Sei sulla pagina 1di 12

Expert Review of Gastroenterology & Hepatology

ISSN: 1747-4124 (Print) 1747-4132 (Online) Journal homepage: http://www.tandfonline.com/loi/ierh20

New molecular insights into inflammatory bowel


disease-induced diarrhea

Yueming Tang, Christopher B Forsyth & Ali Keshavarzian

To cite this article: Yueming Tang, Christopher B Forsyth & Ali Keshavarzian (2011) New
molecular insights into inflammatory bowel disease-induced diarrhea, Expert Review of
Gastroenterology & Hepatology, 5:5, 615-625, DOI: 10.1586/egh.11.64

To link to this article: http://dx.doi.org/10.1586/egh.11.64

Published online: 10 Jan 2014.

Submit your article to this journal

Article views: 47

View related articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ierh20

Download by: [McMaster University] Date: 09 January 2017, At: 19:59


Review
For reprint orders, please contact reprints@expert-reviews.com

New molecular insights into


inflammatory bowel
disease-induced diarrhea
Expert Rev. Gastroenterol. Hepatol. 5(5), 615625 (2011)

Yueming Tang1, Diarrhea is one of the common symptoms that significantly affects quality of life in patients
Christopher B Forsyth1,2 with inflammatory bowel disease (IBD). The clinical manifestation of diarrhea is mainly dependant
and on the type of IBD and the location, extent and severity of intestinal inflammation. Understanding
the pathophysiologic mechanisms of diarrhea in patients with IBD will be beneficial to developing
AliKeshavarzian1,3,4
effective treatments for IBD-associated diarrhea. In recentyears, modern molecular techniques
1
Department of Internal Medicine, have been used intensively to dissect the role of the intestinal microbiota, epithelial barrier and
Division of Digestive Disease and
Nutrition, Rush University Medical the host immune system in the mechanisms of IBD-induced diarrhea. These studies have
Center, Chicago, IL 60612, USA significantly advanced our knowledge of the mechanisms of IBD-induced diarrhea. In this article,
2
Department of Biochemistry, Division we focus on the new and critical molecular insights into the contributions of the intestinal
of Digestive Disease and Nutrition,
microbiota, epithelial tight junctions, proinflammatory cytokines and microRNA as potential
Rush University Medical Center,
Chicago, IL 60612, USA mechanisms underlying to IBD-induced diarrhea.
3
Department of Pharmacology,
Division of Digestive Disease and Keywords : diarrhea epithelial barrier inflammatory bowel diseases intestinal microbiota microRNA
Nutrition, Rush University Medical permeability proinflammatory cytokines tight junction
Center, Chicago, IL 60612, USA
4
Department of Molecular Biophysics
and Physiology, Division of Digestive
Inflammatory bowel diseases (IBDs) are com- General pathophysiology of diarrhea
Disease and Nutrition, Rush University mon, chronic inflammatory disorders of the gut inIBD
Medical Center, Chicago, with unknown etiology. The incidence of IBDs To maintain the normal physiological function
IL60612,USA

Author for correspondence:


have greatly increased in developed, as well as of the GI tract, the intestinal epithelium has
Tel.: +1 312 942 8927 developing countries, in recent years[1] . IBD finely tuned mechanisms to maintain ionic bal-
Fax: +1 312 942 5664 patients have many symptoms, such as abdomi- ance, fluid absorption and secretion [4,7,8] . The
yueming_tang@rush.edu
nal pain, vomiting, diarrhea, rectal bleeding various ionic transport proteins/complexes in the
and weight loss, that can result in significant apical, basal or basolateral membranes of intesti-
disability, loss of work, poor quality of life and nal epithelial cells are important for maintaining
sometimes even surgery and death. intestinal homeostasis. Na+, K+, H+ and Cl- ion
Diarrhea is one of the common symptoms transport proteins are regulated by endogenous
in patients with IBD[2,3] . Approximately 50% mediators (cAMP, cGMP, PGE2, free radicals,
of acute flare-ups of Crohns disease (CD) and histamine, cytokines and Ca 2+) and dietary-
almost 100% of ulcerative colitis (UC) patients related factors (bile acids, short-chain fatty acids
experience diarrhea[4] . The clinical manifesta- through different molecular mechanisms[4,7,8] .
tions of diarrhea are diverse in individual patients. In certain pathophysiologic conditions, the bal-
It is dependent on the type of IBD and the loca- ance of ionic and fluid exchange is disrupted and
tion, extent and severity of inflammation[5,6] . To fails to maintain the homeostasis required to
effectively treat diarrhea in patients with IBD, compensate for the pro-absorptive/antisecretary
it is important to define the pathophysiologic mechanisms[4,7,8] . This dysfunctional homeos-
mechanisms in individual patients. tasis results in excessive secretion of Na+ and Cl-
The underlying mechanisms of diarrhea associ- ions followed by the release of a large amount of
ated with IBD are complex[7,8] . The contributions water into the colonic lumen; this phenomenon
of inflammation and epithelial tight junction per- is manifested as diarrhea which is a common
meability to diarrhea in IBD have recently been symptom in patients with IBD[4,7] .
studied[911] . In this article we focus on recent The overall fluid absorption in the small
progress and new molecular insights relevant to and large intestines was described in a recent
the mechanisms of diarrhea in IBD. review[7] . The fluid load to the small intestine

www.expert-reviews.com 10.1586/EGH.11.64 2011 Expert Reviews Ltd ISSN 1747-4124 615


Review Tang, Forsyth & Keshavarzian

is about 8l per day. The small intestine absorbs about 6l of The importance of the host immune system, intestinal micro
fluid per day with ileocecal flow being approximately 2l per organisms (microbiota) and the intestinal barrier in the mecha-
day. The large intestine absorbs about 1.8l of fluid per day. nisms of diarrhea have been investigated in a large number of
The maximal absorptive capacity of the colon is approximately studies[7,911,19,20] . The interactions between microbiota, intes-
4.55.0l. The fluid in the stool is normally less than 0.2l per tinal barrier and host immune system are complicated during
day. The net water absorption is directly proportional to net normal physiological status or under inflammatory stimula-
Na+ and Cl- absorption and is driven by osmotic gradients[4,7,8] . tion[7,911,19,20] . These studies indicate that there are many
Active Na + and Cl- secretion/absorption is the driving force for molecular mechanisms responsible for the generation of diarrhea
the fluid absorption. Thus, it is critical to understand the role with a wide range of clinical presentation. Thus, understanding
of active Na + absorption and active Cl- secretion in cellular and the molecular mechanisms of diarrhea is important in identifying
molecular mechanisms associated with the diarrhea in IBD. novel molecular targets for the successful treatment of patients
It has been reported that there are at least five different Na + with IBD.
absorptive processes[7] : The intestinal epithelial barrier plays an important role in host
defense by serving as a critical fence between invading pathogens
Nutrient-stimulated Na+ absorption is the primary mechanism
and the host immune system. The integrity of the intestinal barrier
for either glucose- or amino acid-stimulated post-prandial fluid
depends on both healthy epithelial cells and on an intact paracel-
absorption in the small intestine;
lular pathway, which appears to be the main route for permea-
Na + H + exchange is responsible for the absorption of large tion of macromolecules such as endotoxins[21] . This pathway is a
amounts of HCO3 derived from biliary and pancreatic secretion complex array of structures that includes tight junctions between
in the duodenum and proximal jejunum. This Na + H + gut epithelial cells. Tight junctions function as gates that regulate
exchange is inhibited by increased cAMP levels; intestinal permeability[2225] . These dynamic tight junctions are
highly regulated and are able to change their size under various
Coupled Na+ Cl- absorption in the distal small intestine and
physiological and pathological conditions[26] . During intestinal
proximal colon is the primary pathway for fluid absorption in
inflammation, the intestinal barrier is disrupted. This inflamma-
the interdigestive period, inhibited by increased cAMP levels
tion-induced intestinal barrier dysfunction results in ions and water
and intracellular Ca 2+;
passively diffusing from the circulation to the intestinal lumen
Short-chain fatty acid-stimulated Na+ absorption is observed and causes leak flux diarrhea[8,10,11] . Under normal physiological
in the colon. Short-chain fatty acids are synthesized by colonic conditions, only a very limited amount of bacterial antigens and
bacteria from nonabsorbed carbohydrates; macromolecules cross the epithelial fence[10] . However, during
inflammation, the intestinal barrier is disturbed and the passage of
Aldosterone-sensitive Na+ absorption via the Na+ channel in
antigens is increased[10] . On the luminal side, invading pathogens
the distal colon represents a scavenger mechanism for fluid and
can disrupt the epithelial barrier and increase intestinal permeabil-
Na+ retention.
ity through releasing a variety of agents including pore-forming
The homeostasis status of ionic balance, fluid absorption and toxin, cytoskeleton-modifying proteins and bacterial lipopolysac-
secretion was studied in patients with UC using perfusion and charide[9] . Thus, some pathogens can cross the epithelial barrier
dialysis technology[1214] . The molecular mechanisms of dis- into the basal side to interact with the host immune system. On
turbed electrolyte transport in intestinal inflammation were the basal side, the pathogen-activated immune cells also disrupt
reviewed by others[4] . The net Na+, Cl- and water absorption the intestinal barrier to increase intestinal permeability and facili-
was reduced in UC patients without any evidence of net fluid, tate the pathogen invasion through secreting proinflammatory
Na+ or Cl- secretion[1214] . Further studies using an invitro flux cytokines such as IFN-g, TNF-a and IL-1b[9] . The contribution
chamber to measure the net ionic absorption and short-circuit of the microbiota, intestinal barrier and host immune system to
current in colonic mucosa from UC patients also confirmed the mechanisms of diarrhea are discussed in the following section.
the above observations[15,16] . The function of the Na+ channel
which is responsible for aldosterone-induced Na+ absorption in Diarrhea & the intestinal microbiota
the epithelial apical membrane in the distal colon of UC patients There are a vast range of microorganisms that colonize the mam-
was impaired[17] . Other studies indicated that both protein and malian GI tract. These microorganisms are known as intestinal
mRNA levels of down-regulated in adenoma (DRA), which is the microbiota, that are required for intestinal homeostasis and func-
apical membrane transporter responsible for colonic Cl-HCO3- tion and appear to play a role in the pathogenesis of IBD[19,20,27,28] .
exchange, were reduced in the colonic mucosa of patients with Host innate and adaptive immune systems respond to intestinal
UC[18] . These studies suggest that the impairment of apical microbiota through different pathways. One of the pathways is
membrane transport proteins, Na+ channel for Na+ absorption that of Toll-like receptors (TLRs), which can recognize conserved
and DRA for Cl- absorption, results in reduced active Na+ and microbial molecules. Another pathway is that of Tregs that can
Cl- absorption and diarrhea in UC[18] . These dysfunctions in ion maintain tolerance and immune homeostasis to prevent chronic
transport were also observed in the colonic mucosa of patients inflammation[20,27,28] . However, the specific contribution of the
with active CD[13,15] . intestinal mircobiota to the pathogenesis of IBD and the role of

616 Expert Rev. Gastroenterol. Hepatol. 5(5), (2011)


New molecular insights into inflammatory bowel disease-induced diarrhea Review

the microbiota in mechanisms for IBD-associated diarrhea has not etal. reported that starch-entrapped microspheres show a ben-
been systematically investigated. The colonization of the intesti- eficial fermentation profile and decrease in potentially harmful
nal microbiota plays an important role in the host physiological bacterial during invitro fermentation in fecal microbiota obtained
processes, such as nutrient uptake and exchange, the formation from patients with IBDs[47] . Recently, another study showed that
and stabilization of the intestinal barrier and the development of new prebiotics from rice bran ameliorate inflammation in murine
the intestinal immune system[19,20,27,28] . colitis models through the modulation of the intestinal mircobiota
The microbial community in the gut is complex. Owing to and the mucosal immune system[48] .
methodological limitations, the diversity of intestinal microbiota In humans, one recent study reported that Escherichia coli
is still incompletely characterized and the role of the microbiota in phylogenetic group B2 was associated with IBD patients with
IBD mechanisms remains poorly defined[19,20] . Modern molecu- distal disease activity[49] . Under different environmental or
lar techniques are rapidly advancing our research progress and inflammatory conditions, the expressions of some specific bacte-
knowledge of the intestinal microbiota[19,20,29] . Previous micro- rial species increased or decreased and the overall compositions
biology studies of the intestinal microbiota were mainly depend- of the intestinal microbiota also changed[20,50,51] . These observed
ent on culture techniques. Such cultivation has only been able changes in intestinal microbiota are termed dysbiosis[20,50,51] . It
to characterize a very small fraction of the microbial diversity in remains unclear whether the dysbiosis is a causative factor or a side
the gut. The recent molecular ecological studies based on 16S effect in the mechanisms of human IBDs[20] . Thus, the effects
ribosomal RNA sequences have revealed the characterization and of different environmental factors or inflammatory conditions on
quantification of the gut microbiota[19,20,2729] . The composition the intestinal microbiota need to be further investigated.
of the mucosa and luminal microbiota has been investigated in The dysbiosis in infectious colitis is characterized by a decrease
humans and mice using large-scale analyses of 16S rDNA genes of Faecalibacterium prausnitzii [52] . The metabolites from F.praus-
and metagenomics, which are culture-independent methods[20,30] . nitzii can block NF-kB activation and reduce IL-8 secretion from
The number of species constituting the human gastrointestinal Caco-2 cells and increase anti-inflammatory IL-10 and reduce
microbiota may be as high as 15,000 species as detected by 16S proinflammatory IL-12 release from peripheral blood mono-
deep sequencing analyses[20,31] . Recent studies have shown that cytes[53] . In the healthy GI tract, E.coli is the most numeri-
the intestinal microbiota vary not only between individuals[20,30] , cally dominant Gram negative species[19] . One important study
but also with anatomical location[20,30] , inflammatory status of reported that the numbers of E.coli in the mucosa of CD patients
IBD[32] , diet[19,20,33] and alcohol consumption[34] . The impact were increased[54] . These E.coli isolated from the mucosa showed
of gut microbiota on the pathogenesis of mouse IBD models has an increased ability to adhere to intestinal epithelial cells and to
been extensively reviewed[20] . The gut microbiota of patients with disrupt the intestinal barrier by producing an a hemolysin[54] .
IBD has been studied by several groups[3538] . Their studies show This is a new pathotype of E.coli termed adhesive and invasive
that there is a compositional shift in the IBD-associated micro- E. coli, which stimulates the release of the proinflammatory
biota[32,37] , including depletion of specific types of commensal cytokine IL-8[55] . The microfold (M) cells make up approximately
bacteria (Lachnospiraceae and Bacteroidetes) and enrichment in 5% of the epithelial cells that overlie Peyers patches in the intes-
Proteobacteria, using phylogenetic comparisons of the mucosa and tine and lyphoid follicles in the colon[56,57] . Recent studies indi-
luminal microbiota of patients with IBDs and non-IBDs con- cate that the Mcell is a portal of entry for pathogens, such as the
trols[32] . However, the specific species associated with intestinal adhesive and invasive E.coli that were found in CD lesions where
inflammation or a specific pathogenic pattern of microbiota has Mcells were located[19,56,57] . Mcells have the unique ability to
not been identified in samples from patients with IBDs[3538] . sample antigens from the gut microbiota[19] . They play an impor-
In murine intestinal inflammation models, including induc- tant role in mucosal immunity by presenting antigens from a wide
tion by bacterial infection (Citrobacter rodentium, Salmonella range of microorganisms, including bacteria, viruses and parasites,
enterica)[3941] , dextran sulfate sodium-induced colitis[40,42] or a from the gut lumen to dendritic cells and lymphocytes[19,58] .
genetic deficiency IL-10-/- mouse model[40] , the intestinal micro- Several studies have indicated that intestinal bacteria such as
biota was changed. The major alteration of the microbiota in the Campylobacter jejuni RM1221 are able to disrupt the epithelial
inflammatory model was the reduction in both the total number barrier and increase paracellular permeability to allow noninvasive
of resident bacteria and the diversity of microbiota[3941] . Garrett organisms to translocate through the epithelial layer[19,59] . When
etal. reported that Enterobacteriaceae act in concert with the gut the mucosal barrier has been weakened, the TLR5 located on the
microbiota to induce spontaneous and maternally transmitted basolateral aspect of epithelial cells may become accessible to bacte-
colitis[43] . The alteration of the murine gut microbiota during rial flagellin[60] . The repositioning of TLRs increases the interac-
infection with the parasitic helminth Heligmosomoides polygyrus tion between epithelial cells and gut microbiota, leading to acti-
was reported by Walk etal.[44] . A pyrosequencing study in twins vation of NF-kB and induction of inflammation[60] . It has been
showed that the profiles of gut microbiota varied with IBD phe- reported that soluble fiber from edible plantains prevents adherence
notypes[45] . One recent study demonstrated that the probiotic to the epithelium by E.coli isolated from IBD patients[19,61,62] .
VLS#3 alters the composition of gut microbiota and these changes The hostmicrobe interaction in the GI tract has been reviewed
correlate with VSL#3-induced disease protection in a 2,4,6-trini- and summarized by other groups[20,27,40,51,63] . These stud-
trobenzenesulfonic acid-induced colitis mouse model[46] . Rose ies indicated that there are many pathways by which intestinal

www.expert-reviews.com 617
Review Tang, Forsyth & Keshavarzian

bacteria can attenuate or aggravate the development of intestinal changes in expression and distribution of claudins-2, -5 and -8
inflammation[63,64] . These pathways include: lead to discontinuous tight junctions and barrier dysfunction
in active CD[69] . Epithelial cell apoptosis also increased two to
Influencing the inflammatory process and modulating oxidative
threefold in patients with active CD[69] . Since inflammatory
stress via TLR signaling and NF-kB activation;
cytokine-induced epithelial cell apoptosis can contribute to intes-
Influencing intestinal permeability through regulation of tight tinal barrier dysfunction[10] , it is reasonable to conclude that the
junction proteins expression and translocation; epithelial apoptosis contributes to the leakiness and diarrhea in
IBDs. NOD2 is the gene product of CARD15, which is expressed
Influencing the composition of the mucus layer. The gut micro-
in human intestinal epithelial cells and is upregulated syner-
biota not only interferes with the expression of MUC genes,
gistically by TNF-a and IFN-g[70] . CARD15 mutations were
but also affects the expression and/or activity of cell glycosyl
found in patients with CD and their first-degree relatives[71,72] .
transferases, which can induce changes in the carbohydrate
Furthermore, these CARD15 mutations were correlated with bar-
repertoire of mucins[64] ;
rier dysfunction and increased intestinal permeability in CD
Influencing resistance to harmful stimuli and epithelial repair patients[71,72] . In clinical studies, diarrhea in IBD patients was
through TLR4 and NF-kB signaling; significantly improved after treatment with TNF-a antibodies
through downregulation of epithelial apoptosis and upregulation
Influencing the production and release of immune effective
of epithelial barrier repair to normalize the elevated intestinal per-
molecules, such as IgA.
meability (leakiness)[7] . These studies support the importance of
More mechanisms for the hostmicrobe interaction in the GI the intestinal barrier in the mechanism of IBD-induced diarrhea.
tract will be revealed in the future as our methods to investigate The above observations both in patients with IBD and in ani-
these interactions become more sophisticated. mal models of IBD indicate that proinflammatory cytokines
In conclusion, recent advances in the molecular technology and can increase intestinal permeability. On the other hand, there is
microbiology provide us with a much more robust knowledge of another possibility that the increase of permeability (disruption
the role of the mucosal and fecal microbiota in IBD. The role of of intestinal barrier) may be responsible for the initiation of IBD
the gut microbiota in the mechanisms of IBD and IBD associated by increasing the translocation of bacteria and bacterial compo-
diarrhea have been investigated by several groups[3538] . However, nents to the lamina propria and thus increasing the exposure of
the specific contribution of the intestinal mircobiota to the patho- lamina propria immune cells to bacteria antigens. This possibility
genesis of IBD and the role of the microbiota in mechanisms for is supported by the fact that mucosal permeability was increased
IBD associated diarrhea have not been systematically investigated. in asymptomatic IBD patients and their relatives[7] . Thus, it is
One recent study showed that the invasion of bacteria resulted still unclear whether the intestinal barrier dysfunction is the
in an increase in the interaction between bacterial components, chicken or the egg for IBD-associated diarrhea.
such as flagellin, with basolateral receptors (e.g.,TLR5), leading Taken together, epithelial barrier dysfunction contributes to
to activation of NF-kB and inflammation as a consequence[60] . diarrhea in IBD by a leak flux mechanism and causes an increase
of luminal antigen uptake, thus, resulting in mucosal inflamma-
Diarrhea & intestinal barrier function tion. Tight junctions are important components of the intestinal
The intestinal barrier consists of epithelial cells and the tight junc- epithelial barrier. The detailed molecular pathways for regula-
tions between the cells[7,9,10,65] . Reduction or disruption of tight tion of tight junctions and the cell cytoskeleton in the epithelial
junctions results in an increase in intestinal permeability. The role barrier during intestinal inflammation have been reviewed in
of permeability or tight junctions in mechanisms of IBD have other papers[9,66] . The epithelial tight junctions are regulated by
been studied[7,9,10,65] . Disruption and dysfunction of the intestinal cytokines and other inflammatory factors in the mechanisms of
barrier in IBD contributes to diarrhea by the leak flux mechanism IBD-induced diarrhea. The immune regulation of the epithelial
and also increases the uptake of luminal antigens, which interact barrier, intestinal permeability and ion transport in IBDs will be
with immune cells to induce intestinal inflammation. discussed in the next section.
It is well accepted that mucosal inflammation causes an
increase of intestinal epithelial permeability[9] . The inflamma- Diarrhea & immune dysfunction (T-cell activation,
tion-induced hyperpermeability plays an important role in the inflammation & cytokines)
pathophysiology of IBD[9,66] . Intestinal barrier dysfunction in There is significant and persistent inflammation in the colonic
CD has been shown in previous studies using invivo permeability mucosa of patients with IBD. The ongoing inflammation might
tests[11] . The intestinal barrier dysfunction is positively correlated induce diarrhea through triggering the release of proinflammatory
with the degree of inflammation in IBD patients[67] . Intestinal cytokines from immune cells. These cytokines not only affect
permeability can predict the clinical relapse of CD[68] . However, electrolyte transport but also affect the tight junctions in the
the underlying molecular mechanisms for the barrier dysfunction intestinal epithelium (Figure1) . The cytokine-induced abnormal
have only recently been elucidated[9,11] . One study reported that ion transport and disruption of tight junctions contribute to
occludin, claudin-5 and claudin-8 were downregulated in CD, the inflammation-induced gut leakiness and the mechanisms of
while the pore forming claudin-2 was upregulated[69] . These IBD-associated diarrhea.

618 Expert Rev. Gastroenterol. Hepatol. 5(5), (2011)


New molecular insights into inflammatory bowel disease-induced diarrhea Review

Inflammation mediators
Gut microbiota DSS, TNBS in animal models
Diarrhea Genetic susceptibility
Lumen Diet
Alcohol
Na+, H2O

Ion transports TLRs


TJ
TJ NF-B
TJ cAMP
Epithelial cells
cGMP Intestinal
Ca2+ miRNAs
barrier

Proinflammatory
Lamina iNOS cytokines (TNF, IFN)
propria ROS
Histamine
Kinins
Principal pathways Prostaglandins
discussed in this review PAF Immune cells
Possible pathways not Other cells
discussed in this review

Expert Rev. Gastroenterol. Hepatol. Future Science Group (2011)

Figure1. Model for the pathogenesis of irritable bowel disease-induced diarrhea.


DSS: Dextran sulfate sodium; IFN: Interferon; iNOS: Inducible nitric oxide synthase; miRNA: MicroRNA; PAF: Platelet activating factor;
ROS: Reactive oxygen species; TJ: Tight junction; TLR: Toll-like receptor; TNBS: 2,4,6-Trinitrobenzenesulfonic acid.

The effects of proinflammatory cytokines on colonic ion trans- Mucosal T-cell activation is associated with diarrhea in patients
port have been studied by several groups[4,7,8] . Their studies with IBDs[73] . Musch etal. reported that T-cell activation with
focused on the effects of TNF-a, IFN-g and IL-1b on the altera- anti-CD3 monoclonal antibodies induces profound diarrhea in
tion of Na+/H + exchange, Cl- /HCO3 - exchange, Na channels mice, quantified by intestinal weight-to-length ratios[73] . T-cell
and Na+/K+ -ATPase[4,7,8] . These investigations show that these activation significantly increased permeability to mannitol at 1h
three cytokines downregulate all of the transporters but do not and decreases in electroneutral Na+ absorption, Na+ -dependent
increase Cl- secretion[4,7,8] . These results indicate that proinflam- glucose absorption and cAMP-stimulated anion secretion at 3h.
matory cytokines, mainly act on the absorptive processes without Furthermore, enteral fluid accumulation was observed in CFTR-/-
inducing secretory processes. These proinflammatory cytokines mice, indicating a minor role of active anion secretion in T-cell
present in the colonic mucosa of patients with IBD, play an activation induced diarrhea. Their studies suggest that diarrhea
important role in the regulation of intestinal permeability and in IBD is due to TNF-a-mediated abnormal absorption rather
inflammation. This suggests that the proinflammatory cytokine- than to secretory processes. T-cell activation induces luminal fluid
induced alteration of ion transport in the colonic mucosa is a accumulation by increasing mucosal permeability and reducing
principal mechanism responsible for diarrhea in patients with epithelial Na+/K+ -ATPase activity leading to decreased intestinal
IBDs. Most of the diarrhea occurring in patients with IBDs is Na+ and water absorption[73] .
characterized by a decrease in the fluid and solute absorption, Disruption of the intestinal epithelial barrier occurs in IBDs,
but not net fluid and solute secretion[7] . The fact that clinical but neither the mechanisms nor the contribution of the bar-
use of monoclonal antibodies to TNF-a (infliximab) can effec- rier dysfunction to the diarrhea mechanisms have been clearly
tively control the diarrhea in patients with IBDs also supports defined. Clayburgh etal. reported that epithelial barrier dysfunc-
the fact that cytokine-induced changes of ion transport play an tion is required for the development of diarrhea, using a murine
important role in the diarrhea mechanism of IBDs[7] . Since only model of T-cell-mediated acute diarrhea to investigate the role
3540% of the patients treated with infliximab have a significant of the epithelial barrier in diarrheal disease, such as IBD[74] .
remission, it is likely that additional factors may be responsible The T-cell activation-induced diarrhea is characterized by the
for the diarrhea in these patients[7] . The tight junctions in the reversal of net water flux, from absorption to secretion; increased
gut epithelium may be one of these additional pathways that leakage of serum proteins into the intestinal lumen; and altered
proinflammatory cytokines may affect to induce gut leakiness tight junction structure[74] . T-cell activation was associated with
during intestinal inflammation[7] . increased phosphorylation of epithelial myosin II regulatory light

www.expert-reviews.com 619
Review Tang, Forsyth & Keshavarzian

chain, which is correlated with tight junction regulation and the in TG mice[77] . They also demonstrate that T-cell activation-
development of diarrhea[74] . T-cell activation-induced epithelial induced net water secretion was reversed and T-cell activation-
myosinII regulatory light chain phosphorylation, tight junction induced luminal flux of different molecular probes (bovine
disruption, protein leak and diarrhea are prevented in genetic serum albumin, alexa350 and dextran3000) was reduced, using
knockout mice of long myosin light chain kinase (MLCK) or in vivo perfusion loop studies in TG mice[77] . Their studies
treatment of wild-type mice with a highly specific peptide MLCK also confirmed that tight junction proteins (occludin, claudin-1
inhibitor[74] . These studies indicate that barrier dysfunction is and zonula occludens-1) are internalized through an NF-kB-
critical to the pathogenesis of diarrheal disease in this IBD model dependent pathway[77] . Taken together, these studies suggest
and that inhibition of epithelial MLCK may be an effective non- that IBD-associated diarrhea results from NF-kB-mediated
immunosuppressive therapy for treatment of immune-mediated tight junction protein internalization and increased paracellular
intestinal disease. permeability[77] . Since epithelial NF-kB activation induced by
Using the same T-cell activation model, Clayburgh etal. demon- mucosal Tcells occurs in patients with IBDs and actively plays a
strated that although TNF-a-neutralizing antibodies completely role in opening paracellular spaces to promote transmucosal fluid
restore water absorption after systemic T-cell activation, barrier effux into the intestinal lumen, NF-kB may be a molecular target
function is only partially corrected[75] . This suggests that, while for effective therapy of IBD[77] . Reduction of epithelial NF-kB
barrier dysfunction is critical, other processes must be involved in activation in IBDs might thus repair defects in the epithelial
T-cell-mediated diarrhea. To study the mechanism during these barrier and reduce diarrhea.
processes invivo, this group designed a series of experiments to Although epithelial NF-kB plays an important role in regulation
investigate the role of TNF-a and LIGHT[76] , a new member of of intestinal barrier function and inflammation in IBD, whether
the TNF-a superfamily, in the regulation of barrier function and NF-kB directly affects the NHE and/or other epithelial transport
the mechanisms of T-cell activation-induced diarrhea. Their stud- function is unclear. Recently, Amin etal. reported that TNF-a
ies show that both TNF-a and LIGHT caused MLCK-dependent represses the expression of NHE2 through NF-kB activation in
barrier dysfunction[75] . However, while TNF-a caused diarrhea, an intestinal epithelial cell model[78] . TNF-a levels are increased
LIGHT enhanced intestinal water absorption. Moreover, TNF-a, in IBD patients. To study the effect of TNF-a on the expression
but not LIGHT, inhibited Na+ absorption due to TNF-a -induced and activity of NHE2, which is involved in transepithelial Na+
internalization of the brush border Na+/H+ exchanger Na+/H+ absorption in intestinal epithelial cells and plays an important role
exchanger (NHE)3[75] . LIGHT did not cause NHE3 internali- in diarrhea mechanism of IBD, they used TNF-a-treated Caco-2
zation. PKC activation by TNF-a was responsible for NHE3 cells. The NHE2 regulations were measured by reverse transcrip-
internalization and pharmacological or genetic PKCa inhibition tion-PCR, reporter gene assays and Western blot analysis. Their
prevented NHE3 internalization, Na+ absorption and diarrhea studies showed that the human NHE2 isoform is a direct target
despite continued barrier dysfunction[75] . The studies also of the transcription factor NF-kB. TNF-a-mediated activation
indicated that cytokine-induced barrier dysfunction may be an of NF-kB decreases the expression and activity of NHE2 in the
important mechanism in the genesis of diarrhea. However, barrier intestinal epithelial cell line. Their findings indicated that NF-kB
dysfunction does not appear to be the only cause responsible for plays an important role in the modulation of Na+ absorption dur-
IBD-associated diarrhea, other pathways may also be involved as ing intestinal inflammatory conditions such as IBD where a high
mechanisms promoting IBD diarrhea. level of TNF-a is detected[78] .
Although these studies provide molecular insight into mecha-
nisms of intestinal water transport by coordinating Na+ absorp- Diarrhea & microRNA
tion and barrier dysfunction in TNF-a-induced diarrhea, there MicroRNAs (miRNAs) are highly conserved regulatory mol-
are some contradictory data regarding the critical role of barrier ecules expressed in eukaryotic cells. They are short noncoding
dysfunction and diarrhea. In addition, these studies have some RNAs that regulate gene expression by binding to target mRNAs,
limitations in that all the studies were performed in mice with which leads to reduced protein synthesis and sometimes decreased
anti-CD3 monoclonal antibodies and not in patients with IBDs. steady-state mRNA levels[79] . Although hundreds of miRNAs
Aberrant activation of innate and adaptive immune responses have been identified, much less is known about their biological
increase mucosal permeability in IBD, although the mechanisms function[8082] . There is evidence that miRNAs affect pathways
are not completely understood[77] . To examine the role of epi- fundamental to metabolic control in higher organisms such as
thelial NF-kB in IBD-induced enhanced permeability, Tang adipocyte and skeletal muscle differentiation. Furthermore, some
etal. generated epithelial-specific IkBa mutant (NF-kB super miRNAs are implicated in lipid, amino acid and glucose homeos-
repressor) transgenic (TG) mice[77] . Their studies show that tasis[83] . Thus, miRNA abnormalities may contribute to common
NF-kB activation was inhibited in TG mice following T-cell- metabolic diseases and there may be novel therapeutic opportu-
mediated immune cell activation using an anti-CD3 monoclonal nities based on miRNA targeting[81] . Recently, the profiles of
antibody[77] . Furthermore, T-cell activation-induced diarrhea miRNA in IBDs were identified in the mucosa of patients with
was inhibited in TG mice. Meanwhile, T-cell activation-induced IBD[8486] . The role of miRNA in the development of intestinal
changes in transepithelial resistance and transmucosal flux of inflammation was investigated. These studies suggest that miR-
alexa350 (0.35kDa) and dextran3000 (3kDa) were blocked NAs are critical regulators for immune response in IBDs and

620 Expert Rev. Gastroenterol. Hepatol. 5(5), (2011)


New molecular insights into inflammatory bowel disease-induced diarrhea Review

may be novel molecular targets for pharmacological modulation In summary, recent studies provide compelling evidence for
of mucosa inflammation[8486] . However, the role of miRNA in the role of miRNAs in the mechanisms underlying IBD-induced
the mechanisms of IBD-induced diarrhea has not been defined. diarrhea. Therefore, miRNAs represent new therapeutic targets
In a recent study we showed the possibility that miRNAs are for the prevention and/or treatment of inflammation-induced
involved in the disruption of tight junction proteins such as tight diarrhea in patients with IBD.
junction protein zonula occludens-1 (ZO-1)[87] . Thus, inflam-
mation-induced gut leakiness could be owign to the effects of Diarrhea & other mechanisms
proinflammatory cytokines on the expression of miRNAs that In the above sections we discuss how the intestinal microbiota,
target tight junction genes in intestinal epithelial cells. epithelial barrier, host immunity and miRNAs contribute to IBD-
We studied the role of miR-212 in alcohol-induced ZO-1 dis- induced diarrhea through regulating the alteration of ion transport
ruption in intestinal epithelial cells because ZO-1 is predicted, and barrier function during intestinal inflammation. Other mecha-
in an established miRNA database, to be one of miR-212s target nisms may also be involved in the diarrhea of patients with IBD[7] .
genes[88,89] . We were the first to show that miR-212 is highly These mechanisms include: ileal dysfunction and bile-acid diarrhea,
expressed in intestinal tissues using a TaqMan miRNA real- steatorrhea (fatty acid-induced diarrhea), bacterial overgrowth
time PCR assay[87] . We also showed that miR-212 overexpres- syndromes, lactose intolerance and short bowel syndrome[7] .
sion is accompanied by reductions in ZO-1 protein expression, These mechanisms were previously briefly reviewed[7] . In order
disruption of ZO-1 and increased permeability of monolayers of to focus our article on the new and critical molecular insights for
Caco-2 cells. miR-212 overexpression correlated with alcohol- IBD-induced diarrhea, other molecules, such as the putative anion
induced disruption of monolayer integrity. Most importantly, transport 1, DRA, cystic fibrosis transmembrane regulator, cAMP,
we found that miR-212 levels in colon biopsy samples in IBD cGMP, Ca2+, iNOS, free radicals, prostaglandins, histamine from
patients were higher than in healthy controls. To see if miR-212 mast cells, kinins from neurons, platelet factors and complement
regulates ZO-1 levels, we conducted both overexpression studies cascade components, are not included in this article, even though
using miR-212 precursors and inhibition studies using miR-212- they may also contribute to the diarrheal mechanisms in IBD[4] .
specific antisense oligonucleotide inhibitors (anti-miR-212). The Genetic risk factors of IBD may also be important for the
data showed that miR-212 overexpression significantly inhibited molecular mechanism of diarrhea in IBD patients. Recent stud-
ZO-1 protein expression and knocking down of miR-212 expres- ies have indicated some genetic deficiencies associated with the
sion in Caco-2 cells using anti-miR-212 inhibited alcohol-induced mechanisms of IBD. The genetic susceptibility of IBD has been
hyperpermeability by 50% (p<0.05)[87] . well reviewed[91] . However, the contribution of genetic risk fac-
The potential importance of miRNAs in intestinal inflamma- tors to IBD-induced diarrhea is unknown. To date, no study
tion has recently been demonstrated[8486] . Wu etal. measured directly addresses the role of genetic deficiency in the mechanism
miRNA expression in the intestinal mucosa of patients with active of diarrhea in IBD. We believe that the genetic deficiency may
UC, inactive UC, CD, irritable bowel syndrome, infectious colitis contribute to the diarrhea mechanism through affects on intesti-
and microscopic colitis, as well as in healthy subjects by micro- nal microbiota and/or barrier function and/or immune function
array and reverse transcription-PCR[86] . Their results showed which are discussed in this article.
that 11 miRNAs were differentially expressed in active UC (eight Successful treatment of IBD-associated diarrhea is dependant
increased and three decreased significantly). miR-192 expres- on defining the pathophysiologic mechanism in each individual
sion was decreased in colonic epithelial cells in active UC[86] . patient. Diarrhea continues to be a major prevalent symptom in
Macrophage inflammatory peptide-2 was identified as a target patients with IBD. Physicians should properly evaluate the com-
of miR-192. In colonic epithelial cells, TNF-a, a proinflam- plaints of diarrhea in IBD patients by assessing both patient symp-
matory cytokine that is present in high levels in the inflamed toms and potential physiologic impacts on fluid and electrolyte
intestine, induced miR-192 reduction and macrophage inflam- status[2,3] . When treating diarrhea, physicians should pay atten-
matory peptide-2 overexpression[86] . miRNAs not only appear to tion to the following conditions: the location, extent and severity
be involved in gut inflammation, but they may also be involved of inflammation; malabsorption; altered motility; and iatrogenic
in the regulation of intestinal functions such as fluid secretion. causes such as medications, diet and antibiotic-associated colitis
For example, Kapeller etal. reported an association between a (e.g., Clostridium difficile). Therapies for IBD-associated diarrhea
functional variant of miR-510, the target site of the serotonin include aminosalicylates, corticosteroids, immune modifiers and,
receptor-type 3E gene (HTR3E) and diarrhea predominant irrita- most recently, biologic treatment[2,3] . Other medications, includ-
ble bowel syndrome[90] . Their studies indicated that miRNA-510 ing loperamide, diphenoxylate, codeine sulfate and tinctures of
regulated HTR3E expression and when a cis-regulatory variant opium slow motility and an increase in the absorption of fluids
was present in the HTR3E gene, the regulation of miR-510 ceased and nutrients[2,3] . The mechanisms of diarrhea in different indi-
to be effective. This variant of the miRNA target site is associated vidual IBD patients are not the same; thus, it is important to select
with female diarrhea predominant irritable bowel syndrome. It an individual therapy strategy for each patient according to the
remains to be seen whether miRNAs that target genes that are specific mechanisms that appear to predominate in that patient.
key in regulation of intestinal inflammation also play a role in Algorithms for selection of antidiarrheal agents have been well
IBD-associated diarrhea mechanisms. reviewed by other studies[2,3] .

www.expert-reviews.com 621
Review Tang, Forsyth & Keshavarzian

Conclusion Expert commentary & five-year view


The molecular mechanisms for IBD-induced diarrhea are sum- More new molecular techniques, such as mutitag pyrosequencing
marized in Figure 1. The specific mechanism(s) for individual combined with bioinformatics and network-based modeling, will be
patients are different. One or more mechanisms may be involved used to identify the specific microbiota responsible for IBD-induced
in the pathogenesis of IBDs and IBD-associated diarrhea. Thus, diarrhea. The specific pathogenic microorganisms in the human
it is important to select therapies for each individual IBD patient gut as a cause of IBD may be identified soon. Treatment based on
according to the specific mechanisms. The roles of the intesti- the modulation of gut microbiota using specific diets or antibiotics
nal microbiota, epithelial barrier and host immune system in or probiotic bacteria may be developed into clinicaltrials.
the mechanisms of IBD-induced diarrhea have been extensively The mechanisms of inflammation induced epithelial barrier
studied. Substantial progress has been achieved recently in under- dysfunction will be further investigated. The effect of therapeutic
standing the interactions between the microbiota, intestinal bar- agents[92] , such as TGF-b, zinc and quercetin, on the intestinal bar-
rier and the host immune system during normal physiology or rier function will be studied. Therapeutic agents will be developed to
under inflammatory conditions. These studies have significantly modulate or repair the epithelial barrier defects in patients with IBD.
improved our knowledge of the mechanisms of IBD-induced Mucosal inflammation-induced increase of epithelial perme-
diarrhea. Modern molecular techniques provide us with a very ability is a common phenomenon in many diseases. The increase
useful tool to investigate the role of the mucosal and fecal micro- of intestinal permeability results in endotoxemia and multi-organ
biota in the mechanisms of IBD-induced diarrhea. Intestinal dysfunction. Understanding the mechanisms would be helpful in
epithelial barrier dysfunctions contribute to the diarrhea in IBD the development of new therapeutic approaches to prevent and
by a leak flux mechanism. The dysfunction of the epithelial bar- treat these diseases through preserving integrity of the epithelial
rier results in an increase of intestinal permeability, increase of barrier. To effectively prevent or treat the diarrhea or hyperperme-
luminal antigens uptake and initiation of mucosal inflammation. ability in IBD, the molecular mechanism for proinflammatory
The epithelial tight junctions are regulated by cytokines and cytokines to disrupt the tight junctions has to be investigated in
other inflammatory factors in the mechanisms of IBD-induced patients with IBDs as well as in IBD animal models.
diarrhea. Recent studies show that IBD-associated diarrhea results
from NF-kB-mediated tight junction protein internalization and Financial & competing interests disclosure
increased paracellular permeability. Epithelial NF-kB activation This work was supported by NIH grants AA18729 (Yueming Tang),
plays a role in opening paracellular spaces to promote transmu- AA019405 (Ali Keshavarzian) and AA020216 (Ali Keshavarzian), and in
cosal fluid effux into the intestinal lumen in patients with IBD. part by a mentoring program of the Department of Internal Medicine of Rush
Thus, NF-kB may be a molecular target for effective therapy of University Medical Center (Yueming Tang). The authors have no other rel-
IBD. Recent studies also provide new evidence that miRNAs evant affiliations or financial involvement with any organization or entity
may play a critical role in the regulation of intestinal inflamma- with a financial interest in or financial conflict with the subject matter or
tion and intestinal permeability in the mechanisms underlying materials discussed in the manuscript apart from thosedisclosed.
IBD-induced diarrhea. No writing assistance was utilized in the production of thismanuscript.

Key issues
Diarrhea is common in patients with inflammatory bowel diseases (IBDs) and significantly affects the quality of a patients life.
IBD-induced diarrhea is characterized by a decrease in the fluid and solute absorption, but not net fluid and solute secretion.
Understanding the molecular mechanisms of diarrhea is important in identifying novel molecular targets for the successful treatment of
patients withIBD.
There are many molecular mechanisms responsible for the generation of diarrhea with a wide range of clinical presentations. The
interactions between intestinal microbiota, epithelial barrier and host immune system are complex during normal physiology and
become more complex under inflammatory conditions but it is important that we identify how these factors may intereact and play
important roles in IBD-induced diarrhea.
Recently, the role of the gut microbiota in the mechanisms of IBDs and IBD-associated diarrhea have been extensively investigated.
Modern molecular techniques are rapidly advancing our research progress and knowledge of the intestinal microbiota. The invasion of
bacteria results in disruption of the intestinal barrier and in an increase in the interaction between bacterial components and immune
cells, leading to activation of NF-kB and inflammation as a consequence.
Intestinal barrier dysfunction (leaky gut) contributes to the diarrhea in IBDs by a leak flux mechanism and causes an increase of luminal
antigen uptake, thus, resulting in mucosal inflammation. Tight junctions are important components of the intestinal epithelial barrier.
The epithelial tight junctions are regulated by cytokines and other inflammatory factors in the mechanisms of IBD-induced diarrhea.
Epithelial inflammation in IBDs might induce diarrhea through triggering the release of proinflammatory cytokines from immune cells in
patients with IBD. These proinflammatory (TNF-a and interferons) cytokines induce abnormal ion transport and disruption of tight
junctions that contribute to the inflammation-induced gut leakiness and the mechanisms of IBD-associated diarrhea.
Recent studies indicate that microRNA regulate intestinal permeability and may play an important role in the mechanisms underlying
IBD-induced diarrhea.

622 Expert Rev. Gastroenterol. Hepatol. 5(5), (2011)


New molecular insights into inflammatory bowel disease-induced diarrhea Review

References 13 Rask-Madsen J, Hammersgaard EA, 24 Ivanov AI, Nusrat A, Parkos CA.


Papers of special note have been highlighted as: Knudsen E. Rectal electrolyte transport Endocytosis of the apical junctional
of interest and mucosal permeability in ulcerative complex: mechanisms and possible roles
1 Gismera CS, Aladren BS. Inflammatory colitis and Crohns disease. J. Lab. Clin. in regulation of epithelial barriers.
bowel diseases: a disease(s) of modern Med. 81(3), 342353 (1973). Bioessays 27(4), 356365 (2005).
times? Is incidence still increasing? World J. 14 Rask-Madsen J, Jensen PB. Electrolyte 25 Laukoetter MG, Bruewer M, Nusrat A.
Gastroenterol. 14(36), 54915498 (2008). transport capacity and electrical potentials Regulation of the intestinal epithelial
2 Shah SB, Hanauer SB. Treatment of of the normal and the inflamed human barrier by the apical junctional complex.
diarrhea in patients with inflammatory rectum in vivo. Scand. J. Gastroenterol. Curr. Opin. Gastroenterol. 22(2), 8589
bowel disease: concepts and cautions. Rev. 8(2), 169175 (1973). (2006).
Gastroenterol. Disord. 7(Suppl.3), S3S10 15 Archampong EQ, Harris J, Clark CG. 26 Madara JL. Warner-Lambert/Parke-Davis
(2007). Theabsorption and secretion of water and Award lecture. Pathobiology of the
3 Payne CM, Fass R, Bernstein H etal. electrolytes across the healthy and the intestinal epithelial barrier. Am. J. Pathol.
Pathogenesis of diarrhea in the adult: diseased human colonic mucosa measured 137(6), 12731281 (1990).
diagnostic challenges and life-threatening in vitro. Gut 13(11), 880886 (1972). 27 Backhed F, Ley RE, Sonnenburg JL,
conditions. Eur. J. Gastroenterol. Hepatol. 16 Hawker PC, McKay JS, Turnberg LA. Peterson DA, Gordon JI. Host-bacterial
18(10), 10471051 (2006). Electrolyte transport across colonic mucosa mutualism in the human intestine. Science
4 Seidler U, Lenzen H, Cinar A, Tessema T, from patients with inflammatory bowel 307(5717), 19151920 (2005).
Bleich A, Riederer B. Molecular disease. Gastroenterology 79(3), 508511 28 Rakoff-Nahoum S, Paglino J,
mechanisms of disturbed electrolyte (1980). Eslami-Varzaneh F, Edberg S,
transport in intestinal inflammation. Ann. 17 Amasheh S, Barmeyer C, Koch CS etal. Medzhitov R. Recognition of commensal
NY Acad. Sci. 1072, 262275 (2006). Cytokine-dependent transcriptional microflora by Toll-like receptors is
5 Xavier RJ, Podolsky DK. Unravelling the down-regulation of epithelial sodium required for intestinal homeostasis. Cell
pathogenesis of inflammatory bowel disease. channel in ulcerative colitis. 118(2), 229241 (2004).
Nature 448(7152), 427434 (2007). Gastroenterology 126(7), 17111720 29 Andoh A, Benno Y, Kanauchi O,
(2004). FujiyamaY. Recent advances in molecular
6 Keighley MR, Stockbrugger RW.
Inflammatory bowel disease. Aliment. 18 Yang H, Jiang W, Furth EE etal. Intestinal approaches to gut microbiota in
Pharmacol. Ther. 18(Suppl. 3), 6670 inflammation reduces expression of DRA, inflammatory bowel disease. Curr. Pharm.
(2003). atransporter responsible for congenital Des. 15(18), 20662073 (2009).
chloride diarrhea. Am. J. Physiol. 275(6 30 Qin J, Li R, Raes J etal. A human gut
7 Binder HJ. Mechanisms of diarrhea in Pt1), G1445G1453 (1998).
inflammatory bowel diseases. Ann. NY microbial gene catalogue established by
Acad. Sci. 1165, 285293 (2009). 19 Friswell M, Campbell B, Rhodes J. The metagenomic sequencing. Nature
role of bacteria in the pathogenesis of 464(7285), 5965 (2010).
Concise summary of the pathophysiology inflammatory bowel disease. Gut Liver 31 Salzman NH, de Jong H, Paterson Y,
of diarrhea in both ulcerative colitis and 4(3), 295306 (2010). Harmsen HJ, Welling GW, Bos NA.
Crohns disease.
Summary of the role of the mucosal and Analysis of 16S libraries of mouse
8 Field M. Intestinal ion transport and the fecal microbiota in the pathogenesis of gastrointestinal microflora reveals a large
pathophysiology of diarrhea. J. Clin. Invest. inflammatory bowel disease. new group of mouse intestinal bacteria.
111(7), 931943 (2003). Microbiology 148(Pt 11), 36513660
20 Nell S, Suerbaum S, Josenhans C. The
9 Ivanov AI, Parkos CA, Nusrat A. (2002).
impact of the microbiota on the
Cytoskeletal regulation of epithelial barrier pathogenesis of IBD: lessons from mouse 32 Frank DN, St Amand AL, Feldman RA,
function during inflammation. Am. J. infection models. Nat. Rev. Microbiol. 8(8), Boedeker EC, Harpaz N, Pace NR.
Pathol. 177(2), 512524 (2010). 564577 (2010). Molecular-phylogenetic characterization
10 Schulzke JD, Ploeger S, Amasheh M etal. of microbial community imbalances in
Summarizes the impact of the microbiota
Epithelial tight junctions in intestinal human inflammatory bowel diseases.
on the pathogenesis in mouse
inflammation. Ann. NY Acad. Sci. 1165, Proc. Natl Acad. Sci. USA 104(34),
infectionmodels. 1378013785 (2007).
294300 (2009).
21 Hollander D. The intestinal permeability 33 Chapman-Kiddell CA, Davies PS,
Describes the different mechanisms
barrier. A hypothesis as to its regulation GillenL, Radford-Smith GL. Role of diet
causing disruption of epithelial tight
and involvement in Crohns disease. Scand. in the development of inflammatory
junctions in intestinal inflammation. J. Gastroenterol. 27(9), 721726 (1992). bowel disease. Inflamm. Bowel Dis. 16(1),
11 Mankertz J, Schulzke JD. Altered Clayburgh DR, Shen L, Turner JR.
22 137151(2010).
permeability in inflammatory bowel Aporous defense: the leaky epithelial
disease: pathophysiology and clinical 34 Mutlu E, Keshavarzian A, Engen P,
barrier in intestinal disease. Lab. Invest. Forsyth CB, Sikaroodi M, Gillevet P.
implications. Curr. Opin. Gastroenterol. 84(3), 282291 (2004).
23(4), 379383 (2007). Intestinal dysbiosis: a possible mechanism
23 Turner JR. Molecular basis of epithelial of alcohol-induced endotoxemia and
12 Harris J, Shields R. Absorption and barrier regulation: from basic mechanisms alcoholic steatohepatitis in rats. Alcohol
secretion of water and electrolytes by the to clinical application. Am. J. Pathol. Clin. Exp. Res. 33(10), 18361846
intact human colon in diffuse untreated 169(6), 19011909 (2006). (2009).
proctocolitis. Gut 11(1), 2733 (1970).

www.expert-reviews.com 623
Review Tang, Forsyth & Keshavarzian

35 Gophna U, Sommerfeld K, Gophna S, First study using pyrosequencing in twins ileal mucosa of a patient with Crohns
Doolittle WF, Veldhuyzen van Zanten SJ. demonstrated that bacterial populations disease. Infect. Immun. 67(9), 44994509
Differences between tissue-associated differ in abundance among individuals (1999).
intestinal microfloras of patients with with different phenotypes of 56 Lodes MJ, Cong Y, Elson CO etal.
Crohns disease and ulcerative colitis. inflammatory bowel disease. Bacterial flagellin is a dominant antigen in
J.Clin. Microbiol. 44(11), 41364141 Crohn disease. J. Clin. Invest. 113(9),
46 Uronis JM, Arthur JC, Keku T etal. Gut
(2006). 12961306 (2004).
microbial diversity is reduced by the
36 Bibiloni R, Mangold M, Madsen KL, probiotic VSL#3 and correlates with 57 Miller H, Zhang J, Kuolee R, Patel GB,
Fedorak RN, Tannock GW. The decreased TNBS-induced colitis. Inflamm. Chen W. Intestinal M cells: the fallible
bacteriology of biopsies differs between Bowel Dis. 17(1), 289297 (2010). sentinels? World J. Gastroenterol. 13(10),
newly diagnosed, untreated, Crohns disease 14771486 (2007).
47 Rose DJ, Venema K, Keshavarzian A,
and ulcerative colitis patients. J.Med.
Hamaker BR. Starch-entrapped 58 Garside P, Millington O, Smith KM. The
Microbiol. 55(Pt 8), 11411149 (2006).
microspheres show a beneficial anatomy of mucosal immune responses.
37 Kassinen A, Krogius-Kurikka L, fermentation profile and decrease in Ann. NY Acad. Sci. 1029, 915 (2004).
Makivuokko H etal. The fecal microbiota potentially harmful bacteria during in vitro 59 Lamb-Rosteski JM, Kalischuk LD,
of irritable bowel syndrome patients differs fermentation in faecal microbiota obtained InglisGD, Buret AG. Epidermal growth
significantly from that of healthy subjects. from patients with inflammatory bowel factor inhibits Campylobacter jejuni-
Gastroenterology 133(1), 2433 (2007). disease. Br. J. Nutr. 103(10), 15141524 induced claudin-4 disruption, loss of
38 Othman M, Aguero R, Lin HC. Alterations (2010). epithelial barrier function, and Escherichia
in intestinal microbial flora and human 48 Komiyama Y, Andoh A, Fujiwara D etal. coli translocation. Infect. Immun. 76(8),
disease. Curr. Opin. Gastroenterol. 24(1), New prebiotics from rice bran ameliorate 33903398 (2008).
1116 (2008). inflammation in murine colitis models 60 Rhee SH, Im E, Riegler M, Kokkotou E,
39 Hoffmann C, Hill DA, Minkah N etal. through the modulation of intestinal OBrien M, Pothoulakis C.
Community-wide response of the gut homeostasis and the mucosal immune Pathophysiological role of Toll-like receptor
microbiota to enteropathogenic Citrobacter system. Scand. J. Gastroenterol. 46(1), 5 engagement by bacterial flagellin in
rodentium infection revealed by deep 4052 (2010). colonic inflammation. Proc. Natl Acad. Sci.
sequencing. Infect. Immun. 77(10), 49 Petersen AM, Nielsen EM, Litrup E, USA 102(38), 1361013615 (2005).
46684678 (2009). Brynskov J, Mirsepasi H, Krogfelt KA. 61 Martin HM, Campbell BJ, Hart CA etal.
40 Lupp C, Robertson ML, Wickham ME Aphylogenetic group of Escherichia coli Enhanced Escherichia coli adherence and
etal. Host-mediated inflammation disrupts associated with active left-sided invasion in Crohns disease and colon
the intestinal microbiota and promotes the inflammatory bowel disease. BMC cancer. Gastroenterology 127(1), 8093
overgrowth of Enterobacteriaceae. Cell Host Microbiol. 9, 171 (2009). (2004).
Microbe 2(2), 119129 (2007). 50 Packey CD, Sartor RB. Commensal 62 Geier MS, Butler RN, Howarth GS.
41 Stecher B, Robbiani R, Walker AW etal. bacteria, traditional and opportunistic Inflammatory bowel disease: current
Salmonella enterica serovar typhimurium pathogens, dysbiosis and bacterial killing in insights into pathogenesis and new
exploits inflammation to compete with the inflammatory bowel diseases. Curr. Opin. therapeutic options; probiotics, prebiotics
intestinal microbiota. PLoS Biol. 5(10), Infect. Dis. 22(3), 292301 (2009). and synbiotics. Int. J. Food Microbiol.
21772189 (2007). 51 Chow J, Mazmanian SK. A pathobiont of 115(1), 111 (2007).
42 Heimesaat MM, Fischer A, Siegmund B the microbiota balances host colonization 63 van Vliet MJ, Harmsen HJ, de Bont ES,
etal. Shift towards pro-inflammatory and intestinal inflammation. Cell Host Tissing WJ. The role of intestinal
intestinal bacteria aggravates acute murine Microbe 7(4), 265276 (2010). microbiota in the development and severity
colitis via Toll-like receptors 2 and 4. PLoS 52 Sokol H, Seksik P, Furet JP etal. Low of chemotherapy-induced mucositis. PLoS
One 2(7), e662 (2007). counts of Faecalibacterium prausnitzii in Pathog. 6(5), e1000879 (2010).
43 Garrett WS, Gallini CA, Yatsunenko T colitis microbiota. Inflamm. Bowel Dis. 64 Hooper LV, Gordon JI. Commensal
etal. Enterobacteriaceae act in concert with 15(8), 11831189 (2009). host-bacterial relationships in the gut.
the gut microbiota to induce spontaneous 53 Sokol H, Pigneur B, Watterlot L etal. Science 292(5519), 11151118 (2001).
and maternally transmitted colitis. Cell Host Faecalibacterium prausnitzii is an anti- 65 Blumberg RS, Li L, Nusrat A, Parkos CA,
Microbe 8(3), 292300 (2010). inflammatory commensal bacterium Rubin DC, Carrington JL. Recent insights
44 Walk ST, Blum AM, Ewing SA, identified by gut microbiota analysis of into the integration of the intestinal
WeinstockJV, Young VB. Alteration of the Crohn disease patients. Proc. Natl Acad. epithelium within the mucosal
murine gut microbiota during infection Sci. USA 105(43), 1673116736 (2008). environment in health and disease. Mucosal
with the parasitic helminth 54 Darfeuille-Michaud A, Neut C, Barnich N Immunol. 1(5), 330334 (2008).
Heligmosomoides polygyrus. Inflamm. etal. Presence of adherent Escherichia coli 66 Turner JR. Intestinal mucosal barrier
Bowel Dis. 16(11), 18411849 (2010). strains in ileal mucosa of patients with function in health and disease. Nat. Rev.
45 Willing BP, Dicksved J, Halfvarson J etal. Crohns disease. Gastroenterology 115(6), Immunol. 9(11), 799809 (2009).
A pyrosequencing study in twins shows that 14051413 (1998).
67 Murphy MS, Eastham EJ, Nelson R,
gastrointestinal microbial profiles vary with 55 Boudeau J, Glasser AL, Masseret E, Joly B, Pearson AD, Laker MF. Intestinal
inflammatory bowel disease phenotypes. Darfeuille-Michaud A. Invasive ability of permeability in Crohns disease. Arch. Dis.
Gastroenterology 139(6), 18441854 an Escherichia coli strain isolated from the Child 64(3), 321325 (1989).
(2010).

624 Expert Rev. Gastroenterol. Hepatol. 5(5), (2011)


New molecular insights into inflammatory bowel disease-induced diarrhea Review

68 Wyatt J, Vogelsang H, Hubl W, 76 Mauri DN, Ebner R, Montgomery RI etal. 85 Wu F, Zhang S, Dassopoulos T etal.
WaldhoerT, Lochs H. Intestinal LIGHT, a new member of the TNF Identification of microRNAs associated
permeability and the prediction of relapse superfamily, and lymphotoxin a are ligands with ileal and colonic Crohns disease.
in Crohns disease. Lancet 341(8858), for herpesvirus entry mediator. Immunity Inflamm. Bowel Dis. 16(10), 17291738
14371439 (1993). 8(1), 2130 (1998). (2010).
69 Zeissig S, Burgel N, Gunzel D etal. 77 Tang Y, Clayburgh DR, Mittal N etal. 86 Wu F, Zikusoka M, Trindade A etal.
Changes in expression and distribution of Epithelial NF-kB enhances transmucosal MicroRNAs are differentially expressed in
claudin 2, 5 and 8 lead to discontinuous fluid movement by altering tight junction ulcerative colitis and alter expression of
tight junctions and barrier dysfunction in protein composition after T cell activation. macrophage inflammatory peptide-2 a.
active Crohns disease. Gut 56(1), 6172 Am. J. Pathol. 176(1), 158167 (2010). Gastroenterology 135(5), 16241635.e1624
(2007). Demonstrated the role of epithelial (2008).
70 Rosenstiel P, Fantini M, Brautigam K etal. NF-kB in the mechanisms of T-cell 87 Tang Y, Banan A, Forsyth CB etal. Effect
TNF-a and IFN-g regulate the expression activation-induced diarrhea and T-cell of alcohol on miR-212 expression in
of the NOD2 (CARD15) gene in human activation-induced disruption of intestinal intestinal epithelial cells and its potential
intestinal epithelial cells. Gastroenterology tight junctions. role in alcoholic liver disease. Alcohol Clin.
124(4), 10011009 (2003). Exp. Res. 32(2), 355364 (2008).
78 Amin MR, Orenuga T, Tyagi S,
71 Buhner S, Buning C, Genschel J etal. DudejaPK, Ramaswamy K, Malakooti J. 88 Blow MJ, Grocock RJ, van Dongen S etal.
Genetic basis for increased intestinal Tumor necrosis factor-a represses the RNA editing of human microRNAs.
permeability in families with Crohns expression of NHE2 through NF-kB Genome Biol. 7(4), R27 (2006).
disease: role of CARD15 3020insC activation in intestinal epithelial cell 89 Griffiths-Jones S, Grocock RJ, van
mutation? Gut 55(3), 342347 (2006). model, C2BBe1. Inflamm. Bowel Dis. DongenS, Bateman A, Enright AJ.
72 DInca R, Annese V, di Leo V etal. 17(3), 720731 (2010). miRBase: microRNA sequences, targets
Increased intestinal permeability and 79 Carthew RW. Gene regulation by and gene nomenclature. Nucleic Acids Res.
NOD2 variants in familial and sporadic microRNAs. Curr. Opin. Genet. Dev. 34(Database issue), D140D144 (2006).
Crohns disease. Aliment. Pharmacol. Ther. 16(2), 203208 (2006). 90 Kapeller J, Houghton LA, Monnikes H
23(10), 14551461 (2006). etal. First evidence for an association of a
80 Ambros V. The functions of animal
73 Musch MW, Clarke LL, Mamah D etal. microRNAs. Nature 431(7006), 350355 functional variant in the microRNA-510
Tcell activation causes diarrhea by (2004). target site of the serotonin receptor-type 3E
increasing intestinal permeability and gene with diarrhea predominant irritable
81 Krutzfeldt J, Stoffel M. MicroRNAs: a new
inhibiting epithelial Na+/K+ -ATPase. bowel syndrome. Hum. Mol. Genet. 17(19),
class of regulatory genes affecting
J.Clin. Invest. 110(11), 17391747 (2002). 29672977 (2008).
metabolism. Cell Metab. 4(1), 912 (2006).
74 Clayburgh DR, Barrett TA, Tang Y etal. 91 Cho JH, Brant SR. Recent insights into the
82 Krutzfeldt J, Poy MN, Stoffel M. Strategies
Epithelial myosin light chain kinase- genetics of inflammatory bowel disease.
to determine the biological function of
dependent barrier dysfunction mediates Gastroenterology 140(6), 17041712
microRNAs. Nat. Genet. 38(Suppl.),
Tcell activation-induced diarrhea invivo. (2010).
S14S19 (2006).
J.Clin. Invest. 115(10), 27022715 (2005). 92 Hering NA, Schulzke JD. Therapeutic
83 Gauthier BR, Wollheim CB. MicroRNAs:
75 Clayburgh DR, Musch MW, Leitges M, options to modulate barrier defects in
ribo-regulators of glucose homeostasis.
FuYX, Turner JR. Coordinated epithelial inflammatory bowel disease. Dig. Dis.
Nat. Med. 12(1), 3638 (2006).
NHE3 inhibition and barrier dysfunction 27(4), 450454 (2009).
are required for TNF-mediated diarrhea in 84 Fasseu M, Treton X, Guichard C etal.
vivo. J. Clin. Invest. 116(10), 26822694 Identification of restricted subsets of
(2006). mature microRNA abnormally expressed in
inactive colonic mucosa of patients with
inflammatory bowel disease. PLoS One
5(10), e13160 (2010).

www.expert-reviews.com 625

Potrebbero piacerti anche