mAb; 145-2C11)
or control hamster mAb (UC8-IB9) and sacrificed at different time points, as previously described.23 Anti-CD3
and control antibodies were purified from cell culture
supernatant over a protein G column (GBioscience, St.
Louis, MO). In some mice, the iNOS inhibitor, L-N6-(1iminoethyl) lysine (L-NIL), was given i.p., 0.2 mg 2 hours
before and at the time of anti-CD3 mAb treatment. IL10⫺/⫺ mice were moved to conventional housing 1 week
before starting piroxicam chow feeding. To accelerate
and synchronize the onset of colitis, IL-10⫺/⫺ mice were
fed 60 mg of piroxicam (Sigma, St Louis, MO)/250 g of
rodent powdered chow for 1 week and then 80 mg of
piroxicam/250 g of chow for another week. Controls were
given powdered chow for only 2 weeks. Mice then resumed standard pelleted chow and were examined on
days 28 and 46 after day 1 of piroxicam feeding. For
chronic dextran sulfate sodium (DSS)-colitis, one cycle
constituted giving the mice 2.0% DSS in their drinking
water for 7 days, followed by 14 days of regular water.
This was done three times, and the mice were sacrificed
at the end of the third cycle. Aminoguanidine was administered at 200 g/day for 2 days, and the mice were
sacrificed 12 hours after the last injection. Anti-TNF, 0.2
mg, was injected into WT mice 24 hours before anti-CD3
treatment, and 0.5 mg of anti-TNF was administered at 36
hours or 1 week, as indicated, before the mice with colitis
were sacrificed.
Immunohistochemical Localization of Apoptotic
Cells
Formalin-fixed, paraffin-embedded sections were stained
for apoptotic cells using the TUNEL method for visualizing the 3=-OH ends of DNA fragments. After digestion in
proteinase K, sections were rinsed and incubated with
0.3% H2O2 at room temperature for 20 minutes. Sections
were incubated in a terminal deoxynucleotide mixture
(Roche Diagnostics Corp., Indianapolis, IN), followed by
anti-fluorescein mAb conjugated with horseradish peroxidase, then 3=,3=-diaminobenzidine as immunodetection
substrate.25 Apoptotic indexes were calculated as the
number of TUNEL-positive epithelial cells/total number of
epithelial cells, multiplied by 100, to yield the apoptotic
index. A total of five mice (four for IL-10⫺/⫺ experiments)
were analyzed in each group, and a minimum of 8 to 20
well-oriented crypts were counted for each mouse. The
data are presented as the mean ⫾ SEM.
Real-Time PCR and Primers
Total RNA from sonicated tissue was isolated using the
RNeasy Mini Kit (Qiagen, Hilden, Germany). The high-
1308
Goretsky et al
AJP October 2012, Vol. 181, No. 10
capacity cDNA RT kit (Applied Biosystems, Foster City,
CA) was used to synthesize cDNA. Expression of genes
was determined by real-time quantitative PCR using the
ABI 7500 Real Time PCR system and the Power SYBR
Green PCR master mix (Applied Biosystems). Primers
were selected based on nucleotide sequences downloaded from the National Center for Biotechnology Information data bank for TNF-␣ (forward primer, 5=CCCAGGGACCTCTCTCTAATCA-3=; reverse primer,
5=-GGTTTGCTACAACATGGGCTACA-3=) and iNOS
(forward primer, 5=-CAAGTACGGCCGCTTCGA-3=; reverse primer, 5=-CACTCGTATTTGGGATGTTCCA-3=).
For each sample assayed, the threshold value (CT) for
target genes and glyceraldehyde-3-phosphate dehydrogenase (internal reference) was determined. All assays were performed in triplicate.
WB Analysis and Antibodies
Small-bowel (SB) epithelial cells were isolated by the
EDTA method, depleted on sheep anti-rat IgG magnetic
Dynabeads (Life Technologies, Grand Island, NY) preloaded with rat anti-mouse CD45 antibodies, and homogenized in radioimmunoprecipitation assay buffer containing
protease and phosphatase inhibitor cocktail (GBioscience).15 Homogenates were centrifuged at 15,700 ⫻ g for
30 minutes. Proteins were separated by SDS-PAGE using
8% to 16% precast gels (Lonza, Basel, Switzerland) and
transferred to polyvinylidene difluoride membrane (EMD
Millipore, Billerica, MA). The membranes were blocked with
protein-free T20 blocking buffer (Thermo Scientific, Rockford, IL) and incubated with primary antibodies specific for
iNOS (BD Transduction Laboratories, San Jose, CA), p53
and nitrotyrosine (Cell Signaling), p-p53 (Ser 15; Santa Cruz
Biotechnology Inc., Santa Cruz, CA), caspase 3 (cleaved
fragment 17, 19 kDa; proform, 35 kDa; Cell Signaling, Danvers, MA), caspase 9 (37 kDa; Cell Signaling), and caspase
8 (20 kDa; Abcam, Cambridge, MA), followed by a corresponding anti-rabbit or anti-mouse IgG (Pierce Protein Research Products, Rockford, IL). The membranes were
stripped and probed with -actin (Sigma-Aldrich, St. Louis,
MO) as a loading control. Proteins were detected by chemiluminescence (West Pico or West Dura kits; Pierce) on
autoradiography film digitally scanned for quantification by
densitometry using Adobe Photoshop (Adobe Systems Inc,
San Jose, CA) analysis tools. Each Western blot (WB) was
repeated at least three times.
Human Colonic Specimens and Histological
Score
Biopsy specimens were obtained from human patients
aged ⱖ18 years, undergoing diagnostic or surveillance
colonoscopy for UC or therapeutic colonic resection, or
healthy individuals undergoing routine colon cancer surveillance. Exclusion criteria were pregnant women, history
of intestinal surgery, bleeding diathesis, or coagulopathy.
Inflammation was scored by a blinded researcher (P.S.) on
a scale from 0 to 8, based on mucosal leukocyte infiltration
(0, no infiltration; 1, basolateral; 2, infiltration halfway up the
crypt; 3, diffuse infiltration; and 4, crypt abscess) added to
a crypt architecture score (0, no epithelial cell distortion; 1,
crypt hyperplasia; 2, mild crypt distortion; 3, severe crypt
distortion; and 4, complete loss of crypt structure). All untreated and anti-TNF–treated patients were inflamed and
had a mean histological score greater than four. A total of
six specimens from six patients were analyzed for each
group. Collection of all patient materials for this study was
approved by Northwestern University’s Office for the Protection of Human Subjects.
Statistical Analysis
A two-tailed Student’s t-test was used to evaluate differences between the groups. For any single experiment, up
to five statistical comparisons were made. Bonferroni correction for multiple comparisons results in differences
being considered statistically significant when P ⬍ 0.01.
This would control the overall type I error rate for an
experiment at 5%.
Results
TNF-Mediated Crypt Cell Apoptosis Is TNFR1
and TNFR2 Dependent
To examine the relative contribution of TNFR1 and TNFR2
signaling to T-cell–induced IEC apoptosis, WT, TNFR1⫺/⫺,
TNFR2⫺/⫺, or TNFR1/2⫺/⫺ mice, we stimulated with antiCD3 mAb to activate T cells. Researchers reported that
treating mice with anti-CD3 increases intestinal (epithelial
and lamina propria) and serum levels of cytokines, including TNF.26 –29 At 24 hours after injection, TUNEL staining of
the SB of WT mice indicated that T-cell activation induced
IEC apoptosis in lower to mid crypt regions (Figure 1A). By
comparison, epithelial cell apoptosis was reduced in mice
deficient for TNFR1 or TNFR2 (Figure 1A). Although individual TNFR1 or TNFR2 deletions reduced apoptotic responses by approximately 45%, mice with combined
TNFR1 and TNFR2 deficiencies exhibited 60% less apoptosis (Figure 1B). Thus, both TNFR1 and TNFR2 contributed
to T-cell–mediated IEC apoptosis in the SB.
Next, caspase 3, 8, and 9 activation was assessed in
IECs as an indicator of intrinsic and extrinsic apoptotic
signaling pathways. The WB analysis of IEC isolates from
control and anti-CD3-treated mice revealed that T-cell
activation induced IEC caspase 3 cleavage 30-fold in WT
mice (Figure 1C). Densitometry showed that caspase 3
cleavage persisted from 3 to 12 hours after treatment,
consistent with times when proteins became nitrated (see
Supplemental Figure S1A at http://ajp.amjpathol.org)26,30
and with times we reported that anti-CD3 induced TNF
locally.26 By comparison, TNFR deficiency largely attenuated caspase 3 cleavage at every time point. The results
of caspase 9 cleavage paralleled those of caspase 3
reductions seen in TNFR-deficient mice (Figure 1C; see
also Supplemental Figure S1B at http://ajp.amjpathol.org).
In contrast, IEC caspase 8 cleavage (p20 protein shown
herein) was unaffected by T-cell activation. Together,
these data indicated that both TNFR1 and TNFR2 signal-
Epithelial Cell Apoptosis in IBD
1309
AJP October 2012, Vol. 181, No. 10
B
A
WT
5
WT + anti-CD3
WT
**
Apoptotic Index
-/TNFR1
TNFR1-/-
4
-/TNFR2
TNFR2-/-/TNFR1/2
TNFR1/2-/-
3
* *
*
2
1
TNFR1/2-/-
TNFR1/2-/- + anti-CD3
0
Untreated
C
TNFR1-/- + anti-CD3
TNFR2-/- + anti-CD3
WT
TNFR1-/-
Anti-CD3
cleaved
TNFR2-/-
TNFR1/2-/- caspase:
17
3
20
8
37
9
0
3
6 12
0 3
6 12
0 3
6
12
0
3
6 12
Anti-CD3, hrs
Figure 1. TNFR1 and TNFR2 signaling induces TNF-mediated crypt cell apoptosis. WT, TNFR1⫺/⫺, TNFR2⫺/⫺, and TNFR1/2⫺/⫺ mice are injected with control
or anti-CD3 antibody and sacrificed at the indicated time points. A: TUNEL staining of SB crypts from mice sacrificed 24 hours after anti-CD3 mAb injection. B:
The apoptotic index is determined based on counting TUNEL-positive cells. *P ⬍ 0.0001 compared with WT stimulated mice; **P ⬍ 0.0001 for anti-CD3-treated
WT mice compared with untreated WT mice. C: WB of activated caspases 3, 8, and 9 from CD45-negative (data not shown) IEC lysates resolved by SDS-PAGE.
-Actin is used as a loading control.
ing mediated IEC apoptosis via caspases 3 and 9. Not all
apoptotic activity was abrogated by TNFR deficiency.
These data suggested that, although TNFR signaling was
important for IEC apoptosis, other pathways contributed
as well (eg, Fas/FasL and perforin).31
TNF-Induced iNOS Mediated Immune-Mediated
Crypt Cell Apoptosis
Next, we tested the hypothesis that TNF-induced iNOS
contributed to T-cell–induced epithelial apoptosis. TNF
and iNOS mRNA levels in anti-CD3-stimulated TNFR1/2⫺/⫺
and iNOS⫺/⫺ mice were assessed. The expression of
iNOS was significantly reduced in TNFR1/2⫺/⫺ mice relative to WT, whereas TNF levels were induced in WT,
TNFR1/2⫺/⫺, and iNOS⫺/⫺ mice (Figure 2A). WB analysis
revealed that iNOS protein levels increased within 3
hours of T-cell activation in WT mice (Figure 2B). By
comparison, iNOS induction was significantly reduced in
the single and double TNF receptor knockout mice (Figure 2B). Therefore, TNFR1 and TNFR2 signaling induced
iNOS after T-cell activation.
Next, we determined whether iNOS mediated T-cell–
induced IEC apoptosis. Data shown in Figure 3A indicate
that IEC apoptosis levels were significantly reduced
when iNOS was blocked either pharmacologically with
the selective inhibitor L-NIL or genetically using iNOS
deficient relative to WT control mice. Furthermore, activation of caspases 3 and 9 was attenuated in iNOS⫺/⫺
mice compared with anti-CD3-treated WT mice (Figure
3B). Procaspase 3 protein levels were induced by immune activation in WT mice and in iNOS⫺/⫺ mice, albeit
to lesser degrees over time. Therefore, procaspase 3
expression may be regulated by inflammatory mediators
in IECs. Caspase 8 activation (p20 activated form) remained unchanged at every time point in IECs from both
WT and iNOS⫺/⫺ mice. Taken together, these data suggested that TNF-induced iNOS mediated IEC apoptosis
after T-cell activation.
TNFR1, TNFR2, and iNOS-Induced IEC
Apoptosis Is p53 Dependent
Data in other tissues, such as the brain, pancreas, and
heart, suggested that iNOS-induced nitrosative oxygen
species induced p53 protein stabilization, which then
induced apoptosis.32 We recently reported that p53 was
an important mediator of inflammation-induced apoptosis
in the colon; however, the upstream mechanisms remained unknown.24 Data herein suggested that both
TNFR1 and TNFR2 contributed to IEC apoptosis via the
intrinsic pathway; thus, we examined p53 protein levels in
IECs after anti-CD3 treatment. Data in Figure 4 showed
that T-cell activation induced a progressive increase in
p53 from 3 to 12 hours in the SB of WT mice. By comparison, p53 stabilization was severely attenuated in TNF
receptor or iNOS-deficient mice (Figure 4A). These results were consistent with TNF and iNOS being upstream
of p53 protein accumulation.
1310
Goretsky et al
AJP October 2012, Vol. 181, No. 10
ND
TNFR1/2-/-
WT
iNOS-/-
B
WT
Anti-CD3, hrs: 0
3
0
3
0 3
130
0
3
iNOS
β-actin
Figure 2. iNOS mediates T-cell–induced crypt cell apoptosis downstream of
TNFR1 and TNFR2. A: Fold induction of iNOS and TNF mRNA expression 3
hours after anti-CD3 injection determined by real-time RT-PCR from SB tissue
from WT, TNFR1/2⫺/⫺, and iNOS⫺/⫺ mice. B: WB analysis of iNOS protein
expression in the indicated mice before, or 3 hours after, T-cell activation.
-Actin is used as a loading control. ND, not determined.
Because T-cell activation enhanced p53 levels, a role
for p53 in IEC apoptosis was explored by TUNEL staining. Although IEC apoptosis increased in lower to mid
crypts of tissue after T-cell activation, TUNEL staining of
IECs was reduced approximately 89% in p53⫺/⫺ mice
(Figure 4, B and C). WB analysis confirmed p53-dependent IEC apoptosis because caspase 3 and 9 activation
was significantly attenuated in p53⫺/⫺ mice compared
with WT (Figure 4D). Taken together, these data were
consistent with the notion that p53 mediated IEC death in
response to TNF and iNOS signaling.
TNF and iNOS Are Required for
Colitis-Induced IEC Apoptosis
To determine the relevance of these findings to IBD colitis, IEC apoptosis was assessed in IL-10⫺/⫺ mice. Colitis
was induced by feeding mice piroxicam for 14 days.33,34
TUNEL staining revealed that, on the day of peak colitis
(day 28), apoptosis increased within IECs localized to
lower crypt regions (Figure 5A). To determine whether
iNOS or TNF signaling contributed to colitis-induced
apoptosis, mice were treated with either aminoguanidine
(a specific iNOS inhibitor) or anti-TNF antibody, respectively. TUNEL staining revealed that apoptotic indexes
decreased by 58% in aminoguanidine-treated mice and
37% in anti-TNF–treated mice, compared with untreated
colitic mice (Figure 5, A and B). Short-term blocking of
TNF by anti-TNF treatment was also effective at reducing
apoptosis in the anti-CD3 model, as determined by
TUNEL staining (see Supplemental Figure S2A at http://
ajp.amjpathol.org). Analysis of cleaved caspases 3 and 9
in colitic mice confirmed that anti-TNF or aminoguanidine
treatment decreased IEC apoptosis in colitic mice (Figure
5C). The expression of caspase 8 active polypeptide
(p20) remained unchanged. Quantitative analysis of TNFinduced iNOS mRNA expression revealed that anti-TNF
treatment of mice reduced downstream effects of TNF
and, thus, colitis-induced epithelial iNOS gene transcription (see Supplemental Figure S2B at http://ajp.amjpathol.
org). These data were also confirmed in another chronic
colitis model. Mice that underwent three cycles of DSS in
their drinking water revealed that anti-TNF reduced expression of relevant gene targets (TNF, mmp7, and iNOS;
see Supplemental Figure S2C at http://ajp.amjpathol.org).
Furthermore, anti-TNF abrogated elevated levels of epithelial cleaved caspases 3 and 9 in mice with chronic
DSS-colitis (Figure 5D). Together, these data suggested
that TNF and iNOS mediated IEC apoptosis in colitis.
To determine whether iNOS and TNF regulated IEC
p53 levels in colitis, IEC lysates were analyzed by WB.
Cytoplasmic p53 levels were much higher with colitis
(Figure 5C). Significantly, anti-TNF and aminoguanidine
treatment reduced p53 accumulation in the cytoplasm.
These data were further supported by findings in threecycle DSS-treated mice in which anti-TNF treatment
abrogated colitis-induced IEC p53 stabilization (Figure
5D). In summary, these data illustrated that TNF-induced p53-mediated signaling accounted for IEC
apoptosis in colitis.
A
Apoptotic Index
A
5
Control
Anti-CD3
4
*
3
2
*
1
0
L-NIL:
- WT
B
+ +
WT
- iNOS-/iNOS-/-
WT
37
cleaved caspase 9
20
cleaved caspase 8
35
procaspase 3
17
cleaved caspase 3
42
β-actin
0
3
6
12
0
3
6
12
Anti-CD3, hrs
Figure 3. iNOS is required for IEC apoptosis. A: Mice are sacrificed 24 hours
after control or anti-CD3 mAb injection, and apoptosis is quantified in SB
sections from WT mice, with or without L-NIL treatment, and from iNOS⫺/⫺
mice. Apoptotic indexes are calculated based on counting TUNEL-positive
cells. *P ⬍ 0.001 compared with anti-CD3-treated WT mice. B: Activation of
caspases 3, 8, and 9 is assessed by WB for the cleaved forms of the indicated
protein in WT and iNOS⫺/⫺ mice at 0, 3, 6, or 12 hours after T-cell activation.
Procaspase 3 is also shown, and -actin is used as a loading control.
Epithelial Cell Apoptosis in IBD
1311
AJP October 2012, Vol. 181, No. 10
A
iNOS-/-
TNFR-1/2-/-
WT
53
p53
β-actin
42
6
Untreated
12
0
anti-CD3
3
6
12
0
3
6 12
C
WT
B
3
Apoptotic index
Anti-CD3, hrs: 0
6
Control
Anti-CD3
5
4
3
2
1
*
0
p53-/-
B6
D
cleaved
caspase:
p53-/-
WT
p53-/-
17
3
37
9
8
20
β-actin
0
3
6
12
0
3
6
12
Anti-CD3, hrs:
Figure 4. p53 Plays a role in T-cell–induced crypt cell apoptosis downstream of TNFR1, TNFR2, and iNOS. A: WB analysis of p53 protein expression and
stabilization in SB IECs from WT, TNFR1/2⫺/⫺, and iNOS⫺/⫺ mice. B: Apoptosis is assessed in the SB crypts and villi in p53⫺/⫺ mice by TUNEL staining 24 hours
after stimulation with anti-CD3 mAb injection relative to WT mice. Insets: Areas magnified (original magnification, ⫻40) are demarcated by boxes in the ⫻10
picture. C: Results are quantified by counting TUNEL-positive cells to determine the apoptotic index. *P ⬍ 0.0001. D: Caspase activation is determined by WB
analysis at the indicated times after anti-CD3 injection. -Actin is used as a loading control.
IEC Apoptosis and p53 Expression Are TNF
Dependent in Human UC
To examine a role for TNF to induce p53-mediated IEC
apoptosis in human UC, IEC apoptosis and p53 staining
were compared between control tissue and actively inflamed colitis tissue from untreated patients or those receiving anti-TNF therapy. All UC samples chosen for this
study had moderately active inflammation, as determined
by histological scores (Figure 6; see also Materials and
Methods). Thus, levels of tissue inflammation were similar
between untreated and anti-TNF–treated tissue. Patients
undergoing routine surveillance colonoscopy rarely displayed TUNEL- or p53-positive cells (Figure 6). Apoptotic
cells were seen primarily in plateau regions, whereas p53
was detected in lower crypts. Strikingly, colitis increased
IEC apoptosis and p53 staining throughout lower and mid
crypt regions (Figure 6). By comparison, IEC apoptosis
and p53 staining were markedly reduced in patients
treated with anti-TNF pharmacological agents. These
data indicated that, despite equivalent levels of inflammation, anti-TNF Ab treatment reduced p53-mediated
IEC apoptosis in human UC (Figure 6D).
Discussion
The intestinal epithelium provides essential roles important for host defense and homeostasis, including maintaining barrier function and participating in mucosal immune responses.1,2 Regulation of apoptosis of IECs is
one way that the intestinal epithelium maintains or returns to homeostasis.35 During IBD, apoptosis is observed in acute inflammatory sites.35,36 Therapy with
TNF-neutralizing antibody effectively reduces IEC
apoptosis and increases mucosal repair; however, the
mechanisms for TNF-mediated tissue injury in IBD remain unclear.37,38 Recent findings from our laboratory
indicate that p53 is the predominant mediator of IEC
apoptosis in IBD.24 The data presented herein suggest
that TNF-induced iNOS stabilizes p53, which induces
crypt IEC apoptosis (Figure 6D).
We show that p53 is a major mediator of inflammationinduced crypt IEC apoptosis. Previous reports showed
elevated levels of p53 in patients with UC.15,16,24,39 Most
significantly, we report a role for p53 in both an acute and
chronic inflammatory model in mice that parallels these
observations made from studying human disease and is,
thus, clinically relevant. First, p53 levels were elevated in
anti-CD3-treated and IL-10⫺/⫺ mouse colitis models (Figures 1 and 5). Second, p53 deficiency reduced IEC
apoptosis, as measured by TUNEL staining and WB of
activated caspase 3 and 9 cleavage (Figure 4). Finally,
regions of elevated p53 staining in patients with UC correlated with increased IEC apoptosis, and regions of
reduced p53 paralleled reductions in IEC apoptosis after
anti-TNF therapy (Figure 6). Taken together, these data
further implicate p53 as a key player in IBD crypt IEC
apoptosis.
Nitrosylated oxygen radicals produced by iNOS are
major inducers of p53 activation and stabilization.40 In
1312
Goretsky et al
AJP October 2012, Vol. 181, No. 10
A
IL-10-/WT, Untreated
B
Apoptotic Index
5
IL-10-/-
aminoguanidine
Untreated
C
*
cleaved
caspase:
37
9
4
*
3
*
2
1
0
Colitis
aminoguanidine
Anti-TNF
-
+
-
+
+
-
+
+
IL-10-/-
Anti-TNF
D
DSS
WT
Anti-TNF
-
-
+
cleaved
caspase:
9
20
8
37
17
3
17
3
53
p53
53
p53
42
β-actin
42
β-actin
Colitis
aminoguanidine
Anti-TNF
-
+
-
+
+
+
+
-
Figure 5. Inhibitors of TNF and iNOS decrease colitis-induced IEC apoptosis. Colitis is induced in IL-10⫺/⫺ mice with piroxicam, and apoptosis is assessed at
day 28. Mice are treated with aminoguanidine to block iNOS or anti-TNF mAb to inhibit TNF. Colonic IECs undergoing apoptosis are identified by TUNEL staining
(A, arrows), and the apoptotic index is calculated based on cell counting of TUNEL-positive cells (B). *P ⬍ 0.004 compared with colitic mice; **P ⬍ 0.0001
compared with noncolitic mice. High-power magnification (⫻40) is shown in the small boxes in the low-magnification (⫻10) pictures. C: WB analysis of colon
IEC whole cell lysates for caspase 3, 8, and 9 activation and for p53 levels. Chronic colitis is also induced in WT mice after three cycles of DSS. D: Mice are either
left untreated or treated with anti-TNF mAb, and levels of p53 and cleaved caspases 3 and 9 are analyzed. -Actin is used as a loading control.
human IBD, iNOS is observed in inflamed areas of the
colon in epithelial cells, lamina propria mononuclear
cells, and neutrophils.14,15 Reductions of iNOS mRNA
and protein in mice with T-cell–stimulated TNFR1/2⫺/⫺
and anti-TNF–treated colitis suggest that TNF is a major
mediator of iNOS in intestinal inflammation. Data presented herein in two mouse models directly implicate
iNOS in inflammation-induced IEC apoptosis. IEC apoptosis was reduced in iNOS-deficient mice (Figure 3) and
in mice treated with the specific inhibitors, L-NIL or aminoguanidine (Figure 5). Furthermore, p53 protein stabilization was attenuated in iNOS⫺/⫺ mice treated with anti-CD3
and in colitic IL-10⫺/⫺ mice treated with aminoguanidine.
These data confirm that NO⫺ and ONOO⫺ actively participate in p53 stabilization. In fact, Singer et al14 previously
demonstrated markedly elevated nitrotyrosine staining (a
product of ONOO⫺) in IECs from patients with IBD. Thus,
the effects of ONOO⫺ likely directly affected p53 stabilization within IECs. Inhibition of p53 stabilization and caspase
activation by L-NIL and aminoguanidine was incomplete.
These data suggest that iNOS-independent mechanisms
may also have some role in inflammation-induced apoptosis
of IECs.
Our data indicate that inflammation-induced TNF is an
important inducer of p53-dependent IEC apoptosis. Previously, expression of both TNFR1 and TNFR2 was increased on IECs during inflammatory conditions.41,42 Our
analysis reveals that signaling through both receptors
contributes to inflammation-induced IEC apoptosis because apoptosis was decreased in the single- and double-receptor knockouts after T-cell activation and by anti-
TNF treatment in the IL-10⫺/⫺ chronic colitis model.
These data are consistent with reports showing that
TNFR1 and TNFR2 cooperate in TNF-mediated apoptosis
and also form functional heterocomplexes.19,43– 45 Most
significantly, TNF neutralization in patients with UC reduced IEC apoptosis and p53 staining (Figure 6, A–C).
These data indicate a direct role for TNF in IEC apoptosis
through p53-mediated pathways (Figure 6D).
In our experiments, data suggest that the mitochondrial intrinsic pathway mediated inflammation-induced
IEC apoptosis. We consistently saw, in murine models,
that inflammation induced procaspase 3 and 9, but not 8,
cleavage into their active forms. Caspase 8 primarily
induces apoptotic death initiated by receptors containing
death domains (eg, TNFR1, Fas, and DR5).46 Its activation, for example, mediates caspase 3 and BH3 interacting-domain death agonist cleavage, leading to the execution of apoptosis. Therefore, the absence of caspase 8
cleavage suggests that although TNFR1 was required, it
did not induce caspases linked to the extrinsic pathway.
Rather, our data indicate that TNFR-mediated IEC apoptosis required activation of the intrinsic or mitochondrial
pathway. Mitochondrial dysfunction initiates apoptosis by
the intrinsic apoptotic pathway and includes p53 translocation to the nucleus, where the protein modulates proand anti-apoptotic Bcl-2 family proteins.47 This pathway
is tightly regulated by a balance between prosurvival and
pro-apoptotic Bcl-2 family members.48 Some evidence
suggests that p53 requires apoptotic protease activating
factor 1, caspase-9, and cytochrome c release to perform
apoptosis.49 Thus, the intrinsic pathway is vital for p53-
Epithelial Cell Apoptosis in IBD
1313
AJP October 2012, Vol. 181, No. 10
A
UC, untreated
UC, anti-TNF treated
p53
TUNEL
Control
*
0.4
Mean histology
score
6
4
2
0.3
0
WT
Un ni afl mmedc ontro l
0.2
*
0.1
0
WT
UC,
UC,
untreated anti-TNF
UC-u ntre ate d
UC-a ntiT NF
C
p53 positive cells/100 IEC
Apoptotic Index
(positive TUNEL/100 IEC)
B
*
0.4
Mean histology
score
6
4
0.3
D
Chronic UC
Anti-TNF
TNF
mesalamine
iNOS
2
0
0.2
WT
Un ni alf mmedc ontro l
UC-u ntre ate d
UC-a ntiT NF
p53
0.1
*
0.0
WT
UC,
UC,
untreated anti-TNF
IEC crypt
apoptosis
Figure 6. Apoptosis and p53 expression are induced by TNF in human UC. Colon tissue biopsy specimens from normal control, UC untreated, and UC
anti-TNF–treated patients are analyzed for p53 and TUNEL staining (A, arrows) and then quantified by counting positive cells per 100 IECs (B and C). Insets:
Areas magnified (⫻40) are demarcated by boxes. Untreated and anti-TNF–treated UC patients selected for study are inflamed, as indicated by mean histological
scores (B and C). *P ⬍ 0.01 (B), P ⬍ 0.0001 (C) for UC untreated compared to anti-TNF–treated patients. The proposed paradigm for IEC crypt apoptosis, in which
chronic inflammation from UC induces TNF production, which causes increased expression of iNOS. The effects of iNOS products (NO⫺ and ONOO⫺) induce
p53 protein in crypt epithelial cells. Points in the pathway where the therapeutic agents, anti-TNF and mesalamine, may inhibit IEC apoptosis are also shown.
dependent apoptosis and tumor suppression. It was previously reported that TNF-induced IEC apoptosis was
only TNFR1 dependent and p53 independent, results that
contradict those reported herein.50 It is possible that the
epithelial populations undergoing apoptosis in these
studies differed from those examined herein. Piguet et
al50 induced villus IEC apoptosis and detachment by a
relatively high-dose TNF injection; however, in our experiments, we observed the increase in apoptosis in IECs in
the lower crypt regions, as seen in patients with human
IBD. Because villus IEC apoptosis occurs after ischemia
reperfusion, their studies likely present a valid model for
this mode of IEC death. Conversely, the models used
herein result in crypt IEC death and, therefore, more
closely resemble mechanisms of IEC apoptosis in IBD.
One clinical implication of these studies is that commonly used therapeutic agents that block TNF (anti-TNF
monoclonal antibody) or iNOS (mesalamine) may enhance mucosal healing by reducing IEC apoptosis.7,51
Our studies show that TNF induces iNOS, which activates
p53 in epithelial cells (Figure 6D). These mediators, increased in IBD, induce IEC apoptosis by activating p53
in lower and mid crypt zones, where proliferation is induced. Thus, we strongly believe that IEC apoptosis in
the lower and mid crypts is most relevant to the increased
epithelial cell death seen in human IBD. It is, therefore,
attractive to speculate that p53-mediated signaling induces apoptosis in proliferating progenitor populations.
In biopsy specimens from patients with UC, we failed to
see a correlation between p53 status (in a sample from
anti-TNF–treated patients) and histological activity. In
other reports,52,53 investigators found that mucosal inflammation was unaffected by p53 status; thus, the predominant effect of p53 function and IEC apoptosis may
be on the development of dysplasia. In this scenario, a
failure to induce apoptosis might allow for a long-lived
progenitor cell population harboring DNA mutations to
persist.
Chronic colitis induced in mice by DSS alone initiates
the neoplastic process,54 as does colitis in IL-10⫺/⫺
mice, suggesting that inflammation plays a key role in
colitis-associated cancer development.54,55 Mutation
of the tumor suppressor gene, p53, is an early event in
the progression toward human colitis-associated can-
1314
Goretsky et al
AJP October 2012, Vol. 181, No. 10
cer.16,56 –59 We show herein that stabilization of epithelial
p53 after immune activation is TNFR1/2 and iNOS dependent. Without p53 function, as would be the case after
inflammation-induced p53 mutation, damaged proliferating cells would avoid apoptosis. Chen et al60 showed
clonal expansion of p53-mutated epithelial cells in areas
of colitis-induced dysplasia. The failure of p53-mutated
IECs to undergo apoptosis may explain why IEC apoptosis is reduced in colonic tumor samples and carcinomas.35 In fact, loss of p53 enhanced cancer rates in mice
with DSS colitis.52,61 Additional exploration of p53-deficient mice in chronic colitis models will further discern the
role p53 plays in IBD-induced oncogenesis.
References
1. Shen L, Turner JR: Role of epithelial cells in initiation and propagation
of intestinal inflammation: eliminating the static: tight junction dynamics exposed. Am J Physiol Gastrointest Liver Physiol 2006, 290:G577–
G582
2. Groschwitz KR, Hogan SP: Intestinal barrier function: molecular regulation and disease pathogenesis. J Allergy Clin Immunol 2009, 124:
3–20; quiz 21–22
3. Arseneau KO, Tamagawa H, Pizarro TT, Cominelli F: Innate and
adaptive immune responses related to IBD pathogenesis. Curr Gastroenterol Rep 2007, 9:508 –512
4. Bouma G, Strober W: The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol 2003, 3:521–533
5. Suenaert P, Bulteel V, Lemmens L, Noman M, Geypens B, Van
Assche G, Geboes K, Ceuppens JL, Rutgeerts P: Anti-tumor necrosis
factor treatment restores the gut barrier in Crohn’s disease. Am J
Gastroenterol 2002, 97:2000 –2004
6. Wong M, Ziring D, Korin Y, Desai S, Kim S, Lin J, Gjertson D, Braun
J, Reed E, Singh RR: TNFalpha blockade in human diseases: mechanisms and future directions. Clin Immunol 2008, 126:121–136
7. Tilg H, Moschen A, Kaser A: Mode of function of biological anti-TNF
agents in the treatment of inflammatory bowel diseases. Expert Opin
Biol Ther 2007, 7:1051–1059
8. Lichtenberger GS, Flavell RA, Alexopoulou L: Innate immunity and
apoptosis in IBD. Inflamm Bowel Dis 2004, 10(Suppl 1):S58 –S62
9. Podolsky DK: Inflammatory bowel disease. N Engl J Med 2002,
347:417– 429
10. Parameswaran N, Patial S: Tumor necrosis factor-␣ signaling in macrophages. Crit Rev Eukaryot Gene Expr 2010, 20:87–103
11. Kleinert H, Schwarz PM, Förstermann U: Regulation of the expression
of inducible nitric oxide synthase. Biol Chem 2003, 384:1343–1364
12. Rachmilewitz D, Karmeli F, Okon E, Bursztyn M: Experimental colitis
is ameliorated by inhibition of nitric oxide synthase activity. Gut 1995,
37:247–255
13. Krieglstein CF, Cerwinka WH, Laroux FS, Salter JW, Russell JM,
Schuermann G, Grisham MB, Ross CR, Granger DN: Regulation of
murine intestinal inflammation by reactive metabolites of oxygen and
nitrogen: divergent roles of superoxide and nitric oxide. J Exp Med
2001, 194:1207–1218
14. Singer II, Kawka DW, Scott S, Weidner JR, Mumford RA, Riehl TE,
Stenson WF: Expression of inducible nitric oxide synthase and nitrotyrosine in colonic epithelium in inflammatory bowel disease. Gastroenterology 1996, 111:871– 885
15. Hofseth LJ, Saito S, Hussain SP, Espey MG, Miranda KM, Araki Y,
Jhappan C, Higashimoto Y, He P, Linke SP, Quezado MM, Zurer I,
Rotter V, Wink DA, Appella E, Harris CC: Nitric oxide-induced cellular
stress and p53 activation in chronic inflammation. Proc Natl Acad Sci
U S A 2003, 100:143–148
16. Hussain SP, Amstad P, Raja K, Ambs S, Nagashima M, Bennett WP,
Shields PG, Ham AJ, Swenberg JA, Marrogi AJ, Harris CC: Increased
p53 mutation load in noncancerous colon tissue from ulcerative
colitis: a cancer-prone chronic inflammatory disease. Cancer Res
2000, 60:3333–3337
17. Gupta S: Molecular steps of tumor necrosis factor receptor-mediated
apoptosis. Curr Mol Med 2001, 1:317–324
18. Aggarwal BB: Signalling pathways of the TNF superfamily: a doubleedged sword. Nat Rev Immunol 2003, 3:745–756
19. Wallach D, Varfolomeev EE, Malinin NL, Goltsev YV, Kovalenko AV,
Boldin MP: Tumor necrosis factor receptor and Fas signaling mechanisms. Annu Rev Immunol 1999, 17:331–367
20. Iwamoto M, Koji T, Makiyama K, Kobayashi N, Nakane PK: Apoptosis
of crypt epithelial cells in ulcerative colitis. J Pathol 1996, 180:152–
159
21. Di Sabatino A, Ciccocioppo R, Luinetti O, Ricevuti L, Morera R, Cifone
MG, Solcia E, Corazza GR: Increased enterocyte apoptosis in inflamed areas of Crohn’s disease. Dis Colon Rectum 2003, 46:1498 –
1507
22. Lee G, Goretsky T, Managlia E, Dirisina R, Singh AP, Brown JB, May
R, Yang GY, Ragheb JW, Evers BM, Weber CR, Turner JR, He XC,
Katzman RB, Li L, Barrett TA: Phosphoinositide 3-kinase signaling
mediates beta-catenin activation in intestinal epithelial stem and progenitor cells in colitis. Gastroenterology 2010, 139:869 – 881,
881.e1-9
23. Musch MW, Clarke LL, Mamah D, Gawenis LR, Zhang Z, Ellsworth W,
Shalowitz D, Mittal N, Efthimiou P, Alnadjim Z, Hurst SD, Chang EB,
Barrett TA: T cell activation causes diarrhea by increasing intestinal
permeability and inhibiting epithelial Na⫹/K⫹-ATPase. J Clin Invest
2002, 110:1739 –1747
24. Dirisina R, Katzman RB, Goretsky T, Managlia E, Mittal N, Williams
DB, Qiu W, Yu J, Chandel NS, Zhang L, Barrett TA: p53 and PUMA
independently regulate apoptosis of intestinal epithelial cells in patients and mice with colitis. Gastroenterology 2011, 141:1036 –1045
25. Gavrieli Y, Sherman Y, Ben-Sasson SA: Identification of programmed
cell death in situ via specific labeling of nuclear DNA fragmentation.
J Cell Biol 1992, 119:493–501
26. Tang Y, Clayburgh DR, Mittal N, Goretsky T, Dirisina R, Zhang Z, Kron
M, Ivancic D, Katzman RB, Grimm G, Lee G, Fryer J, Nusrat A, Turner
JR, Barrett TA: Epithelial NF-kappaB enhances transmucosal fluid
movement by altering tight junction protein composition after T cell
activation. Am J Pathol 2010, 176:158 –167
27. Ferran C, Sheehan K, Dy M, Schreiber R, Merite S, Landais P, Noel
LH, Grau G, Bluestone J, Bach JF, Chatenoud L: Cytokine-related
syndrome following injection of anti-CD3 monoclonal antibody: further
evidence for transient in vivo T cell activation. Eur J Immunol 1990,
20:509 –515
28. Radojevic N, McKay DM, Merger M, Vallance BA, Collins SM, Croitoru
K: Characterization of enteric functional changes evoked by in vivo
anti-CD3 T cell activation. Am J Physiol 1999, 276:R715–R723
29. Clayburgh DR, Barrett TA, Tang Y, Meddings JB, Van Eldik LJ,
Watterson DM, Clarke LL, Mrsny RJ, Turner JR: Epithelial myosin light
chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo. J Clin Invest 2005, 115:2702–2715
30. Pacher P, Beckman JS, Liaudet L: Nitric oxide and peroxynitrite in
health and disease. Physiol Rev 2007, 87:315– 424
31. Merger M, Viney JL, Borojevic R, Steele-Norwood D, Zhou P, Clark
DA, Riddell R, Maric R, Podack ER, Croitoru K: Defining the roles of
perforin, Fas/FasL, and tumour necrosis factor alpha in T cell induced
mucosal damage in the mouse intestine. Gut 2002, 51:155–163
32. Xu W, Liu LZ, Loizidou M, Ahmed M, Charles IG: The role of nitric
oxide in cancer. Cell Res 2002, 12:311–320
33. Berg DJ, Zhang J, Weinstock JV, Ismail HF, Earle KA, Alila H, Pamukcu R, Moore S, Lynch RG: Rapid development of colitis in NSAIDtreated IL-10-deficient mice. Gastroenterology 2002, 123:1527–1542
34. Brown JB, Lee G, Managlia E, Grimm GR, Dirisina R, Goretsky T,
Cheresh P, Blatner NR, Khazaie K, Yang GY, Li L, Barrett TA: Mesalamine inhibits epithelial beta-catenin activation in chronic ulcerative
colitis. Gastroenterology 2010, 138:595– 605
35. Edelblum KL, Yan F, Yamaoka T, Polk DB: Regulation of apoptosis
during homeostasis and disease in the intestinal epithelium. Inflamm
Bowel Dis 2006, 12:413– 424
36. Hagiwara C, Tanaka M, Kudo H: Increase in colorectal epithelial
apoptotic cells in patients with ulcerative colitis ultimately requiring
surgery. J Gastroenterol Hepatol 2002, 17:758 –764
37. Zeissig S, Bojarski C, Buergel N, Mankertz J, Zeitz M, Fromm M,
Schulzke JD: Downregulation of epithelial apoptosis and barrier repair in active Crohn’s disease by tumour necrosis factor alpha antibody treatment. Gut 2004, 53:1295–1302
38. Marini M, Bamias G, Rivera-Nieves J, Moskaluk CA, Hoang SB, Ross
WG, Pizarro TT, Cominelli F: TNF-alpha neutralization ameliorates the
Epithelial Cell Apoptosis in IBD
1315
AJP October 2012, Vol. 181, No. 10
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
severity of murine Crohn’s-like ileitis by abrogation of intestinal
epithelial cell apoptosis. Proc Natl Acad Sci U S A 2003,
100:8366 – 8371
Alkim C, Savas B, Ensari A, Alkim H, Dagli U, Parlak E, Ulker A, Sahin
B: Expression of p53, VEGF, microvessel density, and cyclin-D1 in
noncancerous tissue of inflammatory bowel disease. Dig Dis Sci
2009, 54:1979 –1984
Goodman JE, Hofseth LJ, Hussain SP, Harris CC: Nitric oxide and
p53 in cancer-prone chronic inflammation and oxyradical overload
disease. Environ Mol Mutagen 2004, 44:3–9
Kaiser GC, Polk DB: Tumor necrosis factor alpha regulates proliferation in a mouse intestinal cell line. Gastroenterology 1997, 112:
1231–1240
Mizoguchi E, Mizoguchi A, Takedatsu H, Cario E, de Jong YP, Ooi CJ,
Xavier RJ, Terhorst C, Podolsky DK, Bhan AK: Role of tumor necrosis
factor receptor 2 (TNFR2) in colonic epithelial hyperplasia and chronic
intestinal inflammation in mice. Gastroenterology 2002, 122:134 –144
Tartaglia LA, Pennica D, Goeddel DV: Ligand passing: the 75-kDa
tumor necrosis factor (TNF) receptor recruits TNF for signaling by the
55-kDa TNF receptor. J Biol Chem 1993, 268:18542–18548
Declercq W, Denecker G, Fiers W, Vandenabeele P: Cooperation of
both TNF receptors in inducing apoptosis: involvement of the TNF
receptor-associated factor binding domain of the TNF receptor 75.
J Immunol 1998, 161:390 –399
Pinckard JK, Sheehan KC, Schreiber RD: Ligand-induced formation
of p55 and p75 tumor necrosis factor receptor heterocomplexes on
intact cells. J Biol Chem 1997, 272:10784 –10789
Taylor RC, Cullen SP, Martin SJ: Apoptosis: controlled demolition at
the cellular level. Nat Rev Mol Cell Biol 2008, 9:231–241
Pietsch EC, Sykes SM, McMahon SB, Murphy ME: The p53 family and
programmed cell death. Oncogene 2008, 27:6507– 6521
Adams JM, Cory S: The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 2007, 26:1324 –1337
Soengas MS, Alarcon RM, Yoshida H, Giaccia AJ, Hakem R, Mak TW,
Lowe SW: Apaf-1 and caspase-9 in p53-dependent apoptosis and
tumor inhibition. Science 1999, 284:156 –159
Piguet PF, Vesin C, Guo J, Donati Y, Barazzone C: TNF-induced
enterocyte apoptosis in mice is mediated by the TNF receptor 1 and
does not require p53. Eur J Immunol 1998, 28:3499 –3505
Kennedy M, Wilson L, Szabo C, Salzman AL: 5-Aminosalicylic acid
inhibits iNOS transcription in human intestinal epithelial cells. Int J Mol
Med 1999, 4:437– 443
52. Chang WC, Coudry RA, Clapper ML, Zhang X, Williams KL, Spittle
CS, Li T, Cooper HS: Loss of p53 enhances the induction of colitisassociated neoplasia by dextran sulfate sodium. Carcinogenesis
2007, 28:2375–2381
53. Qiu W, Carson-Walter EB, Liu H, Epperly M, Greenberger JS, Zambetti GP, Zhang L, Yu J: PUMA regulates intestinal progenitor cell
radiosensitivity and gastrointestinal syndrome. Cell Stem Cell 2008,
2:576 –583
54. Berg DJ, Davidson N, Kuhn R, Muller W, Menon S, Holland G,
Thompson-Snipes L, Leach MW, Rennick D: Enterocolitis and colon
cancer in interleukin-10-deficient mice are associated with aberrant
cytokine production and CD4(⫹) TH1-like responses. J Clin Invest
1996, 98:1010 –1020
55. Okayasu I, Yamada M, Mikami T, Yoshida T, Kanno J, Ohkusa T:
Dysplasia and carcinoma development in a repeated dextran sulfate
sodium-induced colitis model. J Gastroenterol Hepatol 2002, 17:
1078 –1083
56. Yoshida T, Mikami T, Mitomi H, Okayasu I: Diverse p53 alterations in
ulcerative colitis-associated low-grade dysplasia: full-length gene sequencing in microdissected single crypts. J Pathol 2003, 199:166 –
175
57. Takaku H, Ajioka Y, Watanabe H, Hashidate H, Yamada S, Yokoyama
J, Kazama S, Suda T, Hatakeyama K: Mutations of p53 in morphologically non-neoplastic mucosa of long-standing ulcerative colitis.
Jpn J Cancer Res 2001, 92:119 –126
58. Fogt F, Vortmeyer AO, Goldman H, Giordano TJ, Merino MJ, Zhuang
Z: Comparison of genetic alterations in colonic adenoma and ulcerative colitis-associated dysplasia and carcinoma. Hum Pathol 1998,
29:131–136
59. Kern SE, Redston M, Seymour AB, Caldas C, Powell SM, Kornacki S,
Kinzler KW: Molecular genetic profiles of colitis-associated neoplasms. Gastroenterology 1994, 107:420 – 428
60. Chen R, Rabinovitch PS, Crispin DA, Emond MJ, Bronner MP, Brentnall TA: The initiation of colon cancer in a chronic inflammatory
setting. Carcinogenesis 2005, 26:1513–1519
61. Fujii S, Fujimori T, Kawamata H, Takeda J, Kitajima K, Omotehara F,
Kaihara T, Kusaka T, Ichikawa K, Ohkura Y, Ono Y, Imura J, Yamaoka
S, Sakamoto C, Ueda Y, Chiba T: Development of colonic neoplasia
in p53 deficient mice with experimental colitis induced by dextran
sulphate sodium. Gut 2004, 53:710 –716