Вы находитесь на странице: 1из 10

Alimentary Pharmacology and Therapeutics

Review article: evidence for the role of gut microbiota in


irritable bowel syndrome and its potential influence on
therapeutic targets
H. L. DuPont

St Luke’s Medical Center, and Baylor SUMMARY


College of Medicine, The University of
Texas School of Public Health,
Houston, TX, USA. Background
Irritable bowel syndrome (IBS) is a prevalent gastrointestinal disease with a
substantial social and economic burden. Treatment options remain limited
Correspondence to: and research on the aetiology and pathophysiology of this multifactorial
Dr H. L. DuPont, UT School of Public disease is ongoing.
Health, 1200 Herman Pressler, E-733,
Houston, TX 77030, USA.
E-mail: hdupont@stlukeshealth.org Aim
To discuss the potential role of gut microbiota in the pathophysiology of
IBS and to identify possible interactions with pathophysiologic targets in
Publication data
IBS.
Submitted 4 November 2013
First decision 18 November 2013
Resubmitted 5 March 2014 Methods
Accepted 6 March 2014 Articles were identified via a PubMed database search [‘irritable bowel syn-
EV Pub Online 25 March 2014 drome’ AND (anti-bacterial OR antibiotic OR flora OR microbiota OR mi-
croflora OR probiotic)]. English-language articles were screened for
This uncommissioned review article was relevance. Full review of publications for the relevant studies was con-
subject to full peer-review.
ducted, including additional publications that were identified from individ-
ual article reference lists.

Results
The role of gut microbiota in IBS is supported by varying lines of evidence
from animal and human studies. For example, post-infectious IBS in
humans is well documented. In addition, certain probiotics and nonsystem-
ic antibiotics appear to be efficacious in the treatment of IBS. Mechanisms
involved in improving IBS symptoms likely go beyond mere changes in the
composition of the gut microbiota, and accumulating animal data support
the interplay of microbiota with other IBS targets, such as the gut–brain
axis, visceral hypersensitivity, mucosal inflammation and motility.

Conclusion
The role of the gut microbiota is still being elucidated; however, it appears
to be one of several important factors that contributes to the aetiology and
pathophysiology of the irritable bowel syndrome.

Aliment Pharmacol Ther 2014; 39: 1033–1042

ª 2014 John Wiley & Sons Ltd 1033


doi:10.1111/apt.12728
H. L. DuPont

INTRODUCTION compared with healthy controls.13–16, 24–31 For example,


Irritable bowel syndrome (IBS) is a chronic disease that Kassinen et al. extracted bacterial genomic DNA from
may affect up to 20% of the population and has a nega- faecal samples of patients with IBS (n = 24) and controls
tive social and economic impact on patients.1–5 Globally, (n = 23); not only was the microbiota significantly
the pooled prevalence of IBS is approximately 11% but altered in patients with IBS, but the composition varied
varies considerably, both by geographical location and by depending on the predominant form of IBS.15 These
diagnostic criteria applied.6 Compared with individuals findings were extended by a study examining the compo-
without IBS, patients with IBS symptoms report signifi- sition of faecal microbiota in patients with IBS (n = 62)
cantly reduced health-related quality of life and reduced compared with healthy controls (n = 46), in which
work productivity.1–3 Despite the substantial burden of researchers reported significant differences based on glo-
IBS, treatment options remain limited and research on bal and deep molecular analysis with duplicate micro-
the aetiology and pathophysiology of this multifactorial array and quantitative polymerase chain reaction.32 In
disease is ongoing.5, 7–9 another investigation, bacterial DNA from faecal samples
The gut microbiota has emerged as an important fac- of patients with diarrhoea-predominant IBS (IBS-D,
tor that may contribute to the pathophysiology of IBS.10 n = 23) were compared with healthy controls (n = 23),
Proposed beneficial effects of the gut microbiota include and taxonomic, structural and diversity differences in gut
maintenance of intestinal homoeostasis, maintenance of microbiota were reported. In particular, IBS-D was asso-
peristalsis, intestinal mucosal integrity, and protection ciated with significantly higher levels of Enterobacteria-
against pathogens through bacterial antagonism and ceae and significantly lower levels of Faecalibacterium
priming of host immune responses.11, 12 The putative prausnitzii vs. healthy controls, which suggests an imbal-
role of gut microbiota in IBS is supported by several ance of advantageous and potentially harmful intestinal
lines of evidence with varying degrees of literature sup- bacteria.14
port. These include the differences between microbiota Biopsy studies have shown reduced mucosal microbi-
in IBS and non-IBS populations, the development of IBS ota diversity in patients with IBS compared with con-
after intestinal infection (i.e. post-infectious IBS), the trols,26, 28 and one study showed that the number of
interesting preliminary data on colonic faecal microbiota mucosal-associated bacteria in patients with IBS nega-
transplantation (FMT) and the efficacy of certain probi- tively correlated with number of stools passed.30 In a
otics, prebiotics, synbiotics and nonsystemic antibiotics related study, two patients with IBS-D followed up for
in the treatment of IBS.13–23 6–8 weeks experienced temporal instability in faecal bac-
teria content that correlated with changes in symptom-
METHODS atology.27 A comparison of microbial diversity in
This narrative review will discuss the role of the gut mic- luminal and mucosal niches of patients with IBS-D
robiota in IBS, including possible interactions with other (n = 16) and healthy controls (n = 21) identified signifi-
pathophysiologic targets in IBS. Articles included in this cantly lower faecal microbial biodiversity in patients with
review were identified via the PubMed database through IBS-D.13 Compared with mucosal samples, microbial
December 2013 using the following search string: ‘irrita- diversity was significantly higher in faecal samples in
ble bowel syndrome’ AND (anti-bacterial OR antibiotic both patient groups. Interestingly, the difference in bio-
OR flora OR microbiota OR microflora OR probiotic). diversity in faecal vs. mucosal samples was greater in the
The search was limited to English-language articles with healthy controls than in patients with IBS-D.13 Reduced
the screening of >800 citation titles and abstracts to diversity in mucosal-associated bacteria in IBS was also
determine potential relevance. Review of more than 200 reported in a second study.26
publications was conducted and included additional pub- In contrast, another investigation reported no signifi-
lications that were identified from citations within indi- cant differences in mucosal and faecal microbial diversity
vidual articles. in patients with IBS (n = 47).16 This investigation did,
however, support the concept of significantly greater var-
MICROBIOTA AS A TARGET IN IBS iability in diversity in faecal samples from healthy con-
trols (n = 33) compared with those from patients with
Microbiota in IBS vs. the healthy human gut IBS. Variations in study findings may be related to dif-
Numerous studies have reported differences in the ferences in patient populations and methods of sample
mucosal and/or faecal microbiota of patients with IBS collection.

1034 Aliment Pharmacol Ther 2014; 39: 1033-1042


ª 2014 John Wiley & Sons Ltd
Review: gut microbiota and therapy targets in irritable bowel syndrome

Small intestinal bacterial overgrowth (SIBO) has been microbiota, the therapeutic approaches that are known
associated with some cases of IBS; however, issues such as to directly target the microbiota include FMT, probiotics
inconsistency of study findings, trial heterogeneity, meth- and nonsystemic antibiotics.
odological problems, lack of validation of small bowel cul-
ture techniques, lack of validation of breath tests and Faecal microbiota transplantation. Preliminary data in
concerns regarding bias have led to the clinical relevance patients with IBS suggest a favourable response to colo-
of SIBO in IBS being questioned.18, 19, 33–35 A key issue is nic FMT, which is consistent with the concept that the
the lack of a gold standard for the diagnosis of SIBO; in gut microbiota has a role in the pathogenesis of IBS. In
particular, it has been demonstrated that the commonly one report, 10 patients (n = 5 with IBS) received antibi-
used lactulose breath test does not reliably distinguish otics and bowel lavage followed by a colonic infusion of
patients with IBS from healthy controls.36 Interestingly, faeces from a healthy donor.21 Examination of stool indi-
scintigraphy studies suggest that breath testing measures cated that FMT resulted in a novel microbiota mainly
variations in small bowel transit time occurring in patients composed of the bacterial species from the donor, and
with IBS, rather than the presence of SIBO.37 Therefore, the microbiota composition remained generally stable
SIBO may be a comorbid condition in a small subset of over 24 weeks. Although of interest, the published evi-
patients with IBS instead of having a direct pathophysio- dence for FMT in IBS is limited, and larger comparative
logic link to the development of IBS. studies are needed to clarify the potential role for FMT
in the management of IBS.
Post-infectious IBS
One of the most compelling arguments for microbiota Probiotics, prebiotics and synbiotics in IBS. The efficacy
involvement in IBS pathophysiology is that in some of probiotics in IBS has been reviewed in several publica-
patients, an acute episode of gastroenteritis precedes the tions.39–43 Overall efficacy has been modest, with varia-
onset of IBS. A meta-analysis of eight studies reported tion in strains of probiotics showing potential benefit.40–
42
an odds ratio (OR) of 7.3 [95% confidence interval (CI): One area of concern is that many of the marketed
4.7–11.1] for developing IBS after a gastrointestinal probiotics have not been adequately evaluated in well-de-
infection, supporting a link between the two.17 Addi- signed clinical trials.44 While meta-analyses have been
tional studies have supported that intestinal infection positive, the pooled relative risks may be influenced by
was strongly associated with a subsequent emergence of the limitations of study methodologies and the possibility
IBS symptoms. When data from nine prospective studies of bias.22, 41 Also, given the heterogeneity of both probi-
were pooled, the OR for developing post-infectious IBS otic composition and patient populations studied, the
was 5.9 (95% CI: 3.6–9.5), and the risk remained ability to extrapolate findings is limited.22, 45 A 2013
increased for up to 3 years post-infection.38 In the meta-analysis of probiotics in IBS attempted to address
meta-analysis, factors associated with a greater risk of some of these limitations. The authors identified 10 stud-
developing IBS post-infection were younger age; psycho- ies meeting relatively robust criteria for inclusion (e.g.
logical disturbance (i.e. increased anxiety and/or depres- randomised placebo-controlled design, IBS defined by
sion as observed in four studies reporting Hospital Rome criteria).22 As expected, efficacy varied by probiot-
Anxiety and Depression Scale scores); and the nature of ic species as well as by the IBS symptom evaluated. Also,
the gastrointestinal illness (i.e. prolonged fever during not all probiotic strains significantly improved pain, dis-
the acute episode, possibly indicative of more severe ill- tention and flatulence, whereas improvements in other
ness).38 In a 2013 study, the faecal microbiota of patients symptoms such as stool frequency, urgency and straining
with post-infectious IBS resembled that seen in idio- were not significant, and effects on quality of life were
pathic IBS-D.29 inconsistent with probiotic therapy.22
The mechanism of action of probiotics in IBS may
IBS therapeutic approaches targeting the gut extend beyond the modulation of the microbiota composi-
microbiota tion.46–48 For example, 4-week administration of a yogurt
Several treatments, including those with effects on the containing probiotic, with known benefits in IBS, to
gut microbiota, may be potentially useful in the manage- patients with IBS (n = 19) did not alter the composition
ment of IBS, based on the current knowledge of IBS of their microbiota.46 However, the authors suggest that
pathophysiology (Figure 1). Although any therapy that probiotic effects on the functionality of the microbiota
changes intestinal motility may have indirect effects on may be of importance.46 In one study, probiotic adminis-

Aliment Pharmacol Ther 2014; 39: 1033-1042 1035


ª 2014 John Wiley & Sons Ltd
H. L. DuPont

Predisposing host factors


Psychological factors, genetic makeup (serotonin transport genes,
inflammatory pathway genes [GN 3, IL-10, proinflammatory genes])

Small and large Microbiota dysbiosis: Disturbed brain


intestine and the bacterial diversity, altered and central and
intestinal nervous balance of Firmicutes peripheral nervous
and immune system (Veillonella, Lactobacillus, and system and
Faecalibacterium) and spinal excitability,
Chronic low-grade
Bacteroidetes (Prevotella), hypothalamus-
inflammation/immune
Bifidobacteriaceae, pituitary-adrenal
activation involving
Proteobacteria stress response
innate and adaptive
(Enterobacteriaceae)
immune activation
with changes in
toll-like receptor
expression and Abdominal pain,
changes in intestinal Transcription of antimicrobial changes in mood
epithelium proteins, cytokines, and or cognition, anxiety,
chemokines via NF- B pathway, and depression
serotonin release, mast cell
activation, regulatory T cells,
Mucosal neurotransmitters, and
permeability neuropeptides

Infection by enteric pathogen


in postinfectious IBS

Visceral hypersensitivity
and dysmotility

Afferent nerves of vagus nerve,


spinal cord, and autonomic
nervous system

Irritable bowel syndrome Intestinal stasis in


constipation form
Abdominal discomfort and
altered bowel habits

Figure 1 | Pathogenesis of IBS summarising multifactorial disordered pathways, the bidirectional gut–brain axis and
potential targets for therapy. IBS, irritable bowel syndrome; IL, interleukin.

tration has been reported to normalise cytokine levels [e.g. tions combining probiotics with prebiotics) have also
interleukin-10 (IL-10) and IL-12] in patients with IBS.49 A been evaluated in the treatment of IBS.51, 52 For exam-
pilot neuroimaging study reported that healthy females ple, the first investigation of a prebiotic (trans-galactooli-
(n = 12) who ingested a fermented milk product with gosaccharide mixture) in patients with IBS was an
probiotic for 4 weeks had significant reductions in the 18-week study in 44 patients randomly assigned to one
activity of brain regions related to a sensory brain network of three treatment groups: group 1 (n = 16), which
compared with females who ingested a nonfermented milk received placebo for 6 weeks followed by 3.5 g prebiotic
control (n = 11) or had no intervention (n = 13).50 for 12 weeks; group 2 (n = 14), which received placebo
While probiotics have been more widely studied, the for 6 weeks followed by 7.0 g prebiotic for 12 weeks;
potential benefits of prebiotics and synbiotics (formula- and group 3 (n = 14), which received placebo for

1036 Aliment Pharmacol Ther 2014; 39: 1033-1042


ª 2014 John Wiley & Sons Ltd
Review: gut microbiota and therapy targets in irritable bowel syndrome

18 weeks.51 The prebiotic treatment groups had qualita- (40.7% vs. 31.7%, pooled data; P < 0.001), and relief was
tive changes in faecal flora compared with the placebo sustained for at least 10 weeks post-treatment.56
group (i.e. relative proportions of Bifidobacterium spp.) Improvement post-treatment was consistent with another
and significant improvements compared with the placebo smaller randomised, double-blind, placebo-controlled
group in terms of stool consistency, flatulence composite study of a lower dose of rifaximin (400 mg three times a
scores and patient subjective global assessments. day; n = 43) vs. placebo (n = 44) for 10 days in patients
A synbiotic preparation (n = 132) containing a probi- with IBS meeting Rome I criteria.58 Patients treated with
otic [viable lyophilised Lactobacillus paracasei B21060 rifaximin experienced a greater mean percentage global
(5 9 109 colony-forming units)] and prebiotic [xylooli- improvement in IBS at 10 weeks post-treatment com-
gosaccharides (700 mg) and glutamine (500 mg)] or a pared with those receiving placebo (36.4  31.5% vs.
control formulation (n = 135) was administered for 21.0  22.1%; P = 0.02).58
12 weeks in patients with IBS based on Rome II crite- Another randomised, double-blind, placebo-controlled
ria.52 The synbiotic preparation alleviated IBS symptoms study (n = 124 patients, with 20%, 38.3% and 41.7%
but failed to demonstrate a significant benefit vs. the receiving IBS-D, IBS-C and alternating IBS diagnoses
administration of a control formulation, except in a sub- respectively) evaluating rifaximin for chronic bloating
group of patients with diarrhoea predominance (n = 47; and flatulence indicated that, in a subgroup of patients
27 received synbiotic, 20 received control). These results with IBS meeting Rome II criteria (n = 70), a signifi-
contrast with those of a previous uncontrolled open-label cantly larger response was observed with rifaximin
study of Bifidobacterium longum W11 and an oligosac- 800 mg/day after 10 days compared with placebo (40.5%
charide-based synbiotic preparation, which improved vs. 18.2% respectively; P = 0.04).57 A meta-analysis of
intestinal function in patients with constipation-variant randomised, placebo-controlled studies evaluating rifaxi-
IBS based on Rome II criteria.53 min for IBS reported that global symptoms of IBS
improved significantly with rifaximin vs. placebo (OR:
Nonsystemic antibiotics in IBS. Most randomised, pla- 1.57; P < 0.001), with a number needed to treat of 10.59
cebo-controlled studies have evaluated nonsystemic In a 2012 observational study, patients with IBS (total
agents (e.g. neomycin, rifaximin) for the treatment of n = 106; 88 IBS-D, 7 IBS-C, 11 alternating IBS) were
IBS (Table 1).54–58 A randomised, double-blind study in treated with rifaximin; patients responded (i.e. experi-
patients with IBS meeting Rome I criteria compared neo- enced reduction in bloating, flatulence, diarrhoea and
mycin [n = 55; 25 patients with IBS-D, 18 with constipa- pain) after 2 weeks, and results were maintained for at
tion-predominant IBS (IBS-C), 10 with other/unknown least 3 months.60 In addition, several retrospective chart
forms of IBS] with placebo (n = 56; 21 patients with reviews of rifaximin treatment for IBS suggest a low
IBS-D, 20 with IBS-C and 15 with other/undetermined potential for loss of efficacy with repeated courses of rif-
forms of IBS).54 A significantly greater reduction from aximin. In two retrospective chart reviews (both in
baseline in IBS symptoms (primary endpoint: composite patients with nonconstipated forms of IBS), clinical
score based on abdominal pain, diarrhoea, constipation) improvement was documented in ~75% of patients with-
was observed with neomycin compared with placebo out loss of response during retreatments.61, 62 Similarly,
(35.0  5.0% vs. 11.4  9.3%; P < 0.05). A subanalysis another retrospective chart review of rifaximin-treated
of patients with IBS-C (neomycin, n = 19; placebo, patients meeting Rome I criteria for IBS showed that
n = 20) indicated that neomycin treatment resulted in a 69% of patients had a clinical response. This was greater
significantly greater global percentage improvement in than the response reported with other antibiotics (i.e.
IBS compared with placebo (36.7  7.9% vs. 38% of 24 patients treated with neomycin experienced a
5.0  3.2%; P < 0.001).55 response).63 Retreatment with rifaximin (n = 16) was
Two randomised, double-blind, placebo-controlled tri- associated with clinical improvement in all cases,
als (n = 1260) evaluated rifaximin 550 mg three times a whereas retreatment with other antibiotics (e.g. doxycy-
day in patients with IBS-D meeting Rome II criteria.56 cline, amoxicillin and clavulanate acid, neomycin) was
After 14 days of treatment, adequate relief of global IBS effective in only two of eight cases (25%).
symptoms for ≥2 of the first 4 weeks post-treatment (i.e. As suggested with probiotics, the action of nonsystem-
primary endpoint) was achieved in significantly more ic antibiotics in IBS may extend beyond direct effects on
patients in the rifaximin group compared with those in the composition of the gut microbiota. For example,
the placebo group in each study and in a pooled analysis additional mechanisms of action for rifaximin have been

Aliment Pharmacol Ther 2014; 39: 1033-1042 1037


ª 2014 John Wiley & Sons Ltd
H. L. DuPont

Table 1 | Published randomised, placebo-controlled, clinical data for antibiotics in the management of IBS

Antibiotic study Dosage Patient population Primary outcome (antibiotic vs. placebo)* P-value
Neomycin
Pimentel et al.54 500 mg b.d. for 10 days IBS (Rome I); n = 111 Per cent reduction in composite <0.05
IBS score†: 35.0% vs. 11.4%
Pimentel et al.55 500 mg b.d. for 10 days IBS (Rome I); n = 39‡ Global improvement in IBS <0.001
symptoms: 36.7% vs. 5.0%
Rifaximin
Pimentel et al.56 550 mg t.d.s. for 14 days Nonconstipated IBS Adequate relief of global IBS symptoms 0.01
(Rome II); n = 623 during ≥2 of first 4 weeks post-treatment:
40.8% vs. 31.2%
Pimentel et al.56 550 mg t.d.s. for 14 days Nonconstipated IBS Adequate relief of global IBS symptoms 0.03
(Rome II); n = 637 during ≥2 of first 4 weeks post-treatment:
40.6% vs. 32.2%
Sharara et al.57 400 mg b.d. for 10 days IBS (Rome II); n = 70§ Global symptom improvement: 0.04
40.5% vs. 18.2%
Pimentel et al.58 400 mg t.d.s. for 10 days IBS (Rome I); n = 87 Global IBS symptom improvement: 0.02
36.4% vs. 21.0%

b.d., two times a day; t.d.s., three times a day.


* Per cent of patients, unless otherwise indicated.
† Composite score of three main IBS symptoms: abdominal pain, diarrhoea, constipation.
‡ Subgroup of patients with constipation-predominant IBS.
§ Subgroup of patients in the study who had IBS.

proposed, such as a reduction in bacterial pathogen viru- Microbiota–gut–brain axis


lence,64 the stabilisation of gut mucosa (i.e. protection The important role of the brain–gut/gut–brain axis in
against bacterial infection),65 anti-inflammatory proper- IBS and in other functional gastrointestinal diseases has
ties,65 and the preservation of normal colonic flora.66 been previously reported on,68–72 while the concept of a
Other potential pathogenic processes in IBS that may be microbiota–gut–brain axis in health and disease is
modulated by rifaximin include visceral hypersensitivity emerging.73–75 Of related interest, in patients with cir-
and mucosal-immune reactivity (based on animal model rhosis (n = 25), differences in gut microbiota were
data).67 observed vs. healthy controls (n = 10), and there was a
direct correlation between several bacterial taxa and cog-
INTERPLAY OF MICROBIOTA AND OTHER nitive function, particularly in 17 patients with hepatic
TREATMENT TARGETS IN IBS encephalopathy.76 As noted above, animal data interpre-
Gut microbiota is one of many factors contributing to tation has limitations, especially given the lack of suitable
the pathophysiology of IBS, and each represents a poten- IBS animal models; however, pre-clinical data can pro-
tial target for therapy (Figure 1). Given the complexity vide insight into the potential relationship between the
of the interactions between these factors/targets, it is pos- microbiota and the gut–brain axis. For example, mice
sible that therapies directly targeting the microbiota may administered a mixture of nonsystemic antibiotics had
also impact other pathways either directly or indirectly an altered microbiota, increased exploratory behaviour
through their effects on the gut microbiota. The majority and less apprehensive behaviour compared with con-
of the evidence supporting the potential interplay of the trols.77 After a 2-week antibiotic washout period, these
gut microbiota with other targets is derived from animal behavioural changes were reversed as the microbiota
studies (where there is no suitable IBS model), meaning normalised. The authors noted that, consistent with the
that in the absence of human studies, clinical relevance behavioural changes, mice receiving the antibiotic mix-
cannot be established. Nonetheless, these pre-clinical ture also had higher levels of central brain-derived neu-
studies have led to the development of hypotheses on rotropic factor in the hippocampus and lower levels in
gut microbiota interaction and are laying the ground- the amygdala compared with controls.77 Collins et al.
work for future investigation.

1038 Aliment Pharmacol Ther 2014; 39: 1033-1042


ª 2014 John Wiley & Sons Ltd
Review: gut microbiota and therapy targets in irritable bowel syndrome

has provided a comprehensive review of data on the administration of certain probiotics for IBS. To assess
bidirectional interactions between the gut microbiota and for a protective effect from antibiotic-induced increases
the brain (animal-based studies), noting that while ani- in visceral sensitivity, a probiotic (L. paracasei) was co-
mal data support the influence of gut microbiota on administered with an antibiotic in mice. Visceral hyper-
brain development and function, information elucidating sensitivity and histology improved; however, total
the possible underlying mechanisms is lacking.75 Current Lactobacilli populations were undetectable,82 suggesting
knowledge on how intestinal microbiota dysbiosis, the that the beneficial effects observed may not simply be
gut–brain axis and pathophysiologic changes help to related to recolonisation of the microbiota.
explain the disease pathophysiology in IBS is summar- In rats with induced post-inflammatory chronic
ised in Figure 1. hypersensitivity to colorectal distention, the administra-
tion of a probiotic resulted in the normalisation of vis-
Inflammation and visceral hypersensitivity ceral sensitivity.83
The gut microbiota may contribute to the low-grade The protective effect of three different probiotic
inflammation and intestinal immune activation described strains was assessed in rat models of psychological stress
in IBS through effects on cytokine levels and toll-like (i.e. chronic maternal deprivation-induced visceral hyper-
receptor activity.24, 78, 79 In a study of patients with IBS sensitivity associated with altered colonic paracellular
(n = 77) undergoing colonoscopy to rule out inflamma- permeability or acute partial restraint-induced visceral
tory bowel disease, patients were categorised based on hypersensitivity).84 Only one of the three probiotic
biopsy as non-inflamed IBS, nonspecific microscopic strains, L. paracasei NCC2461 (Lpa), demonstrated
colitis or lymphocytic colitis.80 Increases in lymphocytic reversal of visceral hypersensitivity to colorectal disten-
populations were observed in all patient subgroups, even tion and complete restoration of gut paracellular perme-
those with no overt signs of inflammation, suggesting a ability. Thus, effects of probiotics on visceral
pathophysiologic role of immune activation in IBS. The hypersensitivity appear to be strain specific. In the partial
authors speculated that bacterial antigens could be one restraint-induced animal model, anti-nociceptive effects
of the factors triggering immune activation.80 on visceral hypersensitivity were observed only when
Visceral hypersensitivity has also been implicated in Lpa and the spent culture medium were both present.
IBS, and the influence of gut microbiota on both visceral The authors suggested a potential synergistic interplay
hypersensitivity and inflammation has been suggested in between the probiotic and intraluminal bacterial prod-
a variety of animal models, several of which are sum- ucts.84
marised in this section. In one study, the transfer of fae- The effect of oral rifaximin on gut microbiota, intesti-
cal microbiota of patients with IBS to germ-free rats was nal inflammation and visceral hyperalgesia was evaluated
accompanied by a transfer of visceral hypersensitivity in a model of visceral hyperalgesia (chronic water avoid-
(assessed by colorectal distention) when these IBS ance and restraint stress in adult Wistar rats).67 Rifaxi-
human microbiota-associated (HMA) rats were com- min administration significantly reduced the ileal
pared with healthy HMA rats.81 An investigation of bacterial load and altered the bacterial composition in
whether changes in gut flora and gut inflammatory cell the distal ileum (i.e. dominance of Lactobacilli post-treat-
activity impacted visceral hypersensitivity in mice dem- ment). In addition, rifaximin inhibited mucosal inflam-
onstrated that, in the absence of sterile precautions (i.e. mation, barrier impairment and visceral hyperalgesia.
allowing for the fluctuation of gut bacterial content), the Although these animal data are interesting, the associa-
mice had a substantial increase in visceral sensitivity over tions between gut microbiota and inflammation and vis-
time that was associated with a slightly increased activity ceral hypersensitivity remain speculative and require
of inflammatory cells.82 When an anti-inflammatory support from clinical investigations in patients with IBS.
agent (i.e. dexamethasone) was administered, both
inflammatory activity and visceral hypersensitivity were Motility
reduced, lending further support to the interplay between In addition, bacterial factors in the gut may cause
inflammation and visceral sensitivity. Overall, these changes in motor function that may underlie IBS symp-
results support the hypothesis that perturbations of the toms.85 Colonisation with different bacterial species in
gut microbiota are associated with small changes rats showed that changes in the composition of gut mic-
in inflammatory activity in the gut that can change vis- robiota were associated with altered (either impeded or
ceral perception; this is a possible rationale for the increased) gastrointestinal motility.86 It has also been

Aliment Pharmacol Ther 2014; 39: 1033-1042 1039


ª 2014 John Wiley & Sons Ltd
H. L. DuPont

reported that bacterial components (e.g., lipopolysaccha- unknown. Studies are needed to further refine the role of
rides) may interact with specific toll-like receptors and the intestinal microbiota in the gut–brain axis and in the
directly affect the contractile function of human colonic chronic inflammation associated with the disorder, and
smooth muscle cells.87 Anti-motility agents are often how these factors influence the important intestinal
used in IBS-D,88 and their effect may be related in part events in the syndrome. As we define these factors, IBS
to changes in gut flora. will move from a functional syndrome to a disease with
a defined biochemical and immunological origin.
CONCLUSIONS
Animal and human data support an interplay between AUTHORSHIP
the gut microbiota and pathophysiologic factors targeted Guarantor of the article: H. L. DuPont.
by agents evaluated for the management of IBS. Further Author contributions: H. L. DuPont provided concept
research on bacterial populations identified in stool and and scientific content for the development of the
associated with intestinal mucosa of healthy individuals manuscript, critically reviewed and edited all drafts of
may provide new insight and opportunities to normalise the manuscript, and approved the final version of the
gut homoeostasis in patients with IBS. Currently, several manuscript.
probiotics and nonsystemic antibiotics are efficacious for
the treatment of patients with IBS. However, given the ACKNOWLEDGEMENTS
complexity of the IBS disease process, it is likely that the Declaration of personal interests: In 2011, Dr DuPont
mechanisms of these agents are not limited to their served as a consultant to Salix Pharmaceuticals, Inc. on
direct effects on microbiota composition.9, 48, 89–92 Gut the topic of antibiotic resistance with chronic use of
microbiota appears to be one of several important factors rifaximin and has received research grants from Santau-
that may contribute to the aetiology and pathophysiology rus, Inc., a company that has been acquired by Salix
of IBS. Future research on metabolic and/or immunolog- Pharmaceuticals, Inc.
ical pathways and microbial genes in the gastrointestinal Declaration of funding interests: Writing and editorial
tract (metagenomics) and their effects on protein expres- support were provided under the direction of the author
sion will help elucidate the role of gut microbiota in this by Mary Beth Moncrief, PhD, and Kulvinder Singh,
burdensome condition.19, 75 PharmD, Synchrony Medical Communications, LLC,
While much is known about the pathophysiology of West Chester, PA, and funded by Salix Pharmaceuticals,
IBS and the role of the microbiota, a great deal more is Inc.

REFERENCES
1. Dean BB, Aguilar D, Barghout V, et al. 6. Lovell RM, Ford AC. Global prevalence 11. Berg RD. The indigenous gastro-
Impairment in work productivity and of and risk factors for irritable bowel intestinal microflora. Trends Microbiol
health-related quality of life in patients syndrome: a meta-analysis. Clin 1996; 4: 430–5.
with IBS. Am J Manag Care 2005; 11: Gastroenterol Hepatol 2012; 10: 712–21. 12. Marchesi J, Shanahan F. The normal
S17–26. 7. Ringel Y, Carroll IM. Alterations in the intestinal microbiota. Curr Opin Infect
2. Gralnek IM, Hays RD, Kilbourne A, intestinal microbiota and functional Dis 2007; 20: 508–13.
Naliboff B, Mayer EA. The impact of bowel symptoms. Gastrointest Endosc 13. Carroll IM, Ringel-Kulka T, Keku TO,
irritable bowel syndrome on health- Clin N Am 2009; 19: 141–50, vii. et al. Molecular analysis of the luminal-
related quality of life. Gastroenterology 8. Ruepert L, Quartero AO, de Wit NJ, and mucosal-associated intestinal
2000; 119: 654–60. van der Heijden GJ, Rubin G, Muris microbiota in diarrhea-predominant
3. Bolino CM, Bercik P. Pathogenic JW. Bulking agents, antispasmodics and irritable bowel syndrome. Am J Physiol
factors involved in the development of antidepressants for the treatment of Gastrointest Liver Physiol 2011; 301:
irritable bowel syndrome: focus on a irritable bowel syndrome. Cochrane G799–807.
microbial role. Infect Dis Clin North Database Syst Rev 2011; 10: CD003460. 14. Carroll IM, Ringel-Kulka T, Siddle JP,
Am 2010; 24: 961–75, ix. 9. Olden KW. Targeted therapies for Ringel Y. Alterations in composition
4. Maxion-Bergemann S, Thielecke F, diarrhea-predominant irritable bowel and diversity of the intestinal
Abel F, Bergemann R. Costs of syndrome. Clin Exp Gastroenterol 2012; microbiota in patients with diarrhea-
irritable bowel syndrome in the UK 5: 69–100. predominant irritable bowel syndrome.
and US. Pharmacoeconomics 2006; 24: 10. Ohman L, Simren M. Intestinal Neurogastroenterol Motil 2012; 24:
21–37. microbiota and its role in irritable e248.
5. Ford AC, Talley NJ. Irritable bowel bowel syndrome (IBS). Curr 15. Kassinen A, Krogius-Kurikka L,
syndrome. BMJ 2012; 345: e5836. Gastroenterol Rep 2013; 15: 323. Makivuokko H, et al. The fecal

1040 Aliment Pharmacol Ther 2014; 39: 1033-1042


ª 2014 John Wiley & Sons Ltd
Review: gut microbiota and therapy targets in irritable bowel syndrome

microbiota of irritable bowel syndrome 29. Jalanka-Tuovinen J, Salojarvi J, Salonen bowel syndrome. BMC Gastroenterol
patients differs significantly from that A, et al. Faecal microbiota composition 2009; 9: 15.
of healthy subjects. Gastroenterology and host-microbe cross-talk following 41. McFarland LV, Dublin S. Meta-analysis
2007; 133: 24–33. gastroenteritis and in postinfectious of probiotics for the treatment of
16. Codling C, O’Mahony L, Shanahan F, irritable bowel syndrome. Gut 2013; irritable bowel syndrome. World J
Quigley EM, Marchesi JR. A molecular doi: 10.1136/gutjnl-2013-305994 [Epub Gastroenterol 2008; 14: 2650–61.
analysis of fecal and mucosal bacterial ahead of print]. 42. Moayyedi P, Ford AC, Talley NJ, et al.
communities in irritable bowel 30. Parkes GC, Rayment NB, Hudspith BN, The efficacy of probiotics in the
syndrome. Dig Dis Sci 2010; 55: 392–7. et al. Distinct microbial populations treatment of irritable bowel syndrome:
17. Halvorson HA, Schlett CD, Riddle MS. exist in the mucosa-associated a systematic review. Gut 2010; 59:
Postinfectious irritable bowel syndrome microbiota of sub-groups of irritable 325–32.
– a meta-analysis. Am J Gastroenterol bowel syndrome. Neurogastroenterol 43. Cui S, Hu Y. Multistrain probiotic
2006; 101: 1894–9. Motil 2012; 24: 31–9. preparation significantly reduces
18. Ford AC, Spiegel BM, Talley NJ, 31. Rigsbee L, Agans R, Shankar V, et al. symptoms of irritable bowel syndrome
Moayyedi P. Small intestinal bacterial Quantitative profiling of gut microbiota in a double-blind placebo-controlled
overgrowth in irritable bowel syndrome: of children with diarrhea-predominant study. Int J Clin Exp Med 2012; 5:
systematic review and meta-analysis. irritable bowel syndrome. Am J 238–44.
Clin Gastroenterol Hepatol 2009; 7: Gastroenterol 2012; 107: 1740–51. 44. Ringel Y, Ringel-Kulka T. The rationale
1279–86. 32. Rajilic-Stojanovic M, Biagi E, Heilig and clinical effectiveness of probiotics
19. Simren M, Barbara G, Flint HJ, et al. HG, et al. Global and deep molecular in irritable bowel syndrome. J Clin
Intestinal microbiota in functional analysis of microbiota signatures in Gastroenterol 2011; 45(Suppl.): S145–8.
bowel disorders: a Rome foundation fecal samples from patients with 45. Whelan K. Probiotics and prebiotics in
report. Gut 2013; 62: 159–76. irritable bowel syndrome. the management of irritable bowel
20. Borody TJ, George L, Andrews P, et al. Gastroenterology 2011; 141: 1792–801. syndrome: a review of recent clinical
Bowel-flora alteration: a potential cure 33. Pyleris E, Giamarellos-Bourboulis EJ, trials and systematic reviews. Curr Opin
for inflammatory bowel disease and Tzivras D, Koussoulas V, Barbatzas C, Clin Nutr Metab Care 2011; 14: 581–7.
irritable bowel syndrome? Med J Aust Pimentel M. The prevalence of 46. Maccaferri S, Candela M, Turroni S,
1989; 150: 604. overgrowth by aerobic bacteria in the et al. IBS-associated phylogenetic
21. Grehan MJ, Borody TJ, Leis SM, small intestine by small bowel culture: unbalances of the intestinal microbiota
Campbell J, Mitchell H, Wettstein A. relationship with irritable bowel are not reverted by probiotic
Durable alteration of the colonic syndrome. Dig Dis Sci 2012; 57: 1321–9. supplementation. Gut Microbes 2012; 3:
microbiota by the administration of 34. Lin HC, Pimentel M. Bacterial concepts 406–13.
donor fecal flora. J Clin Gastroenterol in irritable bowel syndrome. Rev 47. Quigley EM, Flourie B. Probiotics and
2010; 44: 551–61. Gastroenterol Disord 2005; 5(Suppl. 3): irritable bowel syndrome: a rationale
22. Ortiz-Lucas M, Tobias A, Saz P, S3–9. for their use and an assessment of the
Sebastian JJ. Effect of probiotic species 35. Pimentel M, Chow EJ, Lin HC. evidence to date. Neurogastroenterol
on irritable bowel syndrome symptoms: Eradication of small intestinal bacterial Motil 2007; 19: 166–72.
a bring up to date meta-analysis. Rev overgrowth reduces symptoms of 48. Kumar A, Rinwa P, Sharma N. Irritable
Esp Enferm Dig 2013; 105: 19–36. irritable bowel syndrome. Am J bowel syndrome: a review. J Phys
23. Sachdev AH, Pimentel M. Antibiotics Gastroenterol 2000; 95: 3503–6. Pharm Adv 2012; 2: 97–108.
for irritable bowel syndrome: rationale 36. Bratten JR, Spanier J, Jones MP. 49. O’Mahony L, McCarthy J, Kelly P,
and current evidence. Curr Lactulose breath testing does not et al. Lactobacillus and bifidobacterium
Gastroenterol Rep 2012; 14: 439–45. discriminate patients with irritable in irritable bowel syndrome: symptom
24. Pimentel M, Chang C. Inflammation bowel syndrome from healthy controls. responses and relationship to cytokine
and microflora. Gastroenterol Clin Am J Gastroenterol 2008; 103: 958–63. profiles. Gastroenterology 2005; 128:
North Am 2011; 40: 69–85. 37. Yu D, Cheeseman F, Vanner S. 541–51.
25. Chassard C, Dapoigny M, Scott KP, Combined oro-caecal scintigraphy and 50. Tillisch K, Labus J, Kilpatrick L, et al.
et al. Functional dysbiosis within the gut lactulose hydrogen breath testing Consumption of fermented milk
microbiota of patients with constipated- demonstrate that breath testing detects product with probiotic modulates brain
irritable bowel syndrome. Aliment oro-caecal transit, not small intestinal activity. Gastroenterology 2013; 144:
Pharmacol Ther 2012; 35: 828–38. bacterial overgrowth in patients with 1394–401.
26. Durban A, Abellan JJ, Jimenez- IBS. Gut 2011; 60: 334–40. 51. Silk DB, Davis A, Vulevic J, Tzortzis G,
Hernandez N, et al. Structural 38. Thabane M, Kottachchi DT, Marshall Gibson GR. Clinical trial: the effects of
alterations of faecal and mucosa- JK. Systematic review and meta- a trans-galactooligosaccharide prebiotic
associated bacterial communities in analysis: the incidence and prognosis of on faecal microbiota and symptoms in
irritable bowel syndrome. Environ post-infectious irritable bowel irritable bowel syndrome. Aliment
Microbiol Rep 2012; 4: 242–7. syndrome. Aliment Pharmacol Ther Pharmacol Ther 2009; 29: 508–18.
27. Durban A, Abellan JJ, Jimenez- 2007; 26: 535–44. 52. Andriulli A, Neri M, Loguercio C, et al.
Hernandez N, et al. Instability of the 39. Whelan K, Quigley EM. Probiotics in Clinical trial on the efficacy of a new
faecal microbiota in diarrhoea- the management of irritable bowel symbiotic formulation, Flortec, in
predominant irritable bowel syndrome. syndrome and inflammatory bowel patients with irritable bowel syndrome:
FEMS Microbiol Ecol 2013; 86: 581–9. disease. Curr Opin Gastroenterol 2013; a multicenter, randomized study. J Clin
28. Ng SC, Lam EF, Lam TT, et al. Effect 29: 184–9. Gastroenterol 2008; 42(Suppl. 3 Pt 2):
of probiotic bacteria on the intestinal 40. Hoveyda N, Heneghan C, Mahtani KR, S218–23.
microbiota in irritable bowel syndrome. Perera R, Roberts N, Glasziou P. A 53. Dughera L, Elia C, Navino M, Cisaro F.
J Gastroenterol Hepatol 2013; 28: systematic review and meta-analysis: Effects of symbiotic preparations on
1624–31. probiotics in the treatment of irritable constipated irritable bowel syndrome

Aliment Pharmacol Ther 2014; 39: 1033-1042 1041


ª 2014 John Wiley & Sons Ltd
H. L. DuPont

symptoms. Acta Biomed 2007; 78: cells with rifaximin alters bacterial 80. Chadwick VS, Chen W, Shu D, et al.
111–6. attachment and internalization profiles. Activation of the mucosal immune
54. Pimentel M, Chow EJ, Lin HC. Antimicrob Agents Chemother 2010; 54: system in irritable bowel syndrome.
Normalization of lactulose breath 388–96. Gastroenterology 2002; 122: 1778–83.
testing correlates with symptom 66. DuPont HL. Biologic properties and 81. Crouzet L, Gaultier E, Del’Homme C,
improvement in irritable bowel clinical uses of rifaximin. Expert Opin et al. The hypersensitivity to colonic
syndrome. a double-blind, randomized, Pharmacother 2011; 12: 293–302. distension of IBS patients can be
placebo-controlled study. Am J 67. Xu D, Gao J, Gillilland M III, et al. transferred to rats through their fecal
Gastroenterol 2003; 98: 412–9. Rifaximin alters intestinal bacteria and microbiota. Neurogastroenterol Motil
55. Pimentel M, Chatterjee S, Chow EJ, prevents stress-induced gut 2013; 25: e272–82.
Park S, Kong Y. Neomycin improves inflammation and visceral hyperalgesia 82. Verd u EF, Bercik P, Verma-Gandhu M,
constipation-predominant irritable in rats. Gastroenterology 2014; 146: et al. Specific probiotic therapy
bowel syndrome in a fashion that is 484–96. attenuates antibiotic induced visceral
dependent on the presence of methane 68. Koloski NA, Jones M, Kalantar J, hypersensitivity in mice. Gut 2006; 55:
gas: subanalysis of a double-blind Weltman M, Zaguirre J, Talley NJ. The 182–90.
randomized controlled study. Dig Dis brain–gut pathway in functional 83. Johnson AC, Greenwood-Van MB,
Sci 2006; 51: 1297–301. gastrointestinal disorders is bidirectional: McRorie J. Effects of Bifidobacterium
56. Pimentel M, Lembo A, Chey WD, et al. a 12-year prospective population-based infantis 35624 on post-inflammatory
Rifaximin therapy for patients with study. Gut 2012; 61: 1284–90. visceral hypersensitivity in the rat. Dig
irritable bowel syndrome without 69. Mayer EA. The neurobiology of stress Dis Sci 2011; 56: 3179–86.
constipation. N Engl J Med 2011; 364: and gastrointestinal disease. Gut 2000; 84. Eutamene H, Lamine F, Chabo C, et al.
22–32. 47: 861–9. Synergy between Lactobacillus paracasei
57. Sharara AI, Aoun E, Abdul-Baki H, 70. Mayer EA, Tillisch K. The brain-gut and its bacterial products to counteract
Mounzer R, Sidani S, Elhajj I. A axis in abdominal pain syndromes. stress-induced gut permeability and
randomized double-blind placebo- Annu Rev Med 2011; 62: 381–96. sensitivity increase in rats. J Nutr 2007;
controlled trial of rifaximin in patients 71. Drossman DA. Presidential address: 137: 1901–7.
with abdominal bloating and flatulence. gastrointestinal illness and the 85. Azpiroz F, Bouin M, Camilleri M, et al.
Am J Gastroenterol 2006; 101: 326–33. biopsychosocial model. Psychosom Med Mechanisms of hypersensitivity in IBS
58. Pimentel M, Park S, Mirocha J, Kane 1998; 60: 258–67. and functional disorders.
SV, Kong Y. The effect of a 72. Whitehead WE, Crowell MD, Robinson Neurogastroenterol Motil 2007; 19:
nonabsorbed oral antibiotic (rifaximin) JC, Heller BR, Schuster MM. Effects of 62–88.
on the symptoms of the irritable bowel stressful life events on bowel symptoms: 86. Husebye E, Hellstrom PM, Sundler F,
syndrome: a randomized trial. Ann subjects with irritable bowel syndrome Chen J, Midtvedt T. Influence of
Intern Med 2006; 145: 557–63. compared with subjects without bowel microbial species on small intestinal
59. Menees SB, Maneerattannaporn M, dysfunction. Gut 1992; 33: 825–30. myoelectric activity and transit in germ-
Kim HM, Chey WD. The efficacy and 73. Rhee SH, Pothoulakis C, Mayer EA. free rats. Am J Physiol Gastrointest Liver
safety of rifaximin for the irritable Principles and clinical implications of Physiol 2001; 280: G368–80.
bowel syndrome: a systematic review the brain-gut-enteric microbiota axis. 87. Tattoli I, Petitta C, Scirocco A,
and meta-analysis. Am J Gastroenterol Nat Rev Gastroenterol Hepatol 2009; 6: Ammoscato F, Cicenia A, Severi C.
2012; 107: 28–35. 306–14. Microbiota, innate immune system, and
60. Meyrat P, Safroneeva E, Schoepfer AM. 74. Forsythe P, Kunze WA. Voices from gastrointestinal muscle: ongoing studies.
Rifaximin treatment for the irritable within: gut microbes and the CNS. Cell J Clin Gastroenterol 2012; 46(Suppl.):
bowel syndrome with a positive lactulose Mol Life Sci 2013; 70: 55–69. S6–11.
hydrogen breath test improves 75. Collins SM, Surette M, Bercik P. The 88. Efskind PS, Bernklev T, Vatn MH. A
symptoms for at least 3 months. Aliment interplay between the intestinal double-blind placebo-controlled trial
Pharmacol Ther 2012; 36: 1084–93. microbiota and the brain. Nat Rev with loperamide in irritable bowel
61. Pimentel M, Morales W, Chua K, et al. Microbiol 2012; 10: 735–42. syndrome. Scand J Gastroenterol 1996;
Effects of rifaximin treatment and 76. Bajaj JS, Ridlon JM, Hylemon PB, et al. 31: 463–8.
retreatment in nonconstipated IBS Linkage of gut microbiome with 89. Cremonini F, Talley NJ. Treatments
subjects. Dig Dis Sci 2011; 56: 2067–72. cognition in hepatic encephalopathy. targeting putative mechanisms in
62. Weinstock LB. Long-term outcome of Am J Physiol Gastrointest Liver Physiol irritable bowel syndrome. Nat Clin Pract
rifaximin therapy in non-constipation 2012; 302: G168–75. Gastroenterol Hepatol 2005; 2: 82–8.
irritable bowel syndrome. Dig Dis Sci 77. Bercik P, Denou E, Collins J, et al. The 90. Dahlqvist G, Piessevaux H. Irritable
2011; 56: 3389–90. intestinal microbiota affect central levels bowel syndrome: the role of the
63. Yang J, Lee HR, Low K, Chatterjee S, of brain-derived neurotropic factor and intestinal microbiota, pathogenesis and
Pimentel M. Rifaximin versus other behavior in mice. Gastroenterology therapeutic targets. Acta Gastroenterol
antibiotics in the primary treatment 2011; 141: 609. Belg 2011; 74: 375–80.
and retreatment of bacterial overgrowth 78. Dupont AW, DuPont HL. The 91. Camilleri M. Current and future
in IBS. Dig Dis Sci 2008; 53: 169–74. intestinal microbiota and chronic pharmacological treatments for
64. Jiang ZD, Ke S, DuPont HL. Rifaximin- disorders of the gut. Nat Rev diarrhea-predominant irritable bowel
induced alteration of virulence of Gastroenterol Hepatol 2011; 8: 523–31. syndrome. Expert Opin Pharmacother
diarrhoea-producing Escherichia coli 79. McKernan DP, Gaszner G, Quigley EM, 2013; 14: 1151–60.
and Shigella sonnei. Int J Antimicrob Cryan JF, Dinan TG. Altered peripheral 92. Quigley EM. Do patients with
Agents 2010; 35: 278–81. toll-like receptor responses in the functional gastrointestinal disorders
65. Brown EL, Xue Q, Jiang ZD, Xu Y, irritable bowel syndrome. Aliment have an altered gut flora? Therap Adv
DuPont HL. Pretreatment of epithelial Pharmacol Ther 2011; 33: 1045–52. Gastroenterol 2009; 2: 23–30.

1042 Aliment Pharmacol Ther 2014; 39: 1033-1042


ª 2014 John Wiley & Sons Ltd

Вам также может понравиться