Sie sind auf Seite 1von 17

J Gastroenterol 2008; 43:117

DOI 10.1007/s00535-007-2111-3

Review
Inflammatory bowel disease, past, present and future: lessons from
animal models
ATSUSHI MIZOGUCHI1,2 and EMIKO MIZOGUCHI2,3
1

Department of Pathology, Experimental Pathology, Simches 8234, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA,
02114 USA
2
Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
3
Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA

Accumulating data from animal models indicate that


inflammatory bowel disease (IBD) is mediated by a
much more complicated mechanism than previously
predicted. For example, the role of an individual molecule in the pathogenesis of IBD distinctly differs depending on several factors, including the fundamental
mechanism of induction of the disease, the target cell
type, the phase of disease, and the environment. Therefore, it has been difficult in the past to fully explain the
complicated mechanism. Novel concepts have recently
been proposed to further explain the complicated
mechanism of IBD. In this review, we introduce past,
current, and possible future concepts for IBD models
regarding T helper (Th) 1, Th2, and Th17, antigen sampling and presentation, regulatory cell networks, NOD2,
Toll-like receptors, bacteria/epithelia interaction, stem
cells, autophagy, microRNAs, and glycoimmunology,
and we also discuss the relevance of these new concepts,
developed at the bench (in animal models), to the
bedside.
Key words: antigen sampling, autophagy, IBD, Breg,
mucin, NOD2, Th17, TLRs

Introduction
Inflammatory bowel disease (IBD), comprising Crohns
disease (CD) and ulcerative colitis (UC), is a chronic
inflammatory disorder caused by multifactorial conditions in a genetically predisposed host.13 The complicated mechanism underlying IBD is clearly indicated by
the presence of many IBD susceptibility genes.4 Three

Received / Accepted: August 29, 2007


Reprint requests to: A. Mizoguchi

types of genetically engineered mice, T-cell receptor


(TCR-), interleukin (IL)-2-, and IL-10-knockout (KO)
mice were unexpectedly found in 1993, independently
by Mombaerts et al.,5 Sadlack et al.,6 and Kuhn et al.,7
to spontaneously develop chronic colitis. Spontaneous
development of colitis has since been identified in many
kinds of knockout and transgenic mice810 (Fig. 1). The
spontaneous development of colitis in so many different
kinds of genetically engineered mice further emphasizes
the presence of a more complicated mechanism than
previously predicted in the pathogenesis of IBD.
Although the animal models of IBD may not fully
reflect human IBD, these models have provided us great
opportunities to more closely examine the mechanism
of this disease. Indeed, animal models not only have
contributed significantly during the last decade to our
understanding of the complicated mechanism of IBD
but also have provided useful interventions for developing novel therapeutic strategies for this disorder.11 Currently, many animal models of IBD are available,810 and
each model has both merits and disadvantages for studying IBD. For example, acute intestinal injury induced in
mice by continuous administration of dextran sulfate
sodium (DSS) is a useful model for scientifically studying the epithelial barrier and its regeneration and innate
immune responses,12 but it may be an inappropriate
IBD model, for example, for evaluating the potency of
therapeutic agents in IBD patients.

Th17/IL-23
Accumulating data obtained from experimental IBD
models have suggested the involvement of both common
and distinct mechanisms in the pathogenesis of UC
versus CD. One of the major concepts previously used
to explain the different mechanisms is the T helper (Th)
1/Th2 paradigm.811,13 Indeed, Th2 cytokines such as IL-4
and IL-13 contribute significantly to chronic UC-like

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

T H1
Mdr1a

CBI-b
Smad3

PAST

TNF

(ARE)

Common

IL-10

Gi2

IL-2R

IL-2
IL-2R
CD3

TGF
IL-15

IBD

HLAB27

IL-7
gp39

CdCS

IL-4

NCAD

GFAP-HA

Tir8

Tn

A20

TH1

NFATc2 STAT3 GPX1/2


XBP1

LIGHT

Muc2

WASP
Keratin8

STAT4
Runx2

OT

R
E
H

TH2
Fig. 1. Deficiency (knockout mice, red colors) or overexpression (transgenic mice, blue colors) of many different molecules
in mice has been shown to induce the spontaneous development of colitis, which is mediated preferentially by an acquired
(ellipses) or innate (rectangles) immune pathway. The pathogenesis of colitis in some mouse models has significant contributions from the T helper (Th) 1 or Th2 pathway. IBD,
inflammatory bowel disease; IL, interleukin; TGF, transforming growth factor; A20, also known as tumor necrosis factor
(TNF)-induced protein 3; CBl-b, E3 ubiquitin ligase; Gi2, G
protein i2; GFAP-HA, transgenic mice in which enteric glia
are specifically disrupted; GPX, glutathione peroxidase;
HLAB27, HLAB27/human 2 microglobulin transgenic rats;
LIGHT, a TNF superfamily member; Mdr, multiple drug resistance; Muc2, mucin2; NCAD, transgenic mice that overexpress
dominant negative N-cadherin in the intestinal epithelial cells;
NEMO, NF-B essential modulator; NFAT, nuclear factor of
activated T cells; STAT, signal transducer and activator of
transcription; TCR, T-cell receptor; Tir8, also known as
SIGIRR (single Ig interleukin-1-related receptor); TNFARE,
mice lacking TNF AU-rich elements; WASP, Wiskott-Aldrich
syndrome protein; XBP1, X-box binding protein

disease.811 In contrast, the Th1 pathway has been implicated in the pathogenesis of CD.811,13 In addition to the
Th1/Th2 theory, accumulating recent studies have
unveiled the critical involvement of the IL-23/IL-17
pathway in the pathogenesis of CD-like experimental
diseases.1418 IL-12 (p40/p35) has generally been believed
to be a crucial factor involved in the development of
Th1-mediated colitis because of the beneficial effect of
anti-IL-12p40 treatment in CD patients as well as in
Th1-mediated experimental colitis.11 However, recent
studies have demonstrated that IL-23 (a heterodimer of
p40 and p19 subunits) rather than IL-12 (a heterodimer
of p40 and p35 subunits) contributes to the development of Th1-mediated chronic colitis observed in several
murine CD models, including IL-10 KO mice,14 the
CD45RB model,16,17 the cecal bacteria-specific T cellinduced colitis model,18 the Helicobacter hepaticus-

TH2
IL-4

Treg
RA

Tn TGF

IL-12/IFN

NEMO

TCR

TH2

PRESENT

IL-12/IFN

TH1

IL-6

TH17
IL-23

Fig. 2. In the past, the Th1/Th2 paradigm was a major concept


in the pathogenesis of acquired immune-mediated colitis. An
additional Th17/IL-23 pathway has recently been identified.
Interestingly, recent accumulating data have demonstrated the
reciprocal developmental pathways for generation of effector
Th17 and Foxp3+ regulatory T (Treg) cells. Th17 differentiation
is induced by TGF in the presence of proinflammatory cytokines such as IL-6. In contrast, Treg cell differentiation is
induced by TGF in the absence of IL-6 and/or presence of
retinoic acid (RA). IL-23 is required for the survival and maintenance of Th17. IFN, interferon

induced colitis model,16,17 and the CD40-dependent


innate immune-mediated colitis model.15 In addition, a
very recent study has shown that granuloma formation,
a characteristic feature of CD, is mediated by a unique
dendritic cell subset that coexpresses a macrophage
marker (F4/80) and produces a large amount of IL-23.19
Notably, the relevance of the data from these experimental colitis models to human CD is indicated by a
recent study that identifies a negative association
between IL-23 receptor polymorphisms and the development of IBD.20 Another very recent study also shows
that an IL-23R polymorphism (R381Q) is particularly
associated with CD in non-Jewish children.21 In contrast
to Th1-mediated chronic colitis, IL-12 rather than IL23 has been demonstrated to play a pathogenic role in
Th1-mediated acute colitis, induced by administration
of trinitrobenzesulfonic acid (TNBS).22
A current topic in the field of immunology is the discovery of a Th17 T-cell subset characterized by the production of IL-17 (IL-17A) and IL-17F but not interferon
(IFN)- or IL-4.2328 Differentiation of this Th17 subset
is induced through activation of orphan nuclear receptor RORt signaling, depending on the pleiotropic cytokine transforming growth factor (TGF) , which is also
linked to the development of Foxp3+ regulatory T (Treg)
cells.24,26,27 Reciprocal Th17 versus Treg cell differentiation is regulated by proinflammatory cytokines such as
IL-6 (for differentiation to Th17) and retinoic acid (for
differentiation to Treg cells).24,27,29 The expansion and
survival of Th17 T cells are subsequently mediated by
IL-2324,25 (Fig. 2). IL-17 expression is not detectable in
the normal colon of humans, but is readily detectable in

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

the colon of CD patients, regardless of disease severity.30,31 In contrast, IL-17 expression becomes detectable
only in the moderately to severely affected colon of UC
patients, and the expression levels of colonic IL-17 are
significantly lower in UC than in CD.30,31 The pathogenic
role of IL-17 has been demonstrated in some colitis
models. Neutralization of IL-17A activity results in the
suppression of Th1-mediated chronic colitis in IL-10 KO
mice,14 and the absence of IL-17 receptor in mice induces
resistance to the Th1-mediated acute colitis induced by
TNBS administration.32 In addition, activation of an
IL-17 receptor-mediated signaling pathway via Act1 in
epithelial cells contributes to the development of DSSinduced intestinal injury.33 In contrast, neutralization of
systemic IL-17A activity by administration of anti-IL17A monoclonal antibodies has been shown previously
to exacerbate DSS-induced intestinal injury.34 IL-17A
does not seem to be involved in Th2-mediated chronic
colitis, which develops spontaneously in TCR KO
mice.35 The regulatory role of IL-17 has been documented in a Th2-mediated asthma model.36 Therefore,
the role of IL-17 in the pathogenesis of colitis may differ
distinctly, depending upon, for example, whether the
phase of colitis is chronic or acute, whether Th1 or Th2
has contributed to the disease, and whether IL-17 activity is systemic or local. Interestingly, a recent study has
shown that Treg cells, which are well known to suppress
Th1-mediated colitis in the CD45RB model, are incapable of suppressing IL-17-mediated inflammation.37
Finally, it is important to mention a very recent observation that a T-cell subset producing both IL-17 and IFN-
is present in the inflamed intestine but not in the peripheral blood of CD patients.38
IL-22, which belongs to the IL-10 cytokine family,
specifically targets innate, but not acquired, immune
responses through the activation of STAT3.39 Interestingly, similar to the expression pattern of IL-17, colonic
IL-22 expression is specifically induced by inflammation
in both CD and UC, but the expression level of IL-22 is
significantly higher in CD than in UC.4042 Indeed, recent
studies have demonstrated that, in addition to IL-17,
IL-22 is also expressed preferentially by the Th17 cell
subset.43,44 Functionally, IL-22 has recently been shown
to mediate IL-23-induced inflammation in the skin.44 In
contrast, our recent study demonstrated that IL-22 contributes to the amelioration of ongoing Th2-mediated
chronic colitis, in which IL-23 is not upregulated (unpublished data). Since IL-22 possesses the ability to stimulate production of both regulatory (e.g., IL-10,
antibacterial peptide, and SOCS3) and deleterious (e.g.,
IL-8) molecules,3941,45 the reciprocal regulatory versus
deleterious functions of IL-22 may be determined by
the disease dependence on IL-23.

Antigen sampling and antigen-presenting cells


Presentation of antigens to T cells by professional
antigen-presenting cells (APCs) such as dendritic cells
(DCs) is a critical event for the activation of acquired
immune responses. A well-known lesson from animal
models of IBD is that enteric bacterial antigens are
required for the activation of pathogenic T cells involved
in the induction of colitis.9,11 I2 (a superantigen)46 and
flagellin47 have been identified as bacteria-derived
pathogenic antigens in the activation of deleterious
acquired immune responses. For antigen presentation,
luminal enteric bacterial antigens initially need to gain
access to APCs present in the lamina propria (LP).48 In
the past, the antigen-transport mechanism from the
lumen to the LP was explained only by a pathway mediated by M cells.49 A second pathway by which LP DCs
directly capture the luminal antigens by extending their
dendrites to the intestinal lumen across epithelial cells
was identified 6 years ago by Rescigno et al.50 and elegantly supported by an in vivo imaging system by Niess
and colleagues.51 Very recently, Yoshida et al.52,53 discovered an attractive third pathway by using a novel human/
mouse chimera system. Neonatal Fc receptor (FcRn)
serves as the vehicle that transports LP IgG across the
colonic epithelial layer into the lumen, where the IgG
can bind enteric bacterial antigens. The FcRn then recycles the antigen/IgG complex back across the colonic
epithelial cells into the LP for processing by DCs, which
are capable of presenting the antigens to T cells (Fig. 3).
Similarly, a CD23/IgE-dependent mechanism for transepithelial antigen sampling has recently been documented.54 Since the antigen-transport process represents
the initial event in the activation of pathogenic acquired
immune responses inducing colitis, one can predict that
inhibition of this process may contribute to the inhibition of IBD.
Significant advances in the field of DC biology have
been made during the last decade. The current knowledge regarding mucosal DCs is well summarized in
several excellent reviews.5557 Therefore, in this review,
we briefly describe recent findings regarding the functional role of DCs in colitis. DCs comprise phenotypically heterogeneous populations, and even in a given
subset possess considerable functional plasticity,
depending on their maturation stage and location and
environmental conditions.5558 Many phenotypic and
ex vivo functional studies of intestinal DCs associated
with experimental IBD have been performed, but only
a few studies have investigated whether DCs actually
induce or suppress the development of colitis in vivo.
One study has demonstrated that injection of in vitrogenerated DCs by stimulation with vasoactive intestinal
peptide contributes to the suppression of TNBS-induced
colitis by dampening the Th1 response and generating

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

Pathway 1
Bacteria

Pathway 3

Pathway 2
Ags
IC

Ags
M cells

FcRn
DCs
T cells

IgG

APC

Fig. 3. There are at least three pathways involved in the transportation of luminal antigens (Ags) to the intestinal lamina propria
(LP). M cells capture luminal Ags and then present them to T cells (Pathway 1). LP dendritic cells (DCs) directly capture the
luminal antigens by extending their dendrites to the intestinal lumen across epithelial cells (Pathway 2). Neonatal Fc receptor
(FcRn) serves as the vehicle that transports LP immunoglobulin G (IgG) across the colonic epithelial layer into the lumen where
the IgG can bind enteric bacterial antigens to form immune complexes (IC). The FcRn then recycles the antigens/IgG IC back
across the colonic epithelial cells into the LP for processing by antigen-presenting cells (APCs) such as DCs, which are capable
of presenting the antigens to T cells (Pathway 3)

IL-10-producing regulatory T cells.59 In addition,


cotransfer of in vitro-generated bone marrow (BM)derived DCs by stimulation with IL-10 may suppress the
colitis that is induced in recipient severe combined
immunodeficient (SCID) mice by transfer of CD4+CD25
T cells.60 In contrast, a unique CD11c+ DC-like subset,
which exhibits phenotypic and functional features of
immature myeloid DCs and is characterized by the
expression of a macrophage marker (F4/80), has recently
been shown to participate in the granuloma formation
associated with CD-like experimental colitis.19 Interestingly, a similar DC subset sharing features with macrophages has also been identified in the lung after infection
with Mycobacterium tuberculosis, which characteristically causes granulomatous inflammation.61 Future
studies employing a more sophisticated, genetic engineering approach capable of deleting specific DC subsets
in mice would allow us to further investigate the functional role of individual DC subsets in the pathogenesis
of IBD.

Regulatory cell network


In the past, the major concept explaining the mechanism by which deleterious acquired immune responses
in autoimmune diseases are generated was an expansion of specific pathogenic T-cell population(s). However, a novel concept was proposed two decades ago by
Sakaguchi et al.62 and Powrie and Mason.63 They proposed that dysregulated immune homeostasis caused by
the absence of a regulatory T-cell population contributes to the development of gastritis and IBD.62,63 This

novel concept has been widely supported during the last


decade by many studies, including a recent report that
CD4+CD25+ Treg cells are present not only in the spleen
but also in the intestinal LP.64 Many excellent reviews
discuss current advances made by studies of Treg
cells6568 and the intestinal immunoregulatory network.69
Therefore, in this review, we discuss other regulatory
cell populations that possess the potential ability to
inhibit the progression, but not the development, of
chronic colitis.
B cells are functionally characterized by their specific
feature of antibody production, but they were not typically considered in the past to be a major source of
cytokines. Harris and colleagues70 revised this concept
in 2000 by elegantly demonstrating that, like Th1/Th2 T
cells, B cells also possess the potential ability to produce
large amounts of several cytokines under certain inflammatory conditions. We also found that a regulatory cytokine, IL-10, is not generally produced by B cells in a
state of health, but a Th2-mediated intestinal inflammatory condition activates the B cells in the mesenteric
lymph nodes (MLNs) to produce IL-10.71 Of note, the
inducible IL-10-producing B cells subsequently contribute to the amelioration of the ongoing Th2-mediated
colitis but do not affect the development (initiation)
of this colitis. In humans and mice, IL-10-producing
regulatory B cells have since been identified in several
kinds of inflammations7282 but not in a state health or
in endotoxin shock induced by lipopolysaccharide
(LPS).83 IL-12p70 was initially identified nearly two
decades ago in an Epstein-Barr virus-transformed Bcell line.84 However, subsequent studies have failed to
detect the production of IL-12 by normal B cells from

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

PAST

PRESENT

B cells

B cells

2nd APCs

IL-12
Igs

Igs

IL-10

Fig. 4. In the past, immunoglobulin (Ig) production had generally been believed as the major function of B cells. However,
recent accumulating data have unveiled additional novel functions of B cells. B cells possess the abilities to produce several
cytokines such as IL-10 and IL-12 under inflammatory conditions and to serve as a secondary APCs to dampen T-cell
activation

healthy individuals or by LPS-stimulated B cells. Therefore, macrophages and DCs but not B cells have generally been believed to produce IL-12. However, this
concept may need to be revised in light of recent accumulating evidence that production of IL-12 by B cells
becomes easily detectable under certain inflammatory
conditions and when unmethylated CpG dinucleotides
(CpG), which became available for use only recently,
but not LPS, are used for the stimulation.77,85 Indeed,
we recently found that a Th2-mediated intestinal inflammatory condition as well as CpG stimulation induces
the differentiation of IL-12-producing B cells in the
presence of IL-10.35 Importantly, the inducible IL-12producing B cells contributed to the amelioration of
this Th2-mediated colitis. This finding may support
the earlier observations that Toll-like receptor (TLR)
9/CpG signaling contributes to the suppression of
colitis.8688 Interestingly, absence of IL-12-producing B
cells enhances the production of IL-17A by CD4+ T
cells.35 Indeed, IL-12 has previously been demonstrated
to inhibit IL-17 production.89 These data indicate that B
cells possess considerably more functional plasticity
than previously believed (Fig. 4).
The regulatory role of B cells in colitis and ileitis has
been demonstrated by several studies using different
IBD models, including TCR KO mice, Gi2 KO mice,
NFATc KO mice, and the CD45RB model.71,9096 Wei
and colleagues94 have identified a critical cellular
network for B cell-mediated regulation of colitis. MLN
B cells suppress the chronic colitis of Gi2 KO mice by
cooperating with regulatory CD8+ T cells and natural
killer T cells.94 In addition, recent studies in other autoimmune disease models have demonstrated that B cells
indirectly or directly enhance Treg cell function.9799 The
B cells that possess regulatory functions independent of
immunoglobulin production have been termed Breg
cells.95 It is important to mention that B cells, which

serve as a deleterious cell population, have also been


identified in some models of intestinal inflammation. B
cells have been reported to play a pathogenic role in the
ileitis seen in SAMP/YitFc mice,100 and to participate in
the induction of ileal villus atrophy.101 In addition,
genetically engineered ectopic expression of gp39 on B
cells causes the development of colitis and ileitis.102
These findings suggest the coexistence of pathogenic
and regulatory B cell populations in IBD patients.
Future studies are needed to determine whether B cells
can be targeted for the treatment of IBD patients.

NOD2 (CARD15)
One of the significant advances in IBD studies in the
last decade is the discovery of nucleotide-binding oligomerization domain 2 (NOD2)/caspase-recruitment
domain 15 (CARD15) as a CD susceptibility gene by
Hugot et al.103 and Ogura et al.104 Since recent advances
in IBD genetics and NOD2 biology have already been
clearly introduced in excellent reviews,1,3,4,103106 this
review focuses only on findings in murine models regarding the role of NOD2 in the pathogenesis of IBD.
NOD2 induces innate immune responses through
cytosolic recognition of bacterial components such as
muramyl dipeptide (MDP), which is a conserved structure in virtually all types of peptidoglycans (PGN).104 To
explore the role of NOD2 mutation in the pathogenesis
of CD, three independent research groups have generated NOD2 KO mice107,108 or NOD2939ic knockin (KI)
mice, which carry a NOD2 gene mutated at position
2939 (corresponding to 3020 in human NOD2).109 In the
NOD2 KO mice generated by Kobayashi et al.,108 protective innate immune responses mediated by NOD2
recognition of MDP are completely abolished. Although
the NOD2 KO mice do not spontaneously develop
colitis, they are more susceptible, with a significantly
reduced production of an antibacterial peptide (cryptidin) to Listeria monocytogenesis infection but not to
DSS-induced intestinal injury.108 Abnormally low antibacterial peptide production in the ileum (mainly by
Paneth cells) is likely a primary defect in CD.110 Watanabe and colleagues111,112 proposed a role of NOD2 in
the suppression of the TLR2-dependent innate immune
pathway involved in the activation of the Th1 response
in studies using the other NOD2 KO mice generated by
Pauleau and Murray.107 The NOD2 deficiency significantly enhances the production of IFN-, IL-18, and
IL-12p70 by the splenocytes in response to stimulation
with PGN111 and increases the susceptibility of mice to
TLR2-dependent colitis induced by ovalbumin (OVA)specific T cells in the presence of OVA-producing Escherichia coli (ECOVA).112 In contrast, alteration of such
TLR2-mediated immune responses is not observed in

the other NOD2 KO mice108 or in the NOD2939ic KI


mice.109 Therefore, any potential negative role of NOD2
in the TLR2 response may not be a universal phenomenon but may depend on the genetic background of the
mice used.108 The NOD2939ic KI mice also do not develop
colitis spontaneously, but are more susceptible to DSSinduced intestinal injury than wild-type (WT) mice.109
Interestingly, enhancement of NF-B activity and proinflammatory cytokine secretion by macrophages after
stimulation with MDP is observed in the NOD2939ic KI
mice.109 Many research groups have recently identified
and characterized the molecules capable of interacting
with NOD2, which include GRIM-19 (gene associated
with retinoid IFN-induced mortality),113 TAKI (TGFactivated kinase 1),114 Erbin,115 Centaurin 1,116 and
CARD9.117 Since these NOD2-interacting molecules
have been demonstrated to positively or negatively
regulate NOD2-mediated NF-B activation, future
studies using genetically engineered mice targeting
these molecules would further enhance our understanding of the biological function of NOD2 in the pathogenesis of CD.
Since deficiency or mutation of NOD2 in mice cannot
induce the spontaneous development of colitis,108,109
certain combinations of polymorphisms within multiple
IBD susceptible genes may be required to induce full
development of IBD. On the other hand, none of the
identified NOD2 variants are associated with CD in the
Japanese (non-Western) population.118,119 In addition,
the recently reported association of CD with variants in
other major IBD susceptibility genes, autophagy-related
16-like 1 (ATG16L1) and IL23R, is also not found in
the Japanese population.119 Therefore, in addition to
genetic susceptibility, other environmental and host
factors may be required to fully induce development of
IBD.

TLRs
The identification of mammalian TLRs as pattern recognition receptors for specific recognition of bacterial
products by Medzhitov et al.120 and Hoshino et al.121
led to major breakthroughs in many research fields,
including IBD studies. Recent advances in TLR biology
are well summarized in excellent reviews by the pioneers.122124 Therefore, here we focus only on the functional role of TLRs in the pathogenesis of colitis as
revealed by data accumulated during the last 5 years or
so.
TLR2 and TLR4
The role of TLR4 and TLR2 and their common adaptor
protein, myeloid differentiation factor 88 (MyD88), in

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

maintaining the epithelial barrier against DSS-induced


toxic damage has been well demonstrated by several
groups.125128 In DSS-induced acute intestinal damage,
the impaired epithelial barrier function in TLR2 KO,
TLR4 KO, and MyD88 KO mice leads to increases in
intestinal bleeding, the development of colonic erosion
and ulcers, the translocation of Gram-negative bacteria,
and the mortality rate.125128 Reduction of enteric bacteria flora by administration of a combination of antibiotics also significantly increases the susceptibility of mice
to DSS-induced lethal damage, which is presumably
caused by intestinal bleeding.125 This finding is consistent with the finding that administration of DSS induces
lethal damage, causing severe intestinal bleeding in
germ-free mice.129
As mentioned above, it is clear that the TLR2/TLR4/
MyD88 signaling pathway is required to preserve the
intestinal epithelial barrier against DSS-induced acute
stress.125128 In contrast, the severity of colitis induced by
a continuous DSS challenge varies considerably in
TLR2 KO, TLR4 KO, and MyD88 KO mice. For example,
compared with WT mice, the histological score (colitis
score) of DSS-induced acute intestinal damage in
MyD88 KO mice may be ameliorated,126 exacerbated,127
or unaffected.87,128 In addition, the histological score in
TLR4 KO mice may be ameliorated126,130,131 or exacerbated.87,127 One possible explanation is that different
environmental conditions in different mouse facilities
influence the mucosal immune responses after the disruption of the epithelial barrier. Alternatively, DSSinduced acute injury may not be an appropriate model
for studying colitis (IBD), although this model is very
useful for scientific studies of epithelial homeostasis and
barrier function. Recently, many groups have examined
the recovery phase after a single administration of
DSS.93,132136 Both acquired and innate immune responses
have been implicated in the recovery phase,93,132 and the
epithelial cell regeneration observed during the recovery phase resembles that observed in chronic colitis.135
Therefore, this recovery model may be useful for studying the wound healing process (epithelial regeneration)
and the linkage between innate and acquired immune
responses. Since the balance between damage and
healing is a critical determinant of the progression (after
onset) of IBD, the recovery model may more closely
reflect the mechanism of IBD progression or perpetuation. Utilizing the recovery model, Cario et al.128 recently
demonstrated that TLR2 significantly enhances the
healing process of colitis by regulating ZO-1-associated
epithelial barrier integrity.
The role of TLR2/TLR4/MyD88 in acquired immune
responses involved in the pathogenesis of chronic colitis
has also been investigated recently. The bacterial lipoprotein Pam3Cys-SK4 (TLR1/TLR2 agonist) is able to
transiently suppress the function and Foxp3 expression

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

of Treg cells. Suppression of CD45RBhigh T cell-induced


colitis (CD45RB model) by Treg cells is significantly
delayed when the cells are pretreated with Pam3CysSK4.137 Administration of the synthetic TLR4 antagonist CRX-526 attenuates the chronic colitis that
spontaneously develops in multidrug resistant gene 1
(Mdr1) KO mice.130 Spontaneous development of colitis
in IL-10 KO mice may be abolished by the absence of
MyD88.138 These data suggest the pathogenic role of
TLR2/TLR4/MyD88 signaling in some types of acquired
immune-mediated chronic colitis. In contrast, development of colitis in IL-2 KO mice is independent of
TLR signaling, as indicated by the development of
colitis in MyD88- and Toll-interleukin 1 receptor
domain-containing adaptor inducing interferon
(TRIF) 1-deficient IL-2 triple KO mice.138 These data,
together with those from the DSS model, indicate that
the contribution of TLR2/TLR4/MyD88 signaling to
the pathogenesis of IBD is determined by many factors,
including the fundamental mechanism of the colitis
(e.g., IL-10 KO versus IL-2 KO mice), the cell type (e.
g., epithelial versus Treg cells), and the phase of colitis
(e.g., acute versus recovery versus chronic). Future
studies are needed to clarify the complicated role of
TLR2/TLR4/MyD88 signaling in the pathogenesis of
IBD.
TLR5
By utilizing a serological expression cloning technique,
Lodes et al.47 identified flagellin (TLR5 ligand), a
common bacterial antigen present on most motile bacteria in the intestine, as the dominant antigen capable
of stimulating Th1 responses in CD patients. In an
experimental mouse system, they confirmed that flagellin-specific CD4+ T cells induced colitis in the recipient
mice.47 The involvement of flagellin in acquired immune
responses is further supported by data showing that
flagellin-specific immunoglobulin production is T celldependent.139 The relevance of flagellin to the pathogenesis of human CD is strongly emphasized by the
following observations. A dominant-negative TLR5
polymorphism (called TLR-stop) is negatively associated with CD in Jewish, but not in non-Jewish, patients.140
Anti-flagellin antibodies are frequently detected in CD
but not in UC patients.141 Interestingly, CD patients carrying at least 1 NOD2 variant exhibit enhanced antiCBir1 flagellin reactivity.142
In addition to the role of flagellin in IBD-associated
acquired immune responses, flagellin can also activate
innate immune responses, as indicated by its ability to
stimulate colonic epithelial cells (CECs) to produce
proinflammatory cytokines.143,144 Interestingly, the flagellin/TLR5 response in CECs is basolaterally polarized in
healthy individuals, and CECs in intact colon therefore

do not respond to flagellin, suggesting that the response


of CECs to flagellin is specifically elicited under inflammatory conditions.144 Consistent with this, rectal administration of flagellin does not induce host responses in
normal mice but aggravates DSS-induced acute colonic
injury.144
TLR9
Unmethylated CpG dinucleotides (CpG), which are
frequently recognized in the genomes of bacteria and
viruses, were initially found in 1995 by Krieg et al.145
to stimulate B cells. A subsequent study by Hemmi and
colleagues146 identified TLR9 as the receptor for CpG.
The role of CpG/TLR9 in the pathogenesis of IBD has
been vigorously investigated by the research groups of
Raz and Falk. Rachmilewiz et al.86,87 initially showed
that intragastric and subcutaneous, but not intrarectal,
administration of CpG oligodeoxynucleotides (ODNs)
suppresses DSS-induced acute intestinal injury, TNBSinduced acute colitis, and chronic colitis in IL-10 KO
mice through downregulation of proinflammatory
cytokine and matrix metalloproteinase expression. The
dependence of this therapeutic effect on TLR9 was
further confirmed in TLR9 KO mice.87 Mechanistically,
innate immune pathway-mediated production of type
I IFN is required for the CpG-mediated suppression
of DSS-induced acute injury.88 Interestingly, a unique
pathway for activation of CECs by CpG was recently
discovered.147 Basolateral TLR9 signals activation of
the NF-B pathway in CECs, whereas apical TLR9
prevents activation of the NF-B pathway by inducing
accumulation of ubiquitinated IB in the cytoplasm.147
There are three types of synthetic CpG ODNs having
distinct structures and immunostimulatory activities.148
The effect of CpG ODNs on proinflammatory cytokine
production in UC patients has been shown to differ
depending on the type of CpG ODNs.149 Class B CpG
ODNs are able to inhibit colonic tumor necrosis factor
(TNF)- and IL-1 production in UC patients. In contrast, class A or C CpG ODNs do not have inhibitory
activity. A possible role of CpG ODNs in the induction
of regulatory properties in nave T cells has also been
proposed. CD4+CD62L+ T cells from CpG ODNpretreated donor mice are unable to induce colitis in
recipient SCID mice and can suppress CD4+ T cellmediated colitis.150 Our recent study showed that CpG
activates a regulatory B-cell population (Breg) in Th2mediated chronic colitis in TCR KO mice.35 These
findings suggest the regulatory role of CpG/TLR9 in
the pathogenesis of colitis.
In contrast, a potential capacity of CpG/TLR9 to
exacerbate colitis has also been demonstrated. Administration of CpG exacerbates DSS-induced acute
injury.151 DSS-induced chronic colitis is attenuated in

TLR9 KO mice.152 In addition, administration of adenoviral CpG oligodeoxynucleotides (AV-ODNs), which


are capable of blocking the CpG effect, contributes to
the amelioration of several types of experimental colitis,
including DSS-induced chronic colitis, the CD45RB
model, and IL-10 KO mice.152 This suppression of colitis
is associated with a decrease in IL-6, IFN-, and T-bet
expression. In addition, our recent study showed that
CpG stimulates a unique DC subset to produce IL-23
and cause granuloma formation.19 Taken together, these
findings suggest that the functions of CpG/TLR9 signaling in the pathogenesis of IBD are extremely complicated. The role of CpG/TLR9 signaling in the
pathogenesis of IBD may be determined by many
factors, such as the type of CpG (class B versus class A
or C), the target cell type (CEC versus unique DCs),
and the route of CpG exposure (systemic and oral
versus rectal exposure or apical versus basolateral exposure in CECs). Future studies are needed to further
dissect the complicated mechanism of CpG/TLR9 signaling in the pathogenesis of IBD.
Other TLRs
Kuwata and colleagues153 recently identified TLRinducible nuclear IB (IBNS) to be a selective inhibitor of TLR-dependent gene expression. Absence of
IBNS enhances the production of IL-6, IL-12p40, and
IL-12p70 but not TNF- by DCs and macrophages in
response to stimulation via TLRs, including TLRs 1, 2,
4, 6, and 7. IBNS KO mice exhibit higher susceptibility
to LPS-induced endotoxin shock and DSS-induced
acute intestinal damage. Therefore, IBNS appears to
be critical for controlling inflammatory responses by
negatively regulating the induction of TLR-dependent
gene expression through modulation of NF-B activity.153 The role of TLR3 in DSS-induced intestinal injury
has recently been investigated in the DSS model.154 Subcutaneous, but not oral, administration of synthetic viral
RNA poly(I:C) protects against DSS-induced acute
intestinal damage in the presence or absence of IL-10.
This protective action of poly(I:C) is abolished in the
absence of TLR3.

Bacteria/epithelial cell interaction and


epithelial function
Humans and other mammals are host to 1012 viable
bacteria per gram of colonic content, comprising 500
1000 microbial species.155 During the last decade, much
accumulating data have clearly shown that commensal
enteric bacteria are critical in determining the development and course of IBD.3,11 In addition to the potentially pathogenic effect of enteric bacteria, the presence

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

of probiotic bacteria contributing to the suppression of


colitis has also been well documented during the last
decade.156 Most recently, unexpected findings regarding
the function of commensal bacteria have been made.
Bacteroides thetaiotaomicron is able to change its surface
carbohydrates in response to the nutrient glycan environment that it is accessing and also to evade a host
immune response.157 Indeed, a bacterial polysaccharide
derived from Bacteroides fragilis directs the cellular and
physiological maturation of the developing host immune
system.158 Adherent-invasive E. coli (AIEC), which has
been proposed as a potential pathogenic bacteria in CD,
has recently been shown to adhere to CECs through the
interaction of type 1 pili of AIEC and carcinoembryonic
antigen-related cell adhesion molecule 6 (CECAM6) on
the CECs.159 Chitinase 3-like 1, which is a member of
the chi-lectin family capable of binding to chitin, has
also been shown to enhance the binding of AIEC to
CEC.136 In addition to AIEC, Enterococcus faecalis is
capable of inducing the development of colitis in gnotobiotic IL-10 KO mice through activation of MyD88dependent NF-B activation of mucosal immune
cells.160
As the first line of defense against enteric antigens as
well as invasive or noninvasive enteric bacteria, CECs
play a critical role.161 Goblet cell depletion and thin
mucus layers are characteristic features of IBD, particularly UC.162 Because the layer of mucus lining the intestinal tract serves as a lubricant and a physiological
barrier between the luminal contents and mucosal
surface,162,163 it has generally been accepted conceptually, but without direct evidence, that the mucus layer
plays a regulatory role in colitis. Direct evidence
supporting this concept was provided most recently by
An et al.164 The intestinal mucus layer is a dense,
carbohydrate-rich matrix that consists primarily of
O-glycosylated mucins.162,163 Deficiency of 1,3-Nacetylglucosaminyltransferase (C3GnT) in mice impairs
colonic mucus layer formation by eliminating core 3derived O-glycans and significantly increases their susceptibility to DSS-induced acute intestinal injury.164 In
addition, a critical contribution of Muc2 (a secretary
mucin) in the suppression of colitis has been confirmed
by the data that Muc2 KO mice spontaneously develop
colitis.165 A regulatory role of Muc3 (a membrane-bound
mucin) in DSS- and acetic acid-induced acute intestinal
injury has also been demonstrated.166 Most recently, a
study using Muc1 KO mice demonstrated that surface
Muc1 protects epithelial cells from xenobiotic toxins;
increased apoptosis of intestinal epithelial cells was
observed in Muc1 KO mice challenged with Campylobacter jejuni.167 In contrast, genetically engineered overexpression of human Muc1 leads to the exacerbation of
chronic colitis in IL-10 KO mice.168 Therefore, the role
of Muc1 in intestinal inflammation may differ depend-

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

ing on the type of colitis (e.g., infectious versus chronic


colitis). Alternatively, as appropriately glycosylated
mucins are required to form a tight mucus layer assembly,162,163 it is also possible that overexpressed Muc1,
which may be inappropriately glycosylated, causes
mucus layer formation to be insufficient, thus exacerbating colitis.
During the last decade, accumulating studies have
revealed the role of goblet cells in the pathogenesis of
colitis. Goblet cell-derived intestinal trefoil factor (ITF)
contributes to the suppression of colitis by enhancing
CEC regeneration.169 Interestingly, partial depletion of
goblet cells by genetically engineered expression of the
diphtheria toxin gene under control of the ITF promoter enhances the production of ITF and significantly
increases the resistance of mice to DSS-induced acute
intestinal injury.170 As mentioned above, some Muc
proteins, which are produced preferentially by goblet
cells, participate in the suppression of colitis.165,166 In
contrast, resistin-like molecule (RELM), which is
specifically expressed by intestinal goblet cells, is a
pathogenic factor in DSS-induced acute intestinal injury
through the activation of TNF- production by macrophages171 or through activation of IL-13 production.172
Interestingly, IL-13 KO mice are protected against DSSinduced acute intestinal injury.172 In contrast to DSS
injury, RELM plays a regulatory role in Th1-mediated
acute colitis induced by TNBS.172 These data suggest
that the pathophysiological role of goblet cells in intestinal inflammation is more complicated than previously
predicted.
In addition to the barrier function of CECs, novel
biological functions of CECs have been identified during
the last decade. The biological functions of CECs under
intestinal inflammatory conditions vary significantly
depending upon several factors, such as the phase of
colitis (acute versus recovery versus chronic) and the
fundamental mechanism of colitis development (Th1
versus Th2 contributions).135,173 Detoxification pathways
in CECs are significantly downregulated in chronic but
not in acute colitis. Anti-inflammatory molecules such
as antibacterial peptides are commonly upregulated in
CECs in acute and chronic colitis. Production of a Ctype lectin, RegIII, by CECs is markedly increased
under chronic and acute intestinal inflammatory conditions but not in the recovery phase of colitis.135 Interestingly, RegIII has recently been shown to serve as an
antimicrobial protein and to be induced by microbial
epithelial contact through MyD88-mediated signaling.110,174
Moreover,
enhanced
expression
of
antigen-presenting molecules on CECs is specifically
observed in chronic, but not in acute, colitis.135 Indeed,
the ability of CECs to serve as APCs has been well
documented in IBD patients.161 CECs can induce unique
CD8+ regulatory T cells through interaction with CEC-

derived carcinoembryonic antigen 5.161 Interestingly,


CECs also activate a CD-associated pathogenic
CD4+ T-cell population through the interaction of MICA
on CECs and NKG2D on the CD4+ T cells.175 In addition, CECs have recently been demonstrated to be
capable of controlling the T cell-independent immunoglobulin class switching of intestinal B cells to IgA, positively by producing APRIL176, or negatively by producing
secretory leukocyte protease inhibitor.177 Furthermore,
CECs have been shown to condition intestinal DCs
through the constitutive release of thymic stromal
lymphopoietin.178
The activation of CECs is controlled by several signaling molecules. STAT3 activation in acquired immune
cells has been well documented to contribute to the
development or exacerbation of colitis.179 In contrast,
STAT3 activation of CECs contributes to the suppression of colitis, as indicated by the spontaneous development of colitis in conditional KO mice, in which STAT3
is specifically deleted in the CECs.180 Similar observations have been made regarding NF-B signaling. NFB signaling has been well implicated in the pathogenesis
of acquired immune-mediated colitis181 and DSSinduced intestinal injury.182 In contrast, a very recent
study has elegantly demonstrated that specific deletion
of NEMO (NF-B essential modulator) in the CECs
induces the spontaneous development of colitis.183 These
data clearly indicate that activation of the major signaling pathways in acquired versus innate (CEC) immune
responses results in distinctly different outcomes in the
pathogenesis of colitis. Therefore, as suggested by
Plevy,184 given the double-edged functioning of such
major signaling cascades in colitis, careful attention
should be directed toward the cell population that is
being targeted when contemplating the inhibition or
activation of these signaling cascades in human IBD.

Future topics
Basic science has been continuously progressive both
technically and conceptually. In the following section,
we introduce attractive novel avenues of investigation
in IBD research and discuss classical, but clinically relevant, subjects that might contribute to the immediate
translation from bench to bedside of concepts for
improving the lives of patients with IBD.
Stem cells
A major current topic in science and medicine is stem
cell biology. Further advances in this field are guaranteed to contribute significantly to breakthroughs in the
development of novel therapeutic strategies for several
diseases. Importantly, the relevance of stem cell biology

10

to the treatment of IBD was initially introduced in 2002


by Okamoto and colleagues,185 who showed that bone
marrow-derived stem cells possess the potential ability
to differentiate into CECs in humans. In addition, the
intestine has been identified as a location of the differentiation of lin c-kit immune precursors to T cells (in
mice) and natural killer cells (in humans).186,187 Of note,
recent studies have emphasized the therapeutic potency
of stem cell-based treatment of IBD. Administration of
CD34 stem cells, which are capable of differentiating
into endothelial cells, contributes to the suppression of
DSS-induced intestinal injury.188 BM cells that are
capable of differentiating into myofibroblasts, endothelial cells, pericytes, and vascular smooth muscle cells,
particularly under intestinal inflammatory conditions,
can contribute to the amelioration of TNBS-induced
colitis.189
Autophagy
Autophagy is a unique process of membrane trafficking in which the membrane compartment (autophagosomes) engulfs both organelles and cytosolic
macromolecules and delivers them to the lysosome for
degradation.190 Notably, ATG16L1 was identified most
recently as a new susceptibility gene in CD.191,192 Since
the appearance of bacteria in the cytoplasm in APCs
triggers specialized autophagy (xenophagy),190 this
cytoplasmic process fits well with the cytoplasmic recognition of bacterial antigens through NOD2. In addition, the TRIF-dependent, MyD88-independent TLR4
signaling pathway has recently been identified to regulate autophagy (xenophagy).193 This finding strongly
indicates the significant relevance of this field to IBD.
Alternatively, the major function of autophagy is the
regulation of self-responses.190 Autophagy contributes
to apoptotic cell clearance by a mechanism that involves
the generation of energy-dependent engulfment
signals.194 Importantly, macroautophagy constitutively
and efficiently delivers cytosolic protein (e.g., selfantigens and pathogens in host cells) for MHC class II
presentation to activate the acquired immune system.195,196
The role of autophagy in self-responses revives past
findings in IBD models. An involvement of impaired
clearance of apoptotic cells in the exacerbation of
chronic colitis was proposed in 1997.90 A study conducted in 1999 demonstrated the ability of self-antigen
(hsp60)-specific CD8+ T cells to induce ileitis.197 In addition, self-antigens have previously been shown to contribute to the conditioning of the clonality of the
pathogenic CD4+ T-cell population capable of inducing
colitis.198 Interestingly, the self-reactive CD4+ T-cell population induces the mild, but not the severe, form of
colitis. This previous observation may be consistent
with recent data that commensal bacteria are required

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

for exacerbation but not development of ileitis in


SAMP1/YitFc mice.199 Therefore, autophagy may be
an important process in IBD pathogenesis, inducing
not only innate immune responses against exogenous
antigens but also acquired immune responses against
self-antigens.
MicroRNA
MicroRNAs (miRNAs) have recently been implicated
in an increasingly wide variety of biological processes,
including innate and acquired immune responses.200,201
Currently, 474 human miRNA sequences have been
identified, but the total number of miRNA genes according to computer-assisted estimates might be closer to a
thousand.200 In bovines, expression of some miRNAs
(e.g., bya-miR-145) is restricted to the small intestine,202
suggesting a functional role of miRNA in intestinal
homeostasis or intestinal immune responses. Therefore,
miRNA research may represent a new avenue of exploration to further enhance our understanding of the
pathogenesis of IBD. However, no study on miRNAs in
IBD has yet been carried out.
Glycoimmunology
The majority of immune responses are mediated by
protein/protein interactions such as cytokine/receptor
interactions. However, in some cases, there are unusual
results that cannot easily be explained by protein/protein
interactions. Such results have frequently been described
by the terminologically useful phrase conformational
changes of receptors. Importantly, the majority of
immune receptors are N- or O-glycosylated, and the
assembly of receptors is significantly affected by the
glycosylation state, which is controlled by many kinds
of glycosylation-associated enzymes.203,204 The expression pattern of these glycosylation-associated enzymes
is altered by several factors, such as inflammation, suggesting the modification of carbohydrate (sugar) assembly in the cells under inflammatory conditions. Therefore,
carbohydrate assembly actively participates in the regulation of the acquired and innate immune responses that
control autoimmune diseases.205207 The importance of
protein/carbohydrate interactions has been highlighted
by studies on cell homing through selectin pathways.208
In addition, the involvement of protein/carbohydrate
interactions in pathogen recognition through C-type
lectins such as DC-SIGN has recently been well documented.209,210 Protein/carbohydrate interactions also
contribute to activation or dampening of T-cell responses.
A protein (a lectin called galectin-1) induces the apoptosis of T cells211 and also dampens Th1 differentiation.212
Indeed, a regulatory role of galectin-1 in TNBS colitis
has been demonstrated.213 In contrast to galectin-1,

A. Mizoguchi and E. Mizoguchi: IBD past, present and future

11

which preferentially binds to a lactosamine unit of


mature core 2-branched O-glycan,203,204 galectin-4,
which is capable of interacting with immature core 1
O-glycan,214 has been demonstrated to contribute to the
exacerbation of Th2-mediated chronic colitis (TCR
KO mice) by stimulating intestinal, but not systemic,
CD4+ T cells to produce IL-6 only under inflammatory
conditions.93 These findings emphasize the significant
involvement of protein/carbohydrate interactions in the
pathogenesis of IBD.215 However, the mechanism underlying this phenomenon has not been vigorously investigated in IBD.

are capable of specifically targeting more restricted cell


population(s) may be more appropriate for reducing
the risk of development of unwanted adverse effects.

Other topics

References

A large body of epidemiological studies has clearly


demonstrated that prior appendectomy and cigarette
smoking are protective against the development of UC.2
The suppressive role of appendectomy (resection of the
cecal patch in mice) in the development of experimental
chronic colitis sharing several features with UC has previously been demonstrated.198,216 In addition, the therapeutic effect of carbon monoxide (CO) exposure at low
concentration on established chronic colitis in IL-10 KO
mice has been recently demonstrated.217 Since such
studies (appendectomy and cigarette smoking) have significant clinical relevance, a full understanding of the
suppressive mechanism is needed to provide a basis for
developing novel therapeutic strategies for treating
IBD patients. Another attractive field that might be
immediately applicable for treating IBD patients is
natural food products. For example, retinoic acid has
recently been identified to have several regulatory
immune functions, including activation of mucosal IgA
responses218 and induction of Foxp3+ Treg cell differentiation.29,219221 In addition, curcumin, a major component of turmeric, has been demonstrated to exhibit a
beneficial effect on the suppression of TNBS-induced
colitis.222 Beneficial therapeutic effects of omega-3 fatty
acids on IBD have also been supported by recent studies
using genetically engineered mice.223,224

Conclusion
Finding accumulated in the past and more recently
clearly show that IBD is mediated by a much more
complicated mechanism than previously or even currently predicted. Future studies in animal models of
IBD will contribute to the full dissection of the complicated mechanism of IBD and provide important bases
for developing novel therapeutic strategies to save the
lives of patients with IBD. Since the roles of several
molecules in the pathogenesis of IBD distinctly differ
depending on the type of cell, therapeutic strategies that

Acknowledgments. This study was supported by NIH grants


DK64351 (AM) and DK64289, DK74454, and DK43351 (EM).
We also thank the Eli and Edythe L. Broad Medical Foundation for support.
We thank Ms. Sarah Murphy for her secretarial
assistance in preparing this manuscript. We also thank Drs.
Kiyotaka Nagahama, Mayumi Kawada, Yasuyo Shimomura,
Ken Sugimoto, Atsuko Arihiro, Katsunori Shirane, and
Atsuhiro Ogawa for their excellent contributions.

1. Sands BE. Inflammatory bowel disease: past, present and future.


J Gastroenterol 2007;42:1625.
2. Kucharzik T, Maaser C, Lugering A, Kagnoff M, Mayer L,
Targan S, et al. Recent understanding of IBD pathogenesis:
implications for future therapies. Inflamm Bowel Dis 2006;12:
106883.
3. Xavier RJ, Podilsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007;448:42734.
4. Cho JH, Abraham C. Inflammatory bowel disease genetics:
Nod2. Annu Rev Med 2007;58:40116.
5. Mombaerts P, Mizoguchi E, Grusby MJ, Glimcher LH, Bhan AK,
Tonegawa S. Spontaneous development of inflammatory
bowel disease in T cell receptor mutant mice. Cell 1993;75:
27482.
6. Sadlack B, Merz H, Schorle H, Schimpl A, Feller AC, Horak I.
Ulcerative colitis-like disease in mice with a disrupted
interleukin-2 gene. Cell 1993;75:25361.
7. Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W.
Interleukin-10-deficient mice develop chronic enterocolitis. Cell
1993;75:26374.
8. Strober W, Fuss IJ, Blumberg RS. The immunology of mucosal
models of inflammation. Annu Rev Immunol 2002;20:495
549.
9. Mizoguchi A, Mizoguchi E, Bhan AK. Immune networks in
animal models of inflammatory bowel disease. Inflamm Bowel
Dis 2003;9:24659.
10. Elson CO, Cong Y, McCracken VJ, Dimmitt RA, Lorenz RG,
Weaver CT. Experimental models of inflammatory bowel disease
reveal innate, adaptive, and regulatory mechanisms of host dialogue with the microbiota. Immunol Rev 2005;206:26076.
11. Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. J Clin Invest 2007;117:51421.
12. Dieleman LA, Ridwan BU, Tennyson GS, Beagley KW, Bucy RP,
Elson CO. Dextran sulfate sodium-induced colitis occurs in
severe combined immunodeficient mice. Gastroenterology 1994;
107:164352.
13. Neurath MF, Finotto S, Glimcher LH. The role of Th1/Th2 polarization in mucosal immunity. Nat Med 2002;8:56773.
14. Yen D, Cheun J, Scheerens H, Poulet F, McClanahan T, Mckenzie
B, et al. IL-23 is essential for T cell-mediated colitis and promotes
inflammation via IL-17 and IL-6. J Clin Invest 2006;116:1310
6.
15. Uhlig HH, McKenzie BS, Hue S, Thompson C, Joyce-Shaikh B,
Stepankova R, et al. Differential activity of IL-12 and IL-23 in
mucosal and systemic innate immune pathology. Immunity
2006;25:30918.
16. Kullberg MC, Jankovic D, Feng CG, Hue S, Gorelick PL,
McKenzie BS, et al. IL-23 plays a key role in Helicobacter
hepaticus-induced T cell-dependent colitis. J Exp Med 2006;203:
248594.

12
17. Hue S, Ahern P, Buonocore S, Kullberg MC, Cua DJ, McKenzie
BS, et al. Interleukin-23 drives innate and T cell-mediated intestinal inflammation. J Exp Med 2006;203:247383.
18. Elson CO, Cong Y, Weaver CT, Schoeb TR, McClanahan TK,
Fick RB, et al. Monoclonal anti-interleukin 23 reverses active
colitis in a T cell-mediated model in mice. Gastroenterology
2007;132:235970.
19. Mizoguchi A, Ogawa A, Takedatsu H, Sugimoto K, Shimomura
Y, Shirane K, et al. Dependence of intestinal granuloma formation on unique myeloid DC-like cells. J Clin Invest 2007;
117:60515.
20. Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly
MJ, et al. A genome-wide association identifies IL23R as an
inflammatory bowel disease gene. Science 2006;314:14613.
21. Dubinsky MC, Wang D, Picornell Y, Wrobel I, Katzir L, Quiros
A, et al. IL-23 receptor (IL-23R) gene protects against pediatric
Crohns disease. Inflamm Bowel Dis 2007;13:5115.
22. Becker C, Dornhoff H, Neufert C, Fantini MC, Wirtz S,
Huebner S, et al. IL-23 cross regulates IL-12 production in T
cell-dependent experimental colitis. J Immunol 2006;177:2760
4.
23. Mangan PR, Harrington LE, OQuinn DB, Helms WS, Bullard
DC, Elson CO, et al. Transforming growth factor induces development of the Th17 lineage. Nature 2006;441:2314.
24. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al.
Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006;441:
2358.
25. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger
B. TGF in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 2006;24:17989.
26. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A,
Lafaille JJ, et al. The orphan nuclear receptor RORt directs the
differentiation program of proinflammatory IL-17+ T helper
cells. Cell 2006;126:112133.
27. Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy
KM. Th17: an effector CD4 T cell lineage with regulatory T cell
ties. Immunity 2006;24:67788.
28. Steinman L. A brief history of Th17, the first major revision in
the Th1/Th2 hypothesis of T cell-mediated tissue damage. Nat
Med 2007;13:13945.
29. Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M,
et al. Reciprocal Th17 and regulatory T cell differentiation mediated by retinoic acid. Science 2007;317:25660.
30. Fujino S, Andoh A, Bamba S, Ogawa A, Hata K, Araki Y, et al.
Increased expression of interleukin 17 in inflammatory bowel
disease. Gut 2003;52:6570.
31. Nielsen OH, Kirman I, Rudiger N, Hendel J, Vainer B. Upregulation of interleukin-12 and -17 in active inflammatory bowel
disease. Scan J Gastroenterol 2003;38:1805.
32. Zhang Z, Zheng M, Bindas J, Schwarzenberger P, Kolls JK. Critical role of IL-17 receptor signaling in acute TNBS-induced
colitis. Inflamm Bowel Dis 2006;12:3828.
33. Qian Y, Liu C, Hartupee J, Altuntas CZ, Gulen MF, Jane-wit D,
et al. The adaptor Act1 is required for interleukin 17-dependent
signaling associated with autoimmune and inflammatory disease.
Nat Immunol 2007;8:24756.
34. Ogawa A, Andoh A, Araki Y, Bamba T, Fujiyama Y. Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced
colitis in mice. Clin Immunol 2004;110:5562.
35. Sugimoto K, Ogawa A, Shimomura Y, Nagahama K, Mizoguchi
A, Bhan AK. Inducible IL-12-producing B cells regulate Th2mediated intestinal inflammation. Gastroenterology 2007;133:
12436.
36. Schnyder-Candrian S, Togbe D, Couillin I, Mercier I,
Brombacher F, Quesniaux V, et al. Interleukin-17 is a negative
regulator of established allergic asthma. J Exp Med 2006;203:
271525.

A. Mizoguchi and E. Mizoguchi: IBD past, present and future


37. Lohr J, Knoechel B, Wang JJ, Villarino AV, Abbas AK. Role of
IL-17 and regulatory T lymphocytes in a systemic autoimmune
disease. J Exp Med 2006;203:278591.
38. Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F,
Mazzinghi B, et al. Phenotypic and functional features of human
Th17 cells. J Exp Med 2007;204:184961.
39. Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R.
IL-22 increases the innate immunity of tissues. Immunity 2004;
21:24154.
40. Andoh A, Zhang Z, Inatomi O, Fujino S, Deguchi Y, Araki Y,
et al. Interleukin-22, a member of the IL-10 subfamily, induces
inflammatory responses in colonic subepithelial myofibroblasts.
Gastroenterology 2005;129:96984.
41. Brand S, Beigel F, Olszak T, Zitzmann K, Eichhorst ST, Otte JM,
et al. IL-22 is increased in active Crohns disease and promotes
proinflammatory gene expression and intestinal epithelial cell
migration. Am J Physiol 2006;290:G82738.
42. Wolk K, Witte E, Hoffmann U, Doecke W-D, Endesfelder S,
Asadullah K, et al. IL-22 induces lipopolysaccharide-binding
protein in hepatocytes: a potential systemic role of IL-22 in
Crohns disease. J Immunol 2007;178:597381.
43. Liang SC, Tan XY, Luxenberg DP, Karim R, DunussiJoannopoulos K, Collins M, et al. 2006. Interleukin (IL)-22 and
IL-17 are coexpressed by Th17 cells and cooperatively enhance
expression of antimicrobial peptides. J Exp Med 2006;203:
22719.
44. Zheng Y, Danilenko DM, Valdez P, Kasman I, EasthamAnderson J, Wu J, et al. Interleukin-22, a Th17 cytokine, mediates
IL-23-induced dermal inflammation and acanthosis. Nature
2007;445:64851.
45. Nagalakshmi ML, Rascle A, Zurawski S, Menon S, de Waal
Malefyt R. Interleukin-22 activates STAT3 and induces IL-10
by colon epithelial cells. Int Immunopharmacol 2004;4:679
91.
46. Dalwadi H, Wie B, Kronenberg M, Sutton CL, Braun J. The
Crohns disease-associated bacterial protein I2 is a novel enteric
T cell superantigen. Immunity 2001;15:14958.
47. Lodes MJ, Cong Y, Elson CO, Mohamath R, Landers CJ, Targan
SR, et al. Bacterial flagellin is a dominant antigen in Crohn
disease. J Clin Invest 2004;113:1296306.
48. MacDonald TT, Monteleone G. Immunity, inflammation, and
allergy in the gut. Science 2005;307:19205.
49. Kraehenbuhl JP, Neutral MR. Epithelial M cells: differentiation
and function. Annu Rev Cell Dev Biol 2000;16:30132.
50. Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G,
Bonasio R, et al. Dendritic cells express tight junction proteins
and penetrate gut epithelial monolayers to sample bacteria. Nat
Immunol 2001;2:3617.
51. Niess JH, Brand S, Gu X, Landsman L, Jung S, McCormick
BA, et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 2005;307:254
8.
52. Yoshida M, Claypool SM, Wagner JS, Mizoguchi E, Mizoguchi A,
Roopenian DC, et al. Human neonatal Fc receptor mediates
transport of IgG into luminal secretions for delivery of antigens
to mucosal dendritic cells. Immunity 2004;20:76983.
53. Yoshida M, Kobayashi K, Kuo TT, Bry L, Glickman JN, Claypool
SM, et al. Neonatal Fc receptor for IgG regulates mucosal
immune responses to luminal bacteria. J Clin Invest 2006;116:
214251.
54. Li H, Nowak-Wegrzyn A, Charlop-Powers Z, Shreffler W,
Chehade M, Thomas S, et al. Transcytosis of IgE-antigen complexes by CD23a in human intestinal epithelial cells and its role
in food allergy. Gastroenterology 2006;131:4758.
55. Kelsall BL, Leon F. Involvement of intestinal dendritic cells in
oral tolerance, immunity to pathogens, and inflammatory bowel
disease. Immunol Rev 2005;206:13248.
56. Rescigno M. CCR6(+) dendritic cells: the gut tactical-response
unit. Immunity 2006;24:50810.

A. Mizoguchi and E. Mizoguchi: IBD past, present and future


57. Iwasaki A. Mucosal dendritic cells. Annu Rev Immunol 2007;25:
381418.
58. Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell 2001;106:2558.
59. Gonzalez-Rey E, Delgado M. Therapeutic treatment of experimental colitis with regulatory dendritic cells generated with
vasoactive intestinal peptide. Gastroenterology 2006;131:1799
811.
60. Pedersen AE, Gad M, Kristensen NN, Haase C, Nielsen CH,
Claesson MH. Tolerogenic dendritic cells pulsed with enterobacterial extract suppress development of colitis in the severe combined immunodeficiency transfer model. Immunology 2007;121:
52632.
61. Ordway D, Henau-Tamayo M, Orme IM, Gonzalez-Juarrero M.
Foamy macrophages within lung granulomas of mice infected
with Mycobacterium tuberculosis express molecules characteristic of dendritic cells and antiapoptotic markers of the TNF receptor-associated factor family. J Immunol 2005;175:387381.
62. Sakaguchi S, Fukuma K, Kuribayashi K, Masuda T. Organspecific autoimmune disease induced in mice by elimination of
T cell subset. I evidence for the active participation of T cells in
natural self-tolerance; deficit of a T cell subset as a possible cause
of autoimmune disease. J Exp Med 1985;161:7287.
63. Powrie F, Mason D. OX-22high CD4+ T cells induce wasting disease
with multiple organ pathology: prevention by the OX-22low
subset. J Exp Med 1990;172:17018.
64. Makita S, Kanai T, Nemoto Y, Totsuka T, Okamoto R, Tsuchiya
K, et al. Intestinal lamina propria retaining CD4+CD25+ regulatory T cells is a suppressive site of intestinal inflammation. J
Immunol 2007;178:493746.
65. Sakaguchi S. Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses.
Annu Rev Immunol 2004;22:53162.
66. Izcue A, Coombes JL, Powrie F. Regulatory T cells suppress systemic and mucosal immune activation to control intestinal
inflammation. Immunol Rev 2006;212:25671.
67. Roncarolo MG, Battaglia M. Regulatory T cell immunotherapy
for tolerance to self antigens and alloantigens in humans. Nat
Rev Immunol 2007;7:58598.
68. Zheng Y, Rudensky AY. Foxp3 in control of the regulatory T cell
lineage. Nat Immunol 2007;8:45762.
69. Iweala O, Nagler CR. Immune privilege in the gut: the establishment and maintenance of non-responsiveness to dietary antigens
and commensal flora. Immunol Rev 2006;213:82100.
70. Harris DP, Haynes L, Sayles PC, Duso DK, Eaton SM,
Lepak NM, et al. Reciprocal regulation of polarized cytokine
production by effector B and T cells. Nat Immunol 2000;1:475
82.
71. Mizoguchi A, Mizoguchi E, Takedatsu H, Blumberg RS, Bhan
AK. Chronic intestinal inflammatory condition generates IL-10
producing regulatory B cell subset characterized by CD1d upregulation. Immunity 2002;16:21930.
72. Fillatreau S, Sweenie CH, McGeachy MJ, Gray D, Anderton SM.
B cells regulate autoimmunity by provision of IL-10. Nat
Immunol 2002;3:94450.
73. Mauri C, Gray D, Mushtaq N, Londei M. Prevention of arthritis
by interleukin 10-producing B cells. J Exp Med 2003;197:
489501.
74. Burke F, Stagg AJ, Bedford PA, English N, Knight SC. IL-10producing B220+CD11c APC in mouse spleen. J Immunol
2004;173:236272.
75. Duddy ME, Alter A, Bar-Or A. Distinct profiles of human B cell
effector cytokines: a role in immune regulation? J Immunol
2004;172:34227.
76. Mangan NE, Fallon RE, Smith P, van Rooijen N, McKenzie AN,
Fallon PG. Helminth infection protects mice from anaphylaxis
via IL-10-producing B cells. J Immunol 2004;173:634656.
77. Wagner M, Poeck H, Jahrsdoerfer B, Rothenfusser S, Prell D,
Bohle B, et al. IL-12p70-dependent Th1 induction by human B

13

78.

79.

80.

81.

82.
83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

93.

94.

95.
96.

97.

cells requires combined activation with CD40 ligand and CpG


DNA. J Immunol 2004;172:95463.
Gillan V, Lawrence RA, Devaney E. B cells play a regulatory
role in mice infected with the L3 of Brugia Pahangi. Int Immunol
2005;17:37382.
Cohen-Sfady M, Nussbaum G, Pevsner-Fischer M, Mor F, Carmi
P, Zanin-Zhorov A, et al. Heat shock protein 60 activates B cells
via the TLR4-MyD88 pathway. J Immunol 2005;175:3594602.
Matsumura Y, Byrne SN, Nghiem DX, Miyahara Y, Ullrich SE.
A role for inflammatory mediators in the induction of immunoregulatory B cells. J Immunol 2006;177:48107.
Blenman KRM, Duan B, Xu ZW, Wan SG, Atkinson MA, Flotte
TR, et al. IL-10 regulation of lupus in the NZM2410 murine
model. Lab Invest 2006;86:113648.
Inoue S, Leitner WW, Golding B, Scott D. Inhibitory effects of B
cells on antitumor immunity. Cancer Res 2006;66:77417.
Kamanaka M, Kim ST, Wan YY, Sutterwala FS, Lara-Tejero M,
Galan JE, et al. Expression of interleukin-10 in intestinal lymphocytes detected by an interleukin-10 receptor knock in tiger
mouse. Immunity 2006;25:94152.
Kobayashi M, Fitz L, Ryan M, Hewick RM, Clark SC, Chan S,
et al. Identification and purification of natural killer cell stimulatory factor (NKSF) a cytokine with multiple biologic effects on
human lymphocytes. J Exp Med 1989;170:82737.
Liu N, Ohnishi N, Ni L, Akira S, Bacon KB. CpG directly induces
T-bet expression and inhibits IgG1 and IgE switching in B cells.
Nat Immunol 2003;4:68793.
Rachmilewitz D, Karmeli F, Takabayashi K, Hayashi T, LeiderTrejo L, Lee J, et al. Immunostimulatory DNA ameliorates
experimental and spontaneous murine colitis. Gastroenterology
2002;122:142841.
Rachmilewitz D, Katakura K, Karmeli F, Hayashi T, Reinus C,
Rudensky B, et al. Toll-like receptor 9 signaling mediates the
anti-inflammatory effects of probiotics in murine experimental
colitis. Gastroenterology 2004;126:5208.
Katakura K, Lee J, Rachmilewitz D, Li G, Eckmann L, Raz E.
Toll like receptor 9-induced type I IFN protects mice from experimental colitis. J Clin Invest 2005;115:695702.
Zhang GX, Gran B, Yu S, Li J, Siglienti I, Chen X, et al. Induction
of experimental autoimmune encephalomyelitis in IL-12 receptor 2 deficient mice: IL-12 responsiveness is not required in the
pathogenesis of inflammatory demyelination in the central
nervous system. J Immunol 2003;170:215360.
Mizoguchi A, Mizoguchi E, Smith RN, Preffer FI, Bhan AK.
Suppressive role of B cells in chronic colitis of T cell receptor
mutant mice. J Exp Med 1997;186:174956.
Mizoguchi E, Mizoguchi A, Preffer FI, Bhan AK. Regulatory
function role of mature B cells in a murine model of inflammatory bowel disease. Int Immunol 2000;12:597605.
Gerth AJ, Lin L, Neurath MF, Glimcher LH, Peng SL. An innate
cell-mediated, murine ulcerative colitis-like syndrome in the
absence of nuclear factor of activated T cells. Gastroenterology
2004;126:111521.
Hokama A, Mizoguchi E, Sugimoto K, Shimomura Y, Tanaka Y,
Yoshida M, et al. Induced reactivity of intestinal CD4+ T cells
with an epithelial cell lectin, galectin-4, contributes to exacerbation of intestinal inflammation. Immunity 2004;20:68193.
Wei B, Velazquez P, Turovskaya O, Spricher K, Aranda R,
Kronenberg M, et al. Mesenteric B cells centrally inhibit CD4+
T cell colitis through interaction with regulatory T cell subsets.
Proc Natl Acad Sci U S A 2005;102:20105.
Mizoguchi A, Bhan AK. A case for regulatory B cells. J Immunol
2006;176:70510.
Ostanin DV, Pavlick KP, Bharwani S, DSouza D, Furr KL, Brown
CM, et al. T cell-induced inflammation of the small and large
intestine in immuno-deficient mice. Am J Physiol 2006;290:
G10919.
McGeachy MJ, Stephens LA, Anderton SM. Natural recovery
and protection from autoimmune encephalomyelitis: contribu-

14

98.

99.

100.

101.

102.

103.

104.

105.

106.
107.

108.

109.

110.

111.

112.

113.

114.

115.

116.

117.

A. Mizoguchi and E. Mizoguchi: IBD past, present and future


tion of CD4+CD25+ regulatory cells within the central nervous
system. J Immunol 2005;175:302532.
Milani M, Ostlie N, Wu H, Wang W, Conti-Fine BM. CD4+ T and
B cells cooperate in the immunoregulation of experimental autoimmune myasthenia gravis. J Neuroimmunol 2006;179:15262.
Ashour HM, Niederkorn JY. Peripheral tolerance via the anterior chamber of the eye: role of B cells in MHC class I and II
antigen presentation. J Immunol 2006;176:59507.
Olson TS, Bamias G, Naganuma M, Rivera-Nieves J, Burcin TL,
Ross W, et al. Expanded B cell population blocks regulatory T
cells and exacerbates ileitis in a murine model of Crohns disease.
J Clin Invest 2004;114:38998.
Dohi T, Fujihashi K, Koga T, Shirai Y, Kawamura YI, Ejima C,
et al. T helper type-2 cells induce ileal villus atrophy, goblet cell
metaplasia, and wasting disease in T cell-deficient mice. Gastroenterology 2003;124:67282.
Kawamura T, Kanai T, Dohi T, Uraushihara K, Totsuka T, Iiyama
R, et al. Ectopic CD40 ligand expression on B cells triggers
intestinal inflammation. J Immunol 2004;172:638897.
Hugot JP, Chamaillard M, Zouali H, Lesage S, Cezard JP,
Belaiche J, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohns disease. Nature 2001;411:
599603.
Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos
R, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohns disease. Nature 2001;411:6036.
Inohara, Chamaillard, McDonald C, Nunez G. NOD-LRR proteins: role in host-microbial interactions and inflammatory
disease. Annu Rev Biochem 2005;74:35583.
Baumgart DC, Carding SR. Inflammatory bowel disease: cause
and immunobiology. Lancet 2007;369:162740.
Pauleau AL, Murray PJ. Role of nod2 in the response of macrophages to toll-like receptor agonists. Mol Cell Biol 2003;23:
75319.
Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara
N, Nunez G, et al. Nod2-dependent regulation of innate and
adaptive immunity in the intestinal tract. Science 2005;307:
7314.
Maeda S, Hsu LC, Liu H, Bankston LA, Iimura M, Kagnoff MF,
et al. Nod2 mutation in Crohns disease potentiates NF-kappaB
activity and IL-1beta processing. Science 2005;307:7348.
Cash HL, Whitham CV, Behrendt CL, Hooper LV. Symbiotic
bacteria direct expression of an intestinal bactericidal lectin.
Science 2006;313:112630.
Watanabe T, Kitani A, Murray PJ, Strober W. NOD2 is a negative
regulator of toll-like receptor 2-mediated T helper type 1
responses. Nat Immunol 2004;5:8008.
Watanabe T, Kitani A, Murray PJ, Wakatsuki Y, Fuss IJ, Strober
W. Nucleotide binding oligomerization domain 2 deficiency leads
to dysregulated TLR2 signaling and induction of antigen-specific
colitis. Immunity 2006;25:47385.
Barnich N, Hisamatsu T, Aguirre JE, Xavier R, Reinecker HC,
Podolsky DK. GRIM-19 interacts with nucleotide oligomerization domain 2 and serves as downstream effector of antibacterial function in intestinal epithelial cells. J Biol Chem
2005;280:190216.
Chen CM, Gong Y, Zhang M, Chen JJ. Reciprocal cross-talk
between Nod2 and TAK1 signaling pathways. J Biol Chem
2004;279:2587682.
McDonald C, Chen FF, Ollendorff V, Ogura Y, Marchetto S,
Lecine P, et al. A role for Erbin in the regulation of Nod2dependent NF-kappaB signaling. J Biol Chem 2005;280:403019.
Yamamoto-Frusho JK, Barnich N, Xavier R, Hisamatsu T,
Podolsky DK. Centaurin beta 1 down-regulates nucleotidebinding oligomerization domains 1 and 2 dependent NF-kappaB
activation. J Biol Chem 2006;281:3606070.
Hsu YM, Zhang Y, You Y, Wang D, Li H, Duramad O, et al. The
adaptor protein CARD9 is required for innate immune responses
to intracellular pathogens. Nat Immunol 2007;8:198205.

118. Inoue N, Tamura K, Kinouchi Y, Fukuda Y, Takahashi S, Ogura


Y, et al. Lack of common NOD2 variants in Japanese patients
with Crohns disease. Gastroenterology 2002;123:8691.
119. Yamazaki K, Onouchi Y, Takazoe M, Kubo M, Nakamura Y, Hata
A. Association analysis of genetic variants in IL-23R, ATG16L1
and 5p13.1 loci with Crohns disease in Japanese patients. J Hum
Genet 2007;52:57583.
120. Medzhitov R, Preston-Hurlburt P, Janeway Jr CA. A human
homologue of the Drosophila toll protein signals activation of
adaptive immunity. Nature 1997;388:3947.
121. Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y,
et al. Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the LPS gene
product. J Immunol 1999;162:374952.
122. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev
Immunol 2003;21:33576.
123. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol 2004;5,98795.
124. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and
innate immunity. Cell 2006;24:783801.
125. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S,
Medzhitov R. Recognition of commensal microflora by toll-like
receptors is required for intestinal homeostasis. Cell 2004;118:
22941.
126. Fukata M, Michelsen KS, Eri R, Thomas LS, Hu B, Lukasek K,
et al. Toll-like receptor-4 is required for intestinal response to
epithelial injury and limiting bacterial translocation in a murine
model of acute colitis. Am J Physiol 2005;288:G105565.
127. Araki A, Kanai T, Ishikura T, Makita S, Uraushihara K,
Iiyama R, et al. MyD88-deficient mice develop severe intestinal
inflammation in dextran sodium sulfate colitis. J Gastroenterol
2005;40:1623.
128. Cario E, Gerken G, Podolsky DK. Toll-like receptor 2 controls
mucosal inflammation by regulating epithelial barrier function.
Gastroenterology 2007;132:135974.
129. Kitajima S, Morimoto M, Sagara E, Shimizu C, Ikeda Y. Dextran
sodium sulfate-induced colitis in germ-free IQI/Jic mice. Exp
Anim 2001;50:38795.
130. Fort MM, Mozaffarian A, Stover AG, Correia Jda S, Johnson
DA, Crane RT, et al. A systemic TLR4 antagonist has antiinflammatory effects in two murine models of inflammatory
bowel disease. J Immunol 2005;174:641623.
131. Grabig A, Paclik D, Guzy C, Dankof A, Baumgart DC,
Erckenbrecht J, et al. Infect Immun 2006;74:407582.
132. Kabashima K, Saji T, Murata T, Nagamachi M, Matsuoka T, Segi
E, et al. The prostaglandin receptor EP4 suppresses colitis,
mucosal damage and CD4 cell activation in the gut. J Clin Invest
2002;109:88393.
133. Lin F, Spencer D, Hatala DA, Levine AD, Medof ME. Decayaccelerating factor deficiency increases susceptibility to dextran
sulfate sodium-induced colitis: role for complement in inflammatory bowel disease. J Immunol 2004;172:383641.
134. Matsuura M, Okazaki K, Nishio A, Nakase H, Tamaki H, Uchida
K, et al. Therapeutic effects of rectal administration of basic
fibroblast growth factor on experimental murine colitis. Gastroenterology 2005;128:97586.
135. Mizoguchi E, Xavier RJ, Reinecker H-C, Uchino H, Bhan AK,
Podolsky DK, et al. Colonic epithelial functional phenotype
varies with type and phase of experimental colitis. Gastroenterology 2003;125:14861.
136. Mizoguchi E. Chitinase 3-like 1 exacerbates intestinal inflammation by enhancing bacterial adhesion and invasion in colonic
epithelial cells. Gastroenterology 2006;130:398411.
137. Liu H, Komai-Koma M, Xu D, Liew FY. Toll-like receptor 2 signaling modulates the functions of CD4+CD25+ regulatory T cells.
Proc Natl Acad Sci USA 2006;103:704853.
138. Rakoff-Nahoum S, Hao L, Medzhitov R. Role of toll-like receptors in spontaneous commensal dependent colitis. Immunity
2006;25:31929.

A. Mizoguchi and E. Mizoguchi: IBD past, present and future


139. Sanders CJ, Yu Y, Moore DA, Williams IR, Gewirtz AT. Humoral
immune response to flagellin requires T cells and activation of
innate immunity. J Immunol 2006;177:28108.
140. Gewirtz AT, Vijay-Kumar M, Brant SR, Duerr RH, Nicolae DL,
Cho JH. Dominant-negative TLR5 polymorphism reduces adaptive immune response to flagellin and negatively associates with
Crohns disease. Am J Physiol Gastrointest Liver Physiol
2006;290:G115763.
141. Targan SR, Landers CJ, Yang H, Lodes MJ, Cong Y, Papadakis
KA, et al. Antibodies to Cbir1 flagellin define a unique response
that is associated independently with complicated Crohns
disease. Gastroenterology 2005;128:20208.
142. Papadakis KA, Yang H, Ippoliti A, Mei L, Elson CO, Hershberg
RM, et al. Anti-flagellin (CBir1) phenotypic and genetic Crohns
disease associations. Inflamm Bowel Dis 2007;13:52430.
143. Gewirtz AT, Navas TA, Lyons S, Godowski PJ, Madara JL.
Bacterial flagellin activates basolaterally expressed TLR5 to
induce epithelial proinflammatory gene expression. J Immunol
2001;167:18825.
144. Rhee SH, Im E, Riegler M, Kokkotou E, Obrien M, Pothoulakis
C. Pathophysiological role of toll-like receptor 5 engagement by
bacterial flagellin in colonic inflammation. Proc Natl Acad Sci
USA 2005;102:136105.
145. Krieg AM, Yi AK, Matson S, Waldschmidt TJ, Bishop GA,
Teasdale R, et al. CpG motifs in bacterial DNA trigger direct
B-cell activation. Nature 1995;374:5469.
146. Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H,
et al. A toll-like receptor recognizes bacterial DNA. Nature
2000;408:7405.
147. Lee J, Mo JH, Katakura K, Alkalay I, Rucker AN, Liu YT, et al.
Maintenance of colonic homeostasis by distinctive apical TLR9
signaling in intestinal epithelial cells. Nat Cell Biol 2006;8:
132736.
148. Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat Rev Drug Discov 2006;5:47184.
149. Rachmilewitz D, Karmeli F, Shteingart S, Lee J, Takanayashi K,
Raz E. Immunostimulatory oligonucleotides inhibit colonic proinflammatory cytokine production in ulcerative colitis. Inflamm
Bowel Dis 2006;12:33945.
150. Obermeier F, Straugh UG, Dunger N, Grunwald N, Rath HC,
Herfarth H, et al. In vivo CpG DNA/toll-like receptor 9 interaction induces regulatory properties in CD4+CD62L+ T cells which
prevent intestinal inflammation in the SCID transfer model of
colitis. Gut 2005;54:142836.
151. Obermeier F, Dunger N, Deml L, Herfarth H, Scholmeric J, Falk
W. CpG motifs of bacterial DNA exacerbate colitis of dextran
sulfate sodium-treated mice. Eur J Immunol 2002;32:208492.
152. Obermeier F, Dunger N, Strauch UG, Hofmann C, Bleich A,
Grunwald N, et al. CpG motifs of bacterial DNA essentially
contribute to the perpetuation of chronic intestinal inflammation. Gastroenterology 2005;129:91327.
153. Kuwata H, Matsumoto M, Atarashi K, Morishita H, Hirotani T,
Koga R, et al. IBNS inhibits induction of a subset of Toll-like
receptor-dependent genes and limits inflammation. Immunity
2006;24:4151.
154. Vijay-Kumar M, Wu H, Aitken J, Kolachala VL, Neish AS, Sitaraman SV, et al. Activation of toll-like receptor 3 protects against
DSS-induced acute colitis. Inflamm Bowel Dis 2007;13:85664.
155. Hooper LV, Gordon JI. Commensal host-bacterial relationship
in the gut. Science 2001;292:11158.
156. Sartor RB. Therapeutic manipulation of the enteric microflora in
inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology 2004;126:162033.
157. Sonnenburg JL, Xu J, Leip DD, Chen CH, Westover BP,
Weatherford J, et al. Glycan foraging in vivo by an intestineadapted bacterial symbiont. Science 2005;307:19559.
158. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunoregulatory molecules of symbiotic bacteria directs maturation
of the host immune system. Cell 2005;122:10718.

15
159. Barnich N, Cavalho FA, Glasser AL, Darcha C, Jantscheff P,
Allez M, et al. CEACAM6 acts as a receptor for adherentinvasive E. coli, supporting ileal mucosa colonization in Crohn
disease. J Clin Invest 2007;117:156674.
160. Karrasch T, Kim J-S, Muhlbauer M, Magness ST, Jobin C. Gnotobiotic IL-10/;NF-BEGFP mice reveal the critical role of
TLR/NF-B signaling in commensal bacteria-induced colitis. J
Immunol 2007;178:652232.
161. Dahan S, Roth-Walter F, Arnaboldi P, Agarwal S, Mayer L. Epithelia: lymphocyte interactions in the gut. Immunol Rev 2007;
215:24353.
162. Shirazi T, Longman RJ, Corfield AP, Probert CS. Mucins and
inflammatory bowel disease. Postgrad Med J 2000;76:4738.
163. Rhodes JM. Unifying hypothesis for inflammatory bowel disease
and associated colon cancer: sticking the pieces together with
sugar. Lancet 1996;347:404.
164. An G, Wei B, Xia B, McDaniel JM, Ju T, Cummings RD, et al.
Increased susceptibility to colitis and colorectal tumors in mice
lacking core 3-derived O-glycans. J Exp Med 2007;204:1417
29.
165. Van der Sluis M, De Koning BAE, De Bruijn ACJM, Velcich A,
Meijerink JPP, Van Goudoever JB, et al. Muc2-deficient mice
spontaneously develop colitis, indicating that MUC2 is critical
for colonic protection. Gastroenterology 2006;131:11729.
166. Ho SB, Dvorak LA, Moor RE, Jacobson AC, Frey MR, Corredor
J, et al. Cysteine-rich domains of muc3 intestinal mucin promote
cell migration, inhibit apoptosis, and accelerate wound healing.
Gastroenterology 2006;131:150117
167. McAuley JL, Linden SK, Png CW, King RM, Pennington HL,
Gendler SJ, et al. Muc1 cell surface mucin is a critical element
of the mucosal barrier to infection. J Clin Invest 2007;117:
231324.
168. Beatty PL, Plevy SE, Sepulveda AR, Finn OJ. Transgenic expression of human MUC1 in IL-10/ mice accelerates inflammatory
bowel disease and progression to colon cancer. J Immunol 2007;
179:7359.
169. Mashimo H, Wu DC, Podolsky DK, Fishman MC. Impaired
defence of intestinal mucosa in mice lacking intestinal trefoil
factor. Science 1996;274:2625.
170. Itoh H, Beck PL, Inoue N, Xavier R, Podolsky DK. A paradoxical
reduction in susceptibility to colonic injury upon targeted
transgenic ablation of goblet cells. J Clin Invest 1999;104:
153947.
171. McVay LD, Keilbaugh SA, Wong TM, Kierstein S, Shin ME,
Lehrke M, et al. Absence of bacterially induced RELM reduces
injury in the dextran sodium sulfate model of colitis. J Clin Invest
2006;116:291423.
172. Hogan SP, Seidu L, Blanchard C, Groschwitz K, Mishra A, Karow
ML, et al. Resistin-like molecule regulates innate colonic function: barrier integrity and inflammation susceptibility. J Allergy
Clin Immunol 2006;118:25768.
173. Mizoguchi E, Mizoguchi A, Takedatsu H, Cario E, de Jong YP,
Ooi CJ, et al. Role of tumor necrosis factor receptor 2 (TNFR2)
in colonic epithelial hyperplasia and chronic intestinal inflammation. Gastroenterology 2002;122:13444.
174. Brandl K, Plitas G, Schnabl B, DeMatteo RP, Pamer EG. MyD88mediated signals induce the bacterial lectin RegIII and protect
mice against intestinal Listeria monocytogenes infection. J Exp
Med 2007;204:1891900.
175. Allez M, Tieng V, Nakazawa A, Treton X, Pacault V, Dulphy N,
et al. CD4+NKG2D+ T cells in Crohns disease mediate inflammatory and cytotoxic responses through MICA interactions.
Gastroenterology 2007;132:234658.
176. He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J,
et al. Intestinal bacteria trigger T cell-independent immunoglobulin A(2) class switching by inducing epithelial-cell secretion of
the cytokine APRIL. Immunity 2007;26:81226.
177. Xu W, He B, Chiu A, Chadburn A, Shan M, Buldys M, et al.
Epithelial cells trigger frontline immunoglobulin class switching

16

178.

179.

180.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.
191.

192.

193.

194.

195.

A. Mizoguchi and E. Mizoguchi: IBD past, present and future


through a pathway regulated by the inhibitor SLPI. Nat Immunol
2007;8:294303.
Rimoldi M, Chieppa M, Salucci V, Avogadri F, Sonzogni A,
Sampietro GM, et al. Intestinal immune homeostasis is regulated
by the crosstalk between epithelial cells and dendritic cells. Nat
Immunol 2005;6:50714.
Atreya R, Mudter J, Finotto S, Mullberg J, Jostock T, Wirtz S,
et al. Blockade of interleukin 6 trans signaling suppresses T-cell
resistance against apoptosis in chronic intestinal inflammation:
evidence in Crohn disease and experimental colitis in vivo. Nat
Med 2000;6:5838.
Tebbutt NC, Giraud AS, Inglese M, Jenkins B, Waring P, Clay FJ,
et al. Reciprocal regulation of gastrointestinal homeostasis by
SHP2 and STAT-mediated trefoil gene activation in gp130
mutant mice. Nat Med 2002;8:108997.
Neurath MF, Pettersson S, Meyer zum Buschenfelde KH, Strober
W. Local administration of antisense phosphorothioate oligonucleotides to the p65 subunit of NF-kappa B abrogates established
experimental colitis in mice. Nat Med 1996;2:9981004.
Shibata W, Maeda S, Hikiba Y, Yanai A, Ohmae T, Sakamoto K,
et al. The IB kinase (IKK) inhibitor, NEMO-binding domain
peptide, blocks inflammatory injury in murine colitis. J Immunol
2007;179:26815.
Nenci A, Becker C, Wullaret A, Gareus R, van Loo G, Dansen S,
et al. Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature 2007;446:55761.
Plevy S. A STAT need for human immunologic studies to understand inflammatory bowel disease. Am J Gastroenterol 2005;100:
734.
Okamoto R, Yajima T, Yamazaki M, Kanai T, Mukai M, Okamoto
S, et al. Damaged epithelia regenerated by bone marrow-derived
cells in the human gastrointestinal tract. Nat Med 2002;8:
10117.
Suzuki K, Oida T, Hamada H, Hitotsumatsu O, Watanabe M, Hibi
T, et al. Gut cryptopatches: direct evidence of extrathymic anatomical sites for intestinal T lymphopoiesis. Immunity 2000;13:
691702.
Chinen H, Matsuoka K, Sato T, Kamada N, Okamoto S,
Hisamatsu T, et al. Lamina propria c-kit+ immature precursors
reside in human adult intestine and differentiate into natural
killer cells. Gastroenterology 2007;133:55973.
Khalil PN, Weiler V, Nelson PJ, Khalil MN, Moosmann S,
Mutschler WE, et al. Nonmyeloablative stem cell therapy
enhances microcirculation and tissue regeneration in murine
inflammatory bowel disease. Gastroenterology 2007;132:94454.
Brittan M, Chance V, Elia G, Poulsom R, Alison MR, MacDonald
TT, et al. A regenerative role for bone marrow following experimental colitis: contribution to neovasculogenesis and myofibroblasts. Gastroenterology 2005;128:198495.
Yoshimori T. Autophagy: paying Charons toll. Cell 2007;128:
8336.
Rioux JD, Xavier RJ, Taylor KD, Silverberg MS, Goyette P, Huett
A, et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease
pathogenesis. Nat Genet 2007;39:596604.
Prescott NJ, Fisher SA, Franke A, Hampe J, Onnie CM, Soars D,
et al. A nonsynonymous SNP in ATG16L1 predisposes to ileal
Crohns disease and is independent of CARD15 and IBD5.
Gastroenterology 2007;132:166571.
Xu Y, Jagannath C, Liu X-D, Sharafkhaneh A, Kolodziejska KE,
Eissa NT. Toll-like receptor 4 is a sensor for autophagy associated
with innate immunity. Immunity 2007;27:13544.
Qu X, Zou Z, Sun Q, Luby-Phelps K, Cheng P, Hogan RN, et al.
Autophagy gene-dependent clearance of apoptotic cells during
embryonic development. Cell 2007;128:93146.
Schmid D, Pypaert M, Munz C. Antigen-loading compartments
for major histocompatibility complex class II molecules continuously receive input from autophagosomes. Immunity 2007;26:
7992.

196. Schimid D, Munz C. Innate and adaptive immunity through


autophagy. Immunity 2007;27:1121.
197. Steinhoff U, Brinkmann V, Klemm U, Aichele P, Seiler P, Brandt
U, et al. Autoimmune intestinal pathology induced by hsp60 specific CD8 T cells. Immunity 1999;11:34957.
198. Mizoguchi A, Mizoguchi E, Saubermann LJ, Higaki K, Blumberg
RS, Bhan AK. Limited CD4 T-cell diversity associated with
colitis in T-cell receptor mutant mice requires a T helper 2
environment. Gastroenterology 2000;119:98395.
199. Bamias G, Okazawa A, Rivera-Nieves J, Arseneau KO, De La
Rue SA, Pizarro TT, et al. Commensal bacteria exacerbate intestinal inflammation but are not essential for the development of
murine ileitis. J Immunol 2007;178:180918.
200. Taganov KD, Boldin MP, Baltimore D. MicroRNAs and immunity: tiny players in a big field. Immunity 2007;26:1337.
201. Thai T-H, Calado DP, Casola S, Ansel KM, Xiao C, Xue Y, et al.
Regulation of the germinal center response by microRNA-155.
Science 2007;316:6047.
202. Continho LL, Matukumalli LK, Sonstegard TS, Van Tassell CP,
Gasbarre LC, Capuco AV, et al. Discovery and profiling of bovine
microRNAs from immune-related and embryonic tissues. Physiol
Genomics 2007;29:3543.
203. Lowe JB. Glycosylation, immunity, and autoimmunity. Cell 2001;
104:80912.
204. Baum LG. Developing a taste for sweets. Immunity 2002;16:
58.
205. Crocker PR, Paulson JC, Varki A. Siglecs and their roles in the
immune system. Nat Rev Immunol 2007;7:25566.
206. Van Vliet SJ, Gringhuis SI, Geijtenbeek TBH, van Kooyk Y.
Regulation of effector T cells by antigen-presenting cells via
interaction of the C-type lectin MGL with CD45. Nat Immunol
2006;7:12008.
207. Green RS, Stone EL, Tennon M, Lehtonen E, Farquhar MG,
Marth JD. Mammalian N-glycan branching protects against
innate immune self-recognition and inflammation in autoimmune disease pathogenesis. Immunity 2007;27:113
208. Uchimura K, Rosen SD. Sulfated l-selectin ligands as a therapeutic target in chronic inflammation. Trends Immunol 2006;27:
55965.
209. Gringhuis SI, den Dunnen J, Litjens M, van her Hof B, van Kooyk
Y, Geijtenbeek TBH. C-type lectin DC-SIGN modulates toll-like
receptor signaling via Raf-1 kinase-dependent acetylation of
transcription factor NF-B. Immunity 2007;26:60516.
210. Cunningham AL, Harman AN, Donaghy H. DC-SIGN AIDS
HIV immune evasion and infection. Nat Immunol 2007;8:
5568.
211. Perillo NJ, Pace KE, Seihamer JJ, Baum LG. Apoptosis of T cells
mediated by galectin-1. Nature 1995;378:7369.
212. Toscano MA, Bianco GA, Iiarregui JA, Croci DO, Correale J,
Hernandez JD, et al. Differential glycosylation of Th1, Th2, and
Th17 effector cells selectively regulates susceptibility to cell
death. Nat Immunol 2007;8:82534.
213. Santucci L, Fiorucci S, Rubinstein N, Mencarelli A, Palazzetti B,
Federici B, et al. Galectin-1 suppresses experimental colitis in
mice. Gastroenterology 2003;124:138194.
214. Ideo H, Seko A, Ohkura T, Matta KL, Yamashita K.
High affinity binding of recombinant human galectin-4 to
SO(3)()->3Galbeta1->3GalNAc pyranoside. Glycobiology
2002;12:199208.
215. Mizoguchi E, Mizoguchi A. Is the sugar always sweet in intestinal
inflammation? Immunol Res 2007;37:4760
216. Mizoguchi A, Mizoguchi E, Chiba C, Bhan AK. Appendix
lymphoid follicle plays an important role for development of
inflammatory bowel disease in TCR- mutant mice. J Exp Med
1996;184:70715.
217. Hegazi RAF, Rao KN, Mayle A, Sepulveda AR, Otterbein LE,
Plevy SE. Carbon monoxide ameliorates chronic murine colitis
through a heme oxygenase 1-dependent pathway. J Exp Med
2005;202:170313.

A. Mizoguchi and E. Mizoguchi: IBD past, present and future


218. Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, et al.
Generation of gut-homing IgA-secreting B cells by intestinal
dendritic cells. Science 2006;314:115760.
219. Coombes JL, Siddiqui KRR, Arancibia-Carcamo CV, Hall J, Sun
C-M, Belkaid Y, et al. A functionally specialized population of
mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a
TGF- and retinoic acid-dependent mechanism. J Exp Med
2007;204:175764.
220. Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ. AA-trans
retinoic acid mediates enhanced Treg cell growth, differentiation,
and gut homing in the face of high levels of co-stimulation. J Exp
Med 2007;2004:176574.
221. Sun C-M, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR,
et al. Small intestine lamina propria dendritic cells promote de
novo generation of Foxp3 Treg cells via retinoic acid. J Exp Med
2007;204:177585.

17
222. Sugimoto K, Hanai H, Tozawa K, Aoshi T, Uchijima M, Nagata
T, et al. Curcumin prevents and ameliorates trinitrobenzene sulfonic acid-induced colitis in mice. Gastroenterology 2002;123:
191222.
223. Arita M, Yoshida M, Hong S, Tjonahen E, Glickman JN, Petasis
NA, et al. Resolvin E1, an endogenous lipid mediator derived
from omega 3 eicosapentaenoic acid, protects against 2,4,6trinitrobenzene sulfonic acid-induced colitis. Proc Natl Acad
Sci USA 2005;102:76716.
224. Hudert CA, Weylandt KH, Lu Y, Wang J, Hong S, Dignass A,
et al. Transgenic mice rich in endogenous omega-3 fatty acids
are protected from colitis. Proc Natl Acad Sci USA 2006;103:
1127681.

Das könnte Ihnen auch gefallen