Anda di halaman 1dari 7

REVIEW

CURRENT
OPINION Inflammatory bowel disease and irritable bowel
syndrome: similarities and differences
Giovanni Barbara, Cesare Cremon, and Vincenzo Stanghellini

Purpose of review
Inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) are classically viewed as dichotomous
conditions. The former is perceived as a typical organic disease, and the latter is regarded as a disorder of
gut function driven by mood. Recent research identified some shared contributing factors, which will be
discussed here.
Recent findings
Mounting evidence shows the importance in both IBD and IBS of genetic, microbiological, epithelial, and
immunological factors. In some instances, these factors overlap in the two conditions as shown by:
involvement of brain–gut axis dysfunction in IBD, implication of TNFSF gene in Crohn’s disease and IBS,
evidence of abnormal microbiota and its impact on host functions, identification of low-grade inflammation
in subsets of IBS patients, and development of IBS symptoms in patients with IBD in remission.
Summary
IBD and IBS remain separate conditions although there are some overlapping mechanisms. Both research
and clinical management would benefit from considering a functional approach for certain manifestations
of IBD and accepting an organic view in subsets of IBS patients.
Keywords
epithelial barrier, genes, inflammatory bowel disease, irritable bowel syndrome, microbiota

INTRODUCTION substantiates points of contact and fuels the mount-


Inflammatory bowel disease (IBD) and irritable ing evidence supporting a role for immunological
bowel syndrome (IBS) are frequent gastrointestinal dysfunction in the pathophysiology of IBS.
conditions associated with a marked socioeconomic
burden and impairment of patients’ quality of life
& & EPIDEMIOLOGICAL AND CLINICAL
(QoL) [1 ,2 ]. IBD and IBS are clear paradigms of the
FEATURES
dichotomy between organic and nonorganic gastro-
intestinal diseases. IBD is classically viewed as a IBD encompasses two main clinical entities, namely
peripheral condition characterized by intestinal ulcerative colitis and Crohn’s disease, both present-
inflammation, responding to therapies that primar- ing commonly with abdominal pain, diarrhea, and
ily target the immune system. Conversely, IBS has bloody stools. IBS is characterized by abdominal
been considered for a long time a centrally driven pain/discomfort and disordered defecation and fur-
disorder diagnosed after exclusion of organic causes ther subtyped according to bowel habit character-
and manageable with lifestyle modifications and istics. The main subtypes are IBS with constipation
symptomatic treatment. However, several recent (IBS-C), IBS with diarrhea (IBS-D), and IBS with
findings indicate that the boundaries between IBD
and IBS are becoming blurred. There is epidemio-
Department of Medical and Surgical Sciences, University of Bologna,
logic, genetic, immune, and microbiological over- Bologna, Italy
lap. QoL is often similarly impaired in the two Correspondence to Giovanni Barbara, MD, Department of Medical and
conditions, and it is not uncommon to verify that Surgical Sciences, Section of Internal Medicine and Gastroenterology,
symptoms in patients with IBD surpass the severity University of Bologna, St. Orsola–Malpighi Hospital, Building No. 5, Via
of endoscopic lesions and objective markers of Massarenti, 9, I-40138, Italy. E-mail: giovanni.barbara@unibo.it
inflammation. The frequent experience of IBS-like Curr Opin Gastroenterol 2014, 30:352–358
symptoms in patients with IBD in remission further DOI:10.1097/MOG.0000000000000070

www.co-gastroenterology.com Volume 30  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Inflammatory bowel disease and irritable bowel syndrome Barbara et al.

digestive disease with the greatest preponderance


KEY POINTS among females [11]. Mortality is almost nonexistent
 IBD and IBS share some common features, including for IBS, but is also relatively rare for IBD, making
the involvement of genetic factors, disorders of immune gastroesophageal reflux disease a more commonly
system, and altered microbiota. listed cause of death than IBD [11].
 Some genetic polymorphisms (TNFSF15), which
regulate host–microbiota interactions, are more THE BRAIN–GUT AXIS
frequently observed both in Crohn’s disease and IBS.
The brain–gut axis involves interactions among
 While low-grade intestinal inflammation has been the central and the autonomic nervous system,
widely described in IBS, the magnitude and nature of the hypothalamic–pituitary–adrenal axis, and the
these responses is substantially different compared to
gastrointestinal tract, including the luminal
that of IBD.
microbiota, the epithelial barrier, and the intestinal
 IBS-like symptoms, which are frequently reported by immune system. Brain–gut axis interactions have
patients with IBD, originate from a combination of been typically studied in the context of FGIDs
psychological factors, low-grade inflammation, and and IBS in particular; however, several recent data
enteric neuroplastic changes.
suggest their relevance also in the pathophysiology
&&
of IBD [12 ]. An association between psychological
factors and IBD flare-ups has been widely reported
mixed bowel habit features (IBS-M), although this &&
[12 ]. Depression has been shown to predispose to
classification may be artificial as there is wide over- higher inflammatory responses to a stressor [13 ],
&

&&
lap between these different entities [3 ]. Abdominal and one third of patients report improvement of IBD
pain, the cardinal symptom of IBS, is the first with antidepressant therapy [14]. Compared to con-
reported complaint that induces patients to consult trols, patients taking antidepressants had reduced
a doctor and the key symptom impacting on QoL. relapse rates, steroid use, and endoscopic procedures
Pain is also very frequently reported by IBD patients in the year after their introduction [15]. However,
with active disease [4], but it is often neglected when confirmation of these data in prospective, con-
the therapeutic focus is on mucosal healing. How- trolled studies is lacking. FGIDs have been widely
ever, pain becomes central in patients who continue linked to psychological factors, including anxiety,
to complain of abdominal IBS-like symptoms in depression, fobia, and somatization. Koloski et al.
spite of resolution of inflammation [4]. Both IBD &
[16 ] performed a 12-year longitudinal, prospective,
and IBS are associated with a significant impact on population-based study to determine the direction-
physical as well as mental components of health- ality of brain–gut interactions in IBS and functional
& & &&
related QoL [1 ,2 ,5 ,6]. dyspepsia. As expected, higher levels of anxiety and
The prevalence of IBD has been estimated to depression at baseline were predictive of IBS at
range between 1.5 and 294 cases per 100 000 people follow-up. However, FGID without mood disorders
[7]. Frequency figures are much higher for IBS at baseline predisposed individuals to anxiety and
with around 10–20% of the population affected depression at follow-up, suggesting that peripheral
in Europe and the United States [8]. There have been changes may be the trigger of mood disorders at least
similar trends of increasing prevalence of both IBD in a subgroup of IBS patients. Interestingly, disease
&&
[5 ,9] and IBS [10] in the eastern world. It remains activity status and severity of inflammation have
unknown whether this is a primary epidemiological also been linked to the severity of psychological
shift of the disease or rather reflects higher consult- symptoms associated with IBD (e.g. anxiety and
ing rates, improvements in diagnosis, or the result of &&
depression) [12 ]. Regional cortical gray matter
increasing availability of therapeutic opportunities. density changes have been detected in diverse
Although most patients with IBD seek medical inflammatory disorders, including IBD and IBS.
attention for their symptoms, there is an opposite The importance of female sex in brain neuroplastic
trend for IBS with the majority of patients non- changes has been described, particularly in IBS
consulting a physician. While IBD impacts on both [17,18]. Recent data suggest that these changes
outpatient and inpatient care, IBS is mainly seen in could be the consequence of peripheral gut inflam-
&&
the outpatient setting [5 ]. Chronic constipation mation in ulcerative colitis, whereas they may
and IBS together account for 9.4 million of the represent a primary pathogenetic factor in IBS
11.6 million outpatient diagnoses for functional [17]. However, convincing cause–effect relationship
gastrointestinal disorders (FGIDs) in the United in these disorders is still lacking. One major con-
States [11]. In the female population, the rate visit tributor of brain responses and symptom perception
for IBS is 4.4 times that of males, making IBS the in IBS is visceral hypersensitivity, the increased

0267-1379 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-gastroenterology.com 353

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Inflammatory bowel disease

perception of stimuli arising from the gastrointesti- activation [25]. Although these data suggest that IBS,
nal tract [19]. The role of visceral hypersensitivity similarly to IBD, is linked to an abnormal mucosal
and its correlation with the degree of active inflam- immune response to luminal microbiota in geneti-
mation is controversial [20]. On the other hand, cally predisposed individuals, more work has to be
visceral hypersensitivity may become more evident done to confirm or discard this interesting hypo-
in patients with IBD in clinical remission who show thesis. Nonetheless, these data support the concept
persistent symptoms including abdominal pain of a central role for organic changes in the patho-
&&
[21 ]. genesis of at least a subgroup of patients with IBS.

GENETIC FACTORS MICROBIOTA


Gene hunting started over a decade ago in IBD It is well established that IBD patients show reduced
research and led to identification of several genes diversity and different compositions of intestinal
contributing to IBD pathogenesis. These findings microbiota, including reduction of bacteria
substantiate the known familiar aggregation in involved in butyrate and propionate metabolism
these patients. A recent meta-analysis of genome- (e.g. Ruminococcus bromii et rel., Eubacterium rectale
wide association studies identified 163 susceptibility et rel., and Roseburia sp.) as well as mucus degra-
& &
loci in IBD. Many of these loci (e.g. NOD2, IRGM, dation (i.e. Akkermansia sp.) [26 –28 ]. Attention
ATG16L1, and IL23R) are involved in host–micro- has been also directed to the potential role of
biota interactions [22]. The TNFSF15 gene encodes specific bacterial strains including the reduction
for TL1A, a member of the TNF ligand superfamily in tissue concentrations of the anti-inflammatory
that is involved in defense against pathogens and microbiota commensal Faecalibacterium prausnitzii
&
modulates the interactions between host and micro- [26 ]. The pathogenetic role of F. prausnitzii in IBD
biota in the gut. The TNFSF15 gene was first ident- is supported by data showing that low tissue con-
ified as a risk factor for Crohn’s disease and later centration of this bacterium was associated with an
reported to also contribute to the risk of ulcerative increased risk of postoperative recurrence of Crohn’s
&
colitis [22]. On the basis of the observation that a disease [26 ]. However, a recent study failed to find
subgroup of patients with IBS shows evidence of decreased tissue levels of F. prausnitzii at least in a
low-grade immune activation, the involvement of pediatric group of Crohn’s disease patients [29]. The
TNFSF15, previously linked to Crohn’s disease, has evidence linking microbiota to IBS is in its infancy,
&&
been recently investigated. A large IBS case–control but it is already providing exciting results [30 ]. The
study in 1992 individuals from two independent clearest evidence of the participation of bacteria in
cohorts from Sweden and the United States ident- the pathogenesis of IBS is provided by the fact that
ified that the Crohn’s disease-risk variant G from the about 10% of individuals experiencing an acute
rs4263839 single nucleotide polymorphism (SNP) in episode of gastroenteritis (e.g. Shigella, Salmonella,
the TNFSF15 gene was more frequently observed and Campylobacter) develop persistent symptoms
in IBS (particularly IBS-C) than controls [23]. These fulfilling the criteria for IBS [31]. A 16-year, prospec-
data have been recently confirmed in a smaller study tive, controlled, culture-proven, follow-up study
from the United Kingdom demonstrating also a explored the association between a single episode
higher tissue expression of TNFSF15 along with of Salmonella gastroenteritis and new-onset FGIDs.
increased mRNA expression of several cytokines In this long-term outcome study, it was shown that
and chemokines [24]. As the disease risk alleles Salmonella gastroenteritis increased the risk for IBS,
may be the same for Crohn’s disease and IBS, it but only if the infection occurred during childhood
&
has been hypothesized that common mechanisms [32 ]. Interestingly, acute infectious gastroenteritis
could be involved in the predisposition to both is also a trigger for the development of IBD [33].
conditions. It has also been hypothesized that IBS Porter et al. [33] demonstrated that acute infectious
may represent an immunologically incomplete gastroenteritis was an independently associated risk
expression variant of Crohn’s disease [18]. Along factor for IBD [odds ratio (OR): 1.40] with ORs for
the line of an involvement of genes in the patho- Crohn’s disease slightly higher than those for ulcer-
genesis of IBS, it was found that development of IBS ative colitis (ORs: 1.54 and 1.36, respectively).
following a waterborne outbreak of infectious gas- The first compelling evidence of altered fecal
troenteritis was associated with SNPs in toll-like microbiota composition in IBS comes from a Finnish
receptor 9 (TLR9), the tight junction protein cad- study showing increased ratio of Firmicutes:Bacter-
herin (CDH1), and the pro-inflammatory cytokine oidetes and lower levels of Bifidobacteria [34]. A
interleukin 6, which are involved in host–micro- recent study investigated the correlation between
biota interactions, epithelial barrier, and immune microbiota profiles and psychological factors or

354 www.co-gastroenterology.com Volume 30  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Inflammatory bowel disease and irritable bowel syndrome Barbara et al.

&&
bowel physiology [35 ]. It was found that micro- correlated with the severity of abdominal pain
biota abnormalities were associated with peripheral [41,42]. These functional observations are accom-
changes such as disturbances in gut transit, whereas panied by elegant morphological studies showing
individuals with no modifications of the microbiota that the jejunal mucosa of patients with IBS-D
&&
correlated with anxiety and depression [35 ]. More showed alterations in expression and phosphoryl-
recently, a bacterial profile of 27 genus-like groups ation of tight junctions and ultrastructural abnor-
significantly separated both postinfectious IBS and malities such as perijunctional cytoskeleton con-
IBS-D from controls. The study also provided evi- densation and enlarged apical intercellular distance
&
dence of a correlation between changes in micro- between cells [43 ]. Interestingly, these alterations
biota and expression of several host gene pathways were correlated both with mast cell activation and
involved in impaired epithelial barrier function clinical symptoms including diarrhea and pain
&& &
[36 ]. Attention is being directed to the potential severity [43 ]. The cause(s) of the described morpho-
functional consequences of altered microbiota taxa. logical and functional changes remain unknown,
Diet is a major driving force in the control of micro- although previous episodes of infectious gastroen-
biota composition and function. On the other hand, teritis, dysbiosis, allergy, genetic, and epigenetic
microbiota changes may lead to the abnormal factors may be involved. Low-grade inflammation
fermentation of indigestible carbohydrates, with described in subgroups of patients with IBS may
consequent production of gas and metabolites have a role in the maintenance of increase of
potentially involved in symptom generation. The intestinal permeability.
effect of a challenge with a diet rich in fibers on
microbiota composition and abdominal symptoms
has been recently investigated. When patients com- INFLAMMATION
plaining of flatulence were challenged with the Tissue inflammation is central to IBD pathogenesis
fiber-rich diet, microbiota showed higher instability and has been the starting point of mainstream
and reduction of microbial diversity compared to research in this condition. Conversely, the concept
healthy controls. In addition, when challenged, that IBS pathogenesis involves peripheral organic
both groups recorded more abdominal symptoms, changes and minimal inflammation is relatively
number of gas evacuations, and more gas pro- new and represents a new concept resulting from
&
duction [37 ]. Previous data demonstrated increased recent research. Several studies showed increased
luminal and fecal protease activity in IBS. Proteases numbers of immunocytes (e.g. mast cells and T cells)
have been linked to the activation of receptors that in both adult and pediatric IBS [44]. Low-grade
are involved in pain transmission and bowel dys- inflammation has been mainly associated with post-
function. Specific intestinal bacterial groups (Lacto- infectious IBS; however, several studies demonstrate
bacillales, Lachnospiraceae, and Streptococcaceae) are similar findings in IBS-D and IBS-C [45]. The fact
linked to fecal protease activity. These data support that up to one third of patients with IBD in remis-
the hypothesis that bacteria could be important sion and around half of patients with microscopic
contributors to higher fecal protease activity and colitis experience IBS-like symptoms further sup-
support a link between bacteria and abdominal pain ports the involvement of low-grade inflammation
[38]. in IBS [44]. Low-grade inflammation is coupled
with increased expression of innate immunity to
microbial molecular patterns (e.g. TLR) [46],
EPITHELIAL BARRIER suggesting a contributing role for gut microbiota.
The intestinal barrier represents a strategic site of the Biopsies obtained from the colonic mucosa of IBS
interplay between billions of bacteria, nutrients, and patients released higher amounts of inflammatory
the host. A long-term change in epithelial barrier mediators, including proteases, histamine, and
function has been suggested to play a role in the prostaglandins compared with controls [47–49].
pathogenesis of IBD and predispose to IBD relapses Caution should be used when comparing this
[39]. Increased intestinal permeability has been inflammatory response with that of IBD, as marked
recently suggested to participate in the pathogenesis quantitative and qualitative differences exist. As
of symptoms in IBS [40]. Higher paracellular per- expected, the magnitude of the inflammatory
meability was demonstrated in colonic biopsies response seen in IBS was much lower than that of
in Ussing chamber experiments [41]. In Caco-2 active or quiescent ulcerative colitis, although it was
monolayers, the application of cleared supernatants widely overlapping with that of microscopic colitis
of IBS colonic biopsies in culture induced down- [50]. In IBS, most data converge on the implication
regulation of zonula occludens-1 mRNA expression of mast cells. Activated mast cells in close proximity
and increased paracellular permeability, which of enteric nerves were correlated with the intensity

0267-1379 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-gastroenterology.com 355

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Inflammatory bowel disease

and frequency of abdominal pain [49]. Mucosal suggesting that central factors alone cannot justify
mediators of mast cell origin hyperexcited nocicep- completely the development of IBS-like symptoms
tive visceral sensory pathways in recipient rodents in all patients [4].
&
[47,48,51 ] or enteric nervous system excitability in The prevalence of IBS in the general population is
isolated gut tissues from guinea pigs [52]. Although relatively high (10–20%), hence coexistent IBS could
immune cells are likely the major source of mediators be expected in a similar proportion of IBD patients in
impacting nerve function, a microbial origin of some remission. However, the high prevalence of IBS-like
of these factors has also been suggested [53]. Potential symptoms in IBD in remission cannot be explained
contributing factors of this low-grade inflammation by casual overlap alone. Whether IBS-like symptoms
include, among others, allergic reactions and stress. reflect clinically undetected, low-grade inflam-
Acute psychological stress increased small intestinal mation is a matter of controversy. Fecal calprotectin
permeability in humans and the mast cell stabilizer was significantly higher in Crohn’s disease and ulcer-
disodium chromoglycate blocked this effect. These ative colitis patients with criteria for IBS than in
findings provide new insights into the complex inter- those without IBS-type symptoms [55]. In addition,
play between the central nervous system and the increased intraepithelial lymphocytes and tissue
immune system in humans [54]. tumor necrosis factor alpha expression was associated
with IBS-like symptoms in quiescent IBD, but not
&&
with IBS [56 ]. These findings suggest that occult
EXPERIENCE OF IRRITABLE BOWEL inflammation could drive IBS-like symptoms in
SYNDROME SYMPTOMS IN PATIENTS patients with IBD in remission and imply the possible
WITH INFLAMMATORY BOWEL DISEASE need of further testing (e.g. fecal calprotectin) and
IN REMISSION potentiation of immunosuppressive therapy [55].
Although a high proportion of patients with IBD However, increased calprotectin levels have not been
frequently experiences symptoms, which are indis- confirmed in more recent studies, suggesting a more
&&
tinguishable from IBS, the diagnosis is generally complex pathogenesis [4,21 ]. A barostat study in
confirmed by additional laboratory and imaging patients with ulcerative colitis in remission indicated
studies. The clinical dilemma emerges when that IBS-like symptoms were associated with visceral
patients continue to experience symptoms (e.g. hypersensitivity and mast cell infiltration [57]. In
abdominal pain, bloating and changes in bowel addition, Akbar et al. [58] showed a 3.9-fold to 5-fold
habit) while in remission. Is this the expression of increase of the sensory nerve receptor transient
occult active disease or IBS? Should these patients receptor potential vanilloid type 1 (TRPV1) associ-
receive more aggressive anti-inflammatory/immuno- ated with pain perception in the mucosa of IBD
suppressive therapy? patients with IBS symptoms. TRPV1 expression cor-
A meta-analysis of 1703 individuals showed that related with the severity of abdominal pain, suggest-
the prevalence of IBS-like symptoms in patients with ing that postinflammatory neuroplastic changes
IBD in remission was 35%. Crohn’s disease patients occurring at least in a subgroup of patients with
in remission had a higher risk of having IBS-like IBD may have potential consequences for bowel
symptoms compared with ulcerative colitis in remis- physiology, visceral hypersensitivity, and symptom
sion (OR: 1.74; 95% CI: 1.24 – 2.43) [4]. IBS-like generation.
symptoms in these individuals were associated with
female sex, reduced well-being, and QoL as well as
higher fatigue scores than those without [21 ].
&&
CONCLUSION
Nonetheless, a comparative study reported that Recent research suggests that IBD and IBS share
QoL was more severely affected in ‘true’ IBS com- some common pathogenetic features. The possible
pared with IBS occurring in IBD patients in remis- implication of microbiota, inflammation, and
sion. In a recent longitudinal 3-year follow-up genetic factors has received increasing attention
study, it was observed that symptoms of IBS in in IBS research, although their relevance is far less
ulcerative colitis in remission tended to fluctuate defined in IBS compared with IBD. The overlap
over time being present overall in 19% of patients at between IBD and IBS is particularly evident in
&&
yearly visits [21 ]. A frequent finding is the detec- patients who develop IBS-like symptoms while in
tion of higher levels of mood disorders, including remission. A better understanding of the role of
predominantly anxiety, in patients experiencing pathogenetic factors and mechanisms underlying
IBS-like symptoms compared with symptom-free the development of persistent symptoms in patients
&&
individuals [4,21 ]. Although anxiety was inde- with IBD in remission is now needed for the identi-
pendently associated with IBS symptoms, the fication of more objective disease signatures and
risk seems moderate (OR: 1.11, CI: 1.01–1.21), development of targeted therapies.

356 www.co-gastroenterology.com Volume 30  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Inflammatory bowel disease and irritable bowel syndrome Barbara et al.

11. Everhart JE, Ruhl CE. Burden of digestive diseases in the United States
Acknowledgements part II: lower gastrointestinal diseases. Gastroenterology 2009; 136:741–
G.B. and V.S. are supported in part by the Italian 754.
12. Bonaz BL, Bernstein CN. Brain–gut interactions in inflammatory bowel
Ministry of Instruction, University and Research. && disease. Gastroenterology 2013; 144:36–49.
Extensive review evaluating brain–gut interactions in patients with IBD. In this
review, the authors discuss the role of psychoneurological factors on intestinal
Conflicts of interest inflammation in animal models and human IBD. Implications of these interactions
for the clinical course of IBD and potential therapeutic implications are also
G.B. and V.S. have served as consultants/advisory board provided and discussed.
members for Alfa Wassermann, Almirall, Italchimici, 13. Fagundes CP, Glaser R, Hwang BS, et al. Depressive symptoms enhance
stress-induced inflammatory responses. Brain Behav Immun 2013; 31:172–
Ironwood, and Shire. G.B. and V.S. received research &

176.
grant support from Alfa Wassermann, Italchimici, and Study assessing the relationship among stress, depression, and immune dysregula-
tion in 138 healthy adults. Individuals with more depressive symptoms produced
Shire. G.B. received research grant support from Falk more interleukin-6 after a standardized laboratory speech and mental arithmetic
Pharma, Sofar, and Yakult. V.S. received research grant stressor.
14. Mikocka-Walus AA, Gordon AL, Stewart BJ, Andrews JM. A magic pill? A
support from Almirall, Aptalis, Norgine, and Valeas. qualitative analysis of patients’ views on the role of antidepressant therapy in
G.B. served as consultant/advisory board member for inflammatory bowel disease (IBD). BMC Gastroenterol 2012; 12:93.
15. Goodhand JR, Greig FI, Koodun Y, et al. Do antidepressants influence
Danone, Falk Pharma, Malesci, Nestlè, Noos, Sofar, the disease course in inflammatory bowel disease? A retrospective
Synergy, and Yakult. V.S. served as consultant/advisory case-matched observational study. Inflamm Bowel Dis 2012; 18:1232–
1239.
board member for Angelini, Aptalis, CM&D Pharma, 16. Koloski NA, Jones M, Kalantar J, et al. The brain–gut pathway in functional
Farmaderma, Janssen, Norgine, Takeda, Valeas, and & gastrointestinal disorders is bidirectional: a 12-year prospective population-
based study. Gut 2012; 61:1284–1290.
Zeria. C.C. has been the recipient of a scholarship grant Interesting study showing the directionality of the brain–gut axis in functional
from Sofar. gastrointestinal disorders.
17. Hong JY, Labus JS, Jiang Z, et al. Regional neuroplastic brain changes in
patients with chronic inflammatory and non-inflammatory visceral pain. PLoS
One 2014; 9:e84564.
18. Labus JS, Dinov ID, Jiang Z, et al. Irritable bowel syndrome in female patients is
REFERENCES AND RECOMMENDED associated with alterations in structural brain networks. Pain 2014; 155:137–
READING 149.
Papers of particular interest, published within the annual period of review, have 19. Barbara G, Cremon C, De Giorgio R, et al. Mechanisms underlying visceral
been highlighted as: hypersensitivity in irritable bowel syndrome. Curr Gastroenterol Rep 2011;
& of special interest 13:308–315.
&& of outstanding interest 20. Srinath AI, Walter C, Newara MC, Szigethy EM. Pain management in patients
with inflammatory bowel disease: insights for the clinician. Therap Adv
1. Lackner JM, Gudleski GD, Thakur ER, et al. The impact of physical complaints, Gastroenterol 2012; 5:339–357.
& social environment, and psychological functioning on IBS patients’ health 21. Jonefjall B, Strid H, Ohman L, et al. Characterization of IBS-like symptoms in
perceptions: looking beyond GI symptom severity. Am J Gastroenterol 2014; && patients with ulcerative colitis in clinical remission. Neurogastroenterol Motil
109:224–233. 2013; 25:756–e578.
This study assessed if psychosocial factors can predict self-ratings of health (SRH) Important report characterizing IBS-like symptoms in patients with ulcerative colitis
in a cohort of 234 Rome III-positive IBS patients. A partial correlation was in clinical remission and their correlation with inflammatory markers, psychological
demonstrated between SRH and somatization, depression, fatigue, stress, anxiety, symptoms, and QoL. IBS-like symptoms are common in these patients and are
and medical comorbidities (r ¼ 0.36–0.41, P < 0.05). associated with poor psychological well-being and reduced QoL.
2. McCombie AM, Mulder RT, Gearry RB. How IBD patients cope with IBD: a 22. Jostins L, Ripke S, Weersma RK, et al. Host–microbe interactions have
& systematic review. J Crohns Colitis 2013; 7:89–106. shaped the genetic architecture of inflammatory bowel disease. Nature 2012;
Systematic review on coping strategies of IBD patients. Emotion-focused coping 491:119–124.
was associated with worse psychological outcomes. 23. Zucchelli M, Camilleri M, Andreasson AN, et al. Association of TNFSF15
3. Ford AC, Bercik P, Morgan DG, et al. Characteristics of functional bowel polymorphism with irritable bowel syndrome. Gut 2011; 60:1671–1677.
&& disorder patients: a cross-sectional survey using the Rome III criteria. Aliment 24. Swan C, Duroudier NP, Campbell E, et al. Identifying and testing candidate
Pharmacol Ther 2014; 39:312–321. genetic polymorphisms in the irritable bowel syndrome (IBS): association with
Important study showing that functional gastrointestinal disorders are not clearly TNFSF15 and TNFalpha. Gut 2013; 62:985–994.
distinct entities and frequently overlap. Of 3656 patients with complete dataset for 25. Villani AC, Lemire M, Thabane M, et al. Genetic risk factors for postinfectious
lower gastrointestinal symptoms, 1551 (42.4%) fulfilled criteria for a functional irritable bowel syndrome following a waterborne outbreak of gastroenteritis.
bowel disorder. Overlap occurred in 27.6% of patients with IBS-D and in 18.1% of Gastroenterology 2010; 138:1502–1513.
patients with IBS-C. 26. Major G, Spiller R. Irritable bowel syndrome, inflammatory bowel disease
4. Halpin SJ, Ford AC. Prevalence of symptoms meeting criteria for irritable & and the microbiome. Curr Opin Endocrinol Diabetes Obes 2014; 21:15–
bowel syndrome in inflammatory bowel disease: systematic review and meta- 21.
analysis. Am J Gastroenterol 2012; 107:1474–1482. Timely review describing the potential impact of gut microbiota in IBD and irritable
5. Burisch J, Jess T, Martinato M, Lakatos PL. The burden of inflammatory bowel bowel syndrome and the role of microbiota modulation in these disorders.
&& disease in Europe. J Crohns Colitis 2013; 7:322–337. 27. Cader MZ, Kaser A. Recent advances in inflammatory bowel disease:
Review assessing the burden of IBD in Europe by analyzing epidemiological data, & mucosal immune cells in intestinal inflammation. Gut 2013; 62:1653–
economic aspects (including patients’ disability and work impairment), disease 1664.
course and risk for surgery, hospitalization, cancer, and mortality. Up-to-date review highlighting the complex interplay of distinct intestinal immune
6. Naliboff BD, Kim SE, Bolus R, et al. Gastrointestinal and psychological cell types and discussing how mucosal immune system perturbations can lead to
mediators of health-related quality of life in IBS and IBD: a structural equation inflammation.
modeling analysis. Am J Gastroenterol 2012; 107:451–459. 28. Rajilic-Stojanovic M, Shanahan F, Guarner F, de Vos WM. Phylogenetic analysis
7. Kappelman MD, Rifas-Shiman SL, Kleinman K, et al. The prevalence and & of dysbiosis in ulcerative colitis during remission. Inflamm Bowel Dis 2013;
geographic distribution of Crohn’s disease and ulcerative colitis in the United 19:481–488.
States. Clin Gastroenterol Hepatol 2007; 5:1424–1429. Interesting study assessing fecal microbiota composition in patients with ulcerative
8. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel colitis. Results showed that fecal microbiota composition differs significantly in
syndrome: a meta-analysis. Clin Gastroenterol hepatol 2012; 10:712–721; these patients in comparison with controls, but it is stable in time. This dysbiosis is
e714. characterized by significant reduction of members of the Clostridium cluster IV and
9. Kappelman MD, Moore KR, Allen JK, Cook SF. Recent trends in the pre- significant reduction of bacteria involved in butyrate and propionate metabolism,
valence of Crohn’s disease and ulcerative colitis in a commercially insured US including Ruminococcus bromii et rel., Eubacterium rectale et rel., Roseburia sp.,
population. Dig Dis Sci 2013; 58:519–525. and Akkermansia.
10. Quigley EM, Abdel-Hamid H, Barbara G, et al. A global perspective on irritable 29. Hansen R, Russell RK, Reiff C, et al. Microbiota of de-novo pediatric IBD:
bowel syndrome: a consensus statement of the World Gastroenterology increased Faecalibacterium prausnitzii and reduced bacterial diversity in
Organisation Summit Task Force on irritable bowel syndrome. J Clin Gastro- Crohn’s but not in ulcerative colitis. Am J Gastroenterol 2012; 107:1913–
enterol 2012; 46:356–366. 1922.

0267-1379 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-gastroenterology.com 357

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Inflammatory bowel disease

30. Simren M, Barbara G, Flint HJ, et al. Intestinal microbiota in functional bowel 43. Martinez C, Lobo B, Pigrau M, et al. Diarrhoea-predominant irritable bowel
&& disorders: a Rome foundation report. Gut 2013; 62:159–176. & syndrome: an organic disorder with structural abnormalities in the jejunal
In depth review on the current knowledge on the role of intestinal microbiota in the epithelial barrier. Gut 2013; 62:1160–1168.
pathophysiology of functional gastrointestinal disorders. The review explores also Careful study assessing clinical symptoms and jejunal biopsies in 45 patients with
treatment implications including the use of prebiotics, probiotics, symbiotics, and IBS-D and 30 controls. Patients with IBS-D displayed increased mast cell counts and
antibiotics in these syndromes. activation, increased protein expression of claudin-2, reduced occludin phosphor-
31. Spiller R, Garsed K. Postinfectious irritable bowel syndrome. Gastroenterol- ylation and enhanced redistribution from the membrane to the cytoplasm, increased
ogy 2009; 136:1979–1988. myosin kinase expression, and enhanced phosphorylation of myosin. These altera-
32. Cremon C, Stanghellini V, Pallotti F, et al. Salmonella gastroenteritis during tions were associated with ultrastructural abnormalities at the apical junction complex
& childhood is a risk factor for irritable bowel syndrome in adulthood. Gastro- which correlated both with mast cell activation and clinical symptoms.
enterology 2014. [Epub ahead of print] 44. Barbara G, Cremon C, Carini G, et al. The immune system in irritable bowel
Prospective, controlled, cohort study assessing functional digestive symptoms, syndrome. J Neurogastroenterol Motil 2011; 17:349–359.
psychological factors, and QoL 16 years after a single culture-proven foodborne 45. Matricon J, Meleine M, Gelot A, et al. Review article: associations between
Salmonella enteritidis outbreak involving 1811, mostly pediatric, patients in immune activation, intestinal permeability and the irritable bowel syndrome.
Bologna, Italy. Acute Salmonella gastroenteritis was identified as a risk factor Aliment Pharmacol Ther 2012; 36:1009–1031.
for the development of long-term IBS symptoms (OR for IBS 1.92; 95% CI: 1.23– 46. Belmonte L, Beutheu Youmba S, Bertiaux-Vandaele N, et al. Role of toll like
2.98), but only if the infection occurs during childhood (OR for IBS in children 2.12; receptors in irritable bowel syndrome: differential mucosal immune activation
95% CI: 1.21–3.71). according to the disease subtype. PLoS One 2012; 7:e42777.
33. Porter CK, Tribble DR, Aliaga PA, et al. Infectious gastroenteritis and risk of 47. Cenac N, Andrews CN, Holzhausen M, et al. Role for protease activity in
developing inflammatory bowel disease. Gastroenterology 2008; 135:781– visceral pain in irritable bowel syndrome. J Clin Invest 2007; 117:636–647.
786. 48. Barbara G, Wang B, Stanghellini V, et al. Mast cell-dependent excitation of
34. Rajilić-Stojanović M, Biagi E, Heilig HG, et al. Global and deep molecular visceral-nociceptive sensory neurons in irritable bowel syndrome. Gastroen-
analysis of microbiota signatures in fecal samples from patients with irritable terology 2007; 132:26–37.
bowel syndrome. Gastroenterology 2011; 141:1792–1801. 49. Barbara G, Stanghellini V, De Giorgio R, et al. Activated mast cells in proximity
35. Jeffery IB, O’Toole PW, Öhman L, et al. An irritable bowel syndrome subtype to colonic nerves correlate with abdominal pain in irritable bowel syndrome.
&& defined by species-specific alterations in faecal microbiota. Gut 2012; Gastroenterology 2004; 126:693–702.
61:997–1006. 50. Cremon C, Gargano L, Morselli-Labate AM, et al. Mucosal immune activation
Interesting study investigating the association between microbiota and the brain– in irritable bowel syndrome: gender-dependence and association with diges-
gut axis in patients with IBS. Results showed that those with no modifications in tive symptoms. Am J Gastroenterol 2009; 104:392–400.
fecal microbiota had more mood disorders while those with modification in 51. Valdez-Morales EE, Overington J, Guerrero-Alba R, et al. Sensitization of
microbiota had changes in bowel physiology. & peripheral sensory nerves by mediators from colonic biopsies of diarrhea-
36. Jalanka-Tuovinen J, Salojarvi J, Salonen A, et al. Faecal microbiota composi- predominant irritable bowel syndrome patients: a role for PAR2. Am J
&& tion and host–-microbe cross-talk following gastroenteritis and in postinfec- Gastroenterol 2013; 108:1634–1643.
tious irritable bowel syndrome. Gut 2013. [Epub ahead of print] Study examining the impact of mediators from colonic biopsies of patients with IBS
First study to show that fecal microbiota of patients with postinfectious on intrinsic excitability of colonic nociceptive dorsal root ganglion neurons. Super-
IBS differs from that of controls and shares similarities with that of patients natants from patients with IBS-D but not IBS-C induced a marked increase in
with IBS with diarrhea, suggesting a common pathophysiology. Members of neuronal excitability by a protease-activated receptor 2-mediated mechanism.
Bacteroidetes phylum were increased 12-fold in patients, while controls had 52. Balestra B, Vicini R, Cremon C, et al. Colonic mucosal mediators from patients
35-fold more uncultured Clostridia. Microbial dysbiosis correlates with the with irritable bowel syndrome excite enteric cholinergic motor neurons.
expression of several host gene pathways, including impaired epithelial barrier Neurogastroenterol Motil 2012; 24:1118–e1570.
function. 53. Steck N, Mueller K, Schemann M, Haller D. Bacterial proteases in IBD and
37. Manichanh C, Eck A, Varela E, et al. Anal gas evacuation and colonic IBS. Gut 2012; 61:1610–1618.
& microbiota in patients with flatulence: effect of diet. Gut 2014. [Epub ahead 54. Vanuytsel T, van Wanrooy S, Vanheel H, et al. Psychological stress and
of print] corticotropin-releasing hormone increase intestinal permeability in humans by
Study analyzing the potential role of diet on digestive symptoms, anal gas a mast cell-dependent mechanism. Gut 2013. [Epub ahead of print]
evacuation, intestinal gas distribution, and colonic microbiota in 30 patients with 55. Keohane J, O’Mahony C, O’Mahony L, et al. Irritable bowel syndrome-type
flatulence. On a flatulogenic diet, both patients and controls recorded more symptoms in patients with inflammatory bowel disease: a real association or
abdominal symptoms, gas production, and evacuations. Interestingly, only micro- reflection of occult inflammation? Am J Gastroenterol 2010; 105:1788;
biota of patients developed instability in composition, while that of controls was 1789–1794; quiz 1795.
stable, suggesting that patients with flatulence have a poor tolerance of intestinal 56. Vivinus-Nebot M, Frin-Mathy G, Bzioueche H, et al. Functional bowel symp-
gas that is associated with microbiota instability. && toms in quiescent inflammatory bowel diseases: role of epithelial barrier
38. Carroll IM, Ringel-Kulka T, Ferrier L, et al. Fecal protease activity is associated disruption and low-grade inflammation. Gut 2014; 63:744–752.
with compositional alterations in the intestinal microbiota. PLoS One 2013; Important study evaluating the role of colonic barrier defects and low-grade
8:e78017. inflammation in cecal biopsies of 51 patients with IBS, 49 patients with quiescent
39. Salim SY, Soderholm JD. Importance of disrupted intestinal barrier in inflam- IBD and 27 controls. Paracellular permeability was significantly increased in IBS
matory bowel diseases. Inflamm Bowel Dis 2011; 17:362–381. and patients with quiescent IBD and IBS-like symptoms and associated with lower
40. Piche T. Tight junctions and IBS: the link between epithelial permeability, low- expression of ZO-1 and a-catenin. Intraepithelial lymphocytes and TNFa were
grade inflammation, and symptom generation? Neurogastroenterol Motil significantly increased only in patients with quiescent IBD and IBS-like symptoms.
2014; 26:296–302. 57. van Hoboken EA, Thijssen AY, Verhaaren R, et al. Symptoms in patients with
41. Piche T, Barbara G, Aubert P, et al. Impaired intestinal barrier integrity in the ulcerative colitis in remission are associated with visceral hypersensitivity and
colon of patients with irritable bowel syndrome: involvement of soluble mast cell activity. Scand J Gastroenterol 2011; 46:981–987.
mediators. Gut 2009; 58:196–201. 58. Akbar A, Yiangou Y, Facer P, et al. Expression of the TRPV1 receptor differs in
42. Camilleri M. Peripheral mechanisms in irritable bowel syndrome. N Engl J Med quiescent inflammatory bowel disease with or without abdominal pain. Gut
2012; 367:1626–1635. 2010; 59:767–774.

358 www.co-gastroenterology.com Volume 30  Number 4  July 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Anda mungkin juga menyukai