Skip to main content
Gut Microbes logoLink to Gut Microbes
. 2014 Jul 9;5(3):430–436. doi: 10.4161/gmic.29796

Effect of commensals and probiotics on visceral sensitivity and pain in irritable bowel syndrome

Vassilia Theodorou 1,2,*, Afifa Ait Belgnaoui 1,2,3, Simona Agostini 1,2, Helene Eutamene 1,2
PMCID: PMC4153783  PMID: 25184834

Abstract

The last ten years’ wide progress in the gut microbiota phylogenetic and functional characterization has been made evidencing dysbiosis in several gastrointestinal diseases including inflammatory bowel diseases and irritable bowel syndrome (IBS). IBS is a functional gut disease with high prevalence and negative impact on patient’s quality of life characterized mainly by visceral pain and/or discomfort, representing a good paradigm of chronic gut hypersensitivity. The IBS features are strongly regulated by bidirectional gut-brain interactions and there is increasing evidence for the involvement of gut bacteria and/or their metabolites in these features, including visceral pain. Further, gut microbiota modulation by antibiotics or probiotics has been promising in IBS. Mechanistic data provided mainly by animal studies highlight that commensals or probiotics may exert a direct action through bacterial metabolites on sensitive nerve endings in the gut mucosa, or indirect pathways targeting the intestinal epithelial barrier, the mucosal and/or systemic immune activation, and subsequent neuronal sensitization and/or activation.

Keywords: irritable bowel syndrome, visceral pain, brain-gut axis, probiotics, microbiota, gut permeability, intestinal barrier, stress

Introduction

The human intestine harbors a vast and complex microbial ecosystem called microbiota linked to the host by a symbiotic relationship leading to gut homeostasis. Thus, for the bacterial prokaryote the alimentary tract offers a supply of nutrients and inversely the host benefits from bacterial metabolism products and complex interactions leading to immune and non-immune host defense. Bacteria are the main microorganisms (there are also representatives from archaea and eukarya as well as viruses1) composing this ecosystem, but their species and number remained largely underestimated, before the rapid evolution of molecular approaches occurring in the last ten years. Currently, the number of bacterial species identified in human fecal samples exceeds 1000.2 The normal gut microbiota of adult humans is stable over time and highly resilient to transient aggression.3 However, a breakdown in the mutualistic microbiota-host relationship, called dysbiosis, has been observed in several gastrointestinal pathologies, such as inflammatory bowel diseases (IBD)4,5 and irritable bowel syndrome (IBS) reflected by changes in the stability, diversity, composition, and/or metabolism of gut bacteria.6-12 On this basis, therapeutic strategies targeting the gut microbiota have emerged as for example antibiotics13 or probiotics use.14 Definitely, the most striking findings coming mostly from animal studies highlight that gut microbiota, or its modulation by antibiotics or probiotics can exert effects beyond the gut targeting spinal or supraspinal sites of action and strongly support the concept of the microbiota-gut-brain axis (for a review see ref. 15). Interestingly, in addition to gut dysbiosis, visceral pain, and psychiatric co-morbidity are common features in functional gastrointestinal disorders, such as IBS.16 This encouraged investigations using IBS-like animal models to explore the microbiota-gut brain interactions and contribute in the understanding of IBS physiopathology. In this text we will focus on the effects and mechanisms of action of gut microbiota and probiotics on visceral pain based on animal studies and we will discuss the relevance of gut microbiota modulation probiotics in IBS.

Visceral Sensitivity in IBS: The Top Down and Bottom Up Hypotheses

IBS is a common and highly prevalent disorder which affect, according to the diagnosis Rome criteria applied, 5–20% of the global population.17,18 Despite high prevalence the precise etiology and pathophysiology of IBS remains poorly understood and is often referred as multifactorial. IBS is a good paradigm of chronic gut hypersensitivity and visceral pain, defined as an unpleasant sensation which in contrast to somatic pain remains diffuse and difficult to localize.19 Accordingly, Ritchie et al. in 1972 reported that chronic abdominal pain in IBS patients is associated with lower visceral painful sensations in response to repeated inflations of a rectal balloon.20 However, visceral sensitivity to rectal distension is not a universal feature of IBS patients. Accordingly, the literature reports only 21 to 94% of IBS patients as hypersensitive to rectal distension, evidencing the presence of normosensitive individuals among this population and suggesting distinct pathophysiology pathways involved.2 For instance, patients with visceral hypersensitivity to colorectal distension reported increased pain responses to rectal application of capsaicin, a TRPV1 agonist when compared with normosensitive IBS patients and healthy controls.21 Other predominant IBS features are changes in bowel habits and bloating22 originating from intestinal motility changes and gut abnormal fermentations respectively.23 Gut microbiota composition, stability, and functional changes have also been reported in IBS.6-12 The phylogenetic microbial characterization is conducted mainly from fecal samples. Despite the lack of identification of a specific microbial group characteristic for IBS dysbiosis the alterations generally reported concern the Bifidobacterium and Clostridium coccoides-E. rectal subgroup.

The overall of these IBS features occur without identifiable organic cause and are strongly influenced by stress and anxiety suggesting the involvement of brain-gut interactions in this disease.24 The origin of visceral hypersensitivity in IBS can be central and/or peripheral (for review see ref. 25). The first works on the mechanisms involving the brain-gut axis were based on the communication between the gut intrinsic innervation (enteric nervous system; ENS), connected on the extrinsic innervation (the autonomic nervous system), which interacts with the hypothalamic-pituitary axis and affects visceral sensory motor functions.26 The input from splanchnic visceral afferences is received in the spinal dorsal horn and from this site second-order neurons transmit the visceral sensory information to supraspinal sites, the final integration of the painful perception occurring in the cortex.27 Vagal afferences activation plays a modulatory role on the spinal visceral pain pathway.28 In IBS, increased neuron excitability resulting to visceral hypersensitivity has been reported at several levels of the brain-gut axis, i.e., ENS, spinal cord and supraspinal sites.29 In the supraspinal sites interactions with emotional or stressful influences can modulate the visceral sensitivity resulting to increased pain perception.30

Besides central influences on visceral sensory function in IBS, peripheral factors such as luminal agents (bacteria, or microbial antigens) may also establish a cross-talk with the brain-gut axis and contribute to IBS visceral hypersensitivity. A strong support to this hypothesis was the evidence indicating that IBS may be the adverse outcome of an acute episode of gastroenteritis characterized as post-infectious IBS (PI-IBS) and was proposed 50 years ago by Chaudary and Truelove.31 Interestingly, increased intestinal paracellular permeability has been reported in these patients both in a study investigating persistent IBS symptoms after Campylobacter jejuni infection or after a large waterborne outbreak of acute gastroenteritis in Canada.32,33 Further, a posteriori investigations of the Canada outbreak highlighted genes encoding for proteins involved in epithelial cell barrier function and the innate immune response to enteric bacteria, which may be considered as potential genetic determinants of PI-IBS.34

More recently, prospective studies have shown that 3% to 36% of enteric infections lead to persistent new IBS symptoms, the precise incidence depends on the infecting organism.35 Interestingly, the microbiota profile analysis in PI-IBS patients revealed that several members of Bacteroidetes phylum were increased 12-fold in patients, while healthy controls had 35-fold more uncultured Clostridia. Further, the particular PI-IBS microbiota profile was positively correlated with host gene expression related to epithelial cell junction integrity and inflammatory response.36 Systemic immune activation against bacterial luminal antigens has also been described in IBS and particularly in PI-IBS, since antibodies against flagellin (the primary structural component of bacterial flagella) were observed more frequently in IBS vs. healthy controls and among IBS population these antibodies were more frequent in the PI-IBS subset.37 Another argument supporting the contribution of luminal bacteria in the visceral hypersensitivity is related to functional dysbiosis and altered intestinal fermentation described in IBS. Chassard et al.12 report increased sulfate-reducing bacteria population in fecal microbiota of constipated predominant IBS patients (IBS-C), producing more sulfides,12 known to enhance visceral nociception38 and H2 able to generate gas-related symptoms such as bloating and flatus.39 Abnormal intestinal fermentation in IBS has also been described in the small intestinal bacterial overgrowth (SIBO) hypothesis, where a 4-fold greater rate of maximal gas excretion and greater total hydrogen production in the presence of a fermentable substrate was described.40 Interestingly, antibiotic treatment leading to SIBO eradication improved also IBS symptomatology.41 Moreover, antimicrobials targeting IBS management, have also been used in clinical trials and experimental studies. These are mainly locally acting antibiotics for obvious reasons of systemic antibiotics related adverse effects. The efficacy of neomycin (a poorly absorbed aminoglycoside antibiotic) was tested in IBS-C population.42 Constipation was improved by 32.6 ± 9.9% with neomycin vs. 18.7 ± 7.2% for placebo. Further the methane producers IBS-C patients treated by neomycin exhibited higher constipation improvement vs. placebo suggesting that this improvement depends on the elimination of methane on breath test.42 Another study from the same group has shown that neomycin normalized lactulose breath test in IBS leading to a marked decrease of IBS symptoms.43 However for safety reasons44,45 the use of neomycin for IBS symptoms improvement seems inadequate. Strong clinical evidence in IBS emerged concerning the efficacy of rifaximin, a synthetic derivative of rifamycin acting locally in the gastrointestinal tract and used commonly for the traveler’s diarrhea. In a phase III double-blind placebo-controlled trial, a two week treatment by rifaximin induced an improvement of the IBS symptoms such as bloating, abdominal pain and stool consistency and had a response to treatment during the first 4 weeks after completion of the treatment vs. the placebo group.46 In a phase IV trial evaluating the efficacy of rifaximin in IBS patients, the authors confirmed previous data and reported that rifaximin improved IBS symptoms for 3 months following 2 weeks treatment in lactulose hydrogen breath test-positive IBS patients.47 These clinical data clearly support the involvement of microbiota in the IBS pathogenesis and visceral pain.

Regarding the rate of the main bacterial metabolites issue from intrestinal bacteria fermentation, namely short chain fatty acids (SCFA), production in IBS as well as their influence on the associated visceral sensitivity, the literature data remain contradictory. Increased SCFA production in IBS has been described with elevated severity of visceral pain in patients with the highest SCFA levels48 whereas other studies reported lower concentrations of SCFA in IBS.49 Such discrepancies can be attributed to uncontrolled diet differences among patients. Further, increased or decreased visceral nociceptive perception has been described after intracolonic instillation of butyrate.50,51 However whether gut dysbiosis in IBS is a cause or a consequence of the disease remains unclear.

Gut Microbiota and Visceral Sensitivity: Mechanistic Aspects

The incidence of the luminal milieu (bacteria and their by-products) to the IBS associated visceral pain highlights the possibility of a cross talk between gut microorganisms and/or their metabolites, intestinal barrier, mucosal immunity and neural pathways. It has been recently shown that visceral hypersensitivity characterizing IBS-C patients can be transferred to rats by the fecal microbiota.52 Accordingly, germ free rats inoculated with IBS-C fecal suspension exhibited increased visceral sensitivity in response to colorectal distension compared with germ free rats inoculated with healthy controls fecal suspension in absence of any mucosal abnormality and change in gut permeability.52 In contrast, germ free rats inoculated with IBS-C microbiota presented abnormal gut fermentation mostly characterized by increased H2 excretion and sulfides production vs. controls.52 Authors suggest that H2 and hydrogen sulfide may be bacterial metabolites responsible for visceral hypersensitivity since H2 increased production has been described in IBS39 and colorectal infusion of hydrogen sulfide directly triggers visceral nociceptive behavior in mice through sensitization and/or activation of T-type channels probably in the primary afferents.38 Besides direct action of bacterial metabolites on sensitive terminals, pathways involving epithelial barrier and neuro-immune interactions may also occur. Interestingly, the presence of a low grade inflammation in the gastrointestinal tract of several IBS subtypes has been described.53 This inflammatory tone involving neuro-immune interactions in the gut mucosa may generate mediators able to sensitize nerve afferents resulting to viscera hypersensitivity. Indeed, in addition to mechanosensitive receptors and plymodal endings the gut also contains a class of mechanically insensitive receptors called “silent” receptors which may respond to mechanical stimulation after organ insult such as inflammation and amplify visceral sensitivity.54

In rodents, antibiotic treatment altering gut flora, was associated with increased colonic inflammatory tone and substance P (a sensory neurotransmitter) immunoreactivity, resulting to visceral hypersensitivity.55 The involvement of neuro-immune interactions is further supported by findings showing that IBS patients have significantly increased number of immune cells in the gut mucosa, such as T cells32 and mast cells56,57 and increased fecal levels of human β-defensin 2 (HBD-2)58 indicating activation of the innate immune system compared with controls. Interestingly, T cell activation induces changes in the enteric neuromuscular function59 and the colonic mast cells infiltrate in IBS is closer to enteric nerve endings vs. controls as well as positively correlated with severity and frequency of abdominal pain or discomfort.60 Moreover, mediators released from colonic IBS biopsies such as histamine serotonin and mast cell proteases can activate human submucosal enteric neurons possibly contributing by this way to visceral hypersensitivity.61 Besides mast cells, colonic bacteria are also able to release proteases.62 For instance, Lactobacilli exhibiting high ability to cleave proteases (anti-protease activity) are decreased in IBS diarrhea-predominant patients (IBS-D).7,9 Interestingly, an increase in fecal serine-protease activity mostly from microbial origin has been observed in IBS-D patients compared with controls and IBS-C patients.63 Further, the intracolonic application of IBS-D fecal supernatants in mice, increased colonic paracellular permeability and induced visceral hypersensitivity through a protease activated-receptor type 2 (PAR2) pathway.64 More recently an increase in luminal cysteine-protease activity was described in a subset of IBS-C patients positively correlated with disease severity and abdominal pain scoring.65 Further, repeated colonic mucosal application in mice of fecal supernatant from this subset of IBS-C patients induced gut hyperpermeability linked to the enzymatic degradation of occludin, and was associated with enhanced visceral sensitivity.65 As pointed by these studies the intestinal epithelial barrier regulating the trans and paracellular passage in the gut represents another key factor in the communication between the luminal milieu, mucosal immunity and nerve endings. Interestingly, increased gut permeability has been described in IBS patients whatever the sub type considered66 and a positive correlation between increased gut paracellular permeability and visceral pain and discomfort has been shown.67 In a recent study the increased number of mast cells determined in the jejunum of IBS-D patients was found associated with intestinal epithelial apical junction complex alterations as reflected by reduced occludin phosphorylation and redistribution in the cytoplasm and enhanced MLCK phosphorylation.68 Further, these changes at the proteic level were associated with ultrastructural abnormalities at the apical junction complex, i.e., perijunctional cytoskeleton condensation and enlarged apical intercellular distance clearly evoking epithelial barrier impairment.68 In another study of the same group, investigating IBS-D biopsies, reduced zonula occludens 1 (ZO-1) expression in the jejunum and redistribution in the cytoplasm have also been reported.69 More interestingly, these epithelial barrier defects were correlated with increased number of mast cells and clinical symptoms.69 Taken together these data underline the major role of epithelial barrier impairment in the IBS pathogenesis. Moreover, animal studies highlight a cause effect relationship between gut hyperpermeability and visceral hypersensitivity induced by stress.70 Indeed, in rats blockade of the intestinal epithelial cell cytoskeleton contraction (by a MLCK inhibitor, ML-7), and subsequent tight junctions opening, prevented visceral hypersensitivity induced by an acute stress.70

Probiotics and Visceral Sensitivity

Probiotics are defined as live microorganisms which when ingested in adequate amounts confer a health benefit on the host.71 In a recent systematic review aiming at establish a reference guide on the role of specified probiotics in the management of lower gastrointestinal disorders, including IBS, by means of systematic review-based consensus, authors reported evidence of an overall benefit of probiotics use in IBS.72 Among statements worded to reflect the grade of available evidence (high, moderate, or low), “specific probiotics help to reduce abdominal pain, bloating, and/or distension in some patients with IBS” was recorded as high.72 However, clinical trials testing probiotics efficacy in IBS remain generally difficult to compare due to differences in the study design (size of the study, duration of the treatment), probiotic dose and strain used. Further, some of them are conducted using probiotic mixtures rather than single strains, and even combinations including prebiotics rendering difficult to decipher the active moieties and their mechanism of action. Regarding safety, an important issue in the probiotics use, Lactobacilli, Bifidobacteria, and other commensal microorganisms are generally regarded as safe. However the safety can be compromised and a risk of bacteremia can emerge when probiotics are massively administered in immunodepressed or in severe illness suffering patients.73-75 Besides a overall beneficial but modest effect of priobiotic treatments in different meta-analyses, further investigations are clearly needed in order to establish optimal regimens (the most effective probiotic species and strains, individual or mixture administration [synergistic effect]) as well as to identify subgroups of patients most likely to benefit from these treatments.

From the mechanistic point of view, arguments in favor of the probiotics use in IBS come mostly from animal studies and are based mainly in their ability (1) to modulate visceral sensitivity and (2) to enhance intestinal barrier function and immunity. In this paragraph will be discussed literature data concerning these abilities.

Alterations in the gut microbiota and inflammatory tone induced by non absorbable antibiotics treatment enhanced visceral sensitivity and substance P expression in the colonic mucosa.55 These effects were prevented by L paracasei administration.55 In another study, treatment with live, killed, or even the conditioned medium of L reuteri prevented the pain response to colorectal distension, as reflected by inhibition of the constitutive cardio-autonomic response to colorectal distension and decrease in the dorsal root ganglion single unit activity to distension.76 A decrease of normal visceral perception and chronic colonic hypersensitivity, elicited by butyrate enemas, was also observed after an oral treatment by L acidophilus NCFM.77 This effect was associated with an increased expression of μ-opioid (MOR1) and cannabinoid receptors (CB2) in intestinal epithelial cells.77

Acute and chronic stress models are widely used as IBS-like models, since stress induces gut hyperpermeability and visceral hypersensitivity in response to colorectal distension.78,79 The ability of probiotic strains to enhance intestinal epithelial barrier function was reported by Zareie et al.,80 showing that a commercially available probiotic combination (L helveticus R0052 [5%] and L rhamnosus R0011 [95%] treatment) prevented the ileal epithelial barrier impairment and bacterial translocation to the mesenteric lymph nodes induced by a chronic stress (water avoidance stress).80 Similarly, treatment by the same combination, prevented colonic epithelial barrier disruption in adult rats submitted in a maternal deprivation stress during the neonatal period.81

The prevention of stress-induced gut hyperpermeability has been described as responsible for stress-induced visceral hypersensitivity in rodents.70 Accordingly, treatment by L. farciminis prevented stress-induced visceral hypersensitivity and colonic paracellular hyperpermeability, through a decrease of the myosin light chain (MLC) phosphorylation, responsible for epithelial cells cytoskeleton contraction and subsequent tight junction opening.82 Further, the analgesic effect of L farciminis was confirmed by data showing a decrease in the enhanced Fos protein expression at the spinal (spinal cords sections S1, S2, and L6) and supraspinal (hypothalamic paraventricula nucleus) sites induced by colorectal distension in stressed rats.83 In another study comparing the effect of three probiotic strains (L paracasei NCC2461 B lactis NCC362 and L johnsonii NCC533) using two stress models in rats (acute restraint stress and neonatal maternal deprivation) it has be shown that only L paracasei NCC2461 associated with its conditioned medium was able to restore gut permeability and visceral sensitivity alterations induced by the stress, underlying the strain specificity and the synergy between live bacteria and their metabolites generated in the medium, for the stress-induced IBS-like symptoms improvement.84 Similarly, in a study comparing the efficacy of three probiotics (L salivarius UCC118, B infantis 35624, or B breve UCC2003) on the abdominal response to colorectal distension using visceral normosensitive rats (Spargue-Daweley) and visceral hypersensitive rats (Wistar Kyoto), only B infantis 35624 reduced the colorectal distension-induced pain behavior in both rat strains.85 Interestingly, in a clinical trial, comparing an eight weeks treatment by B infantis 35624 and L salivarius UCC118 in IBS patients, only B infantis 35624 treated patients experienced a reduction in composite and individual scores for abdominal pain and/or discomfort, bloating/distension.86 It is of particular interest that in this trial B infantis 35624 treatment normalized the low IL-10/IL-12 ratio (indicative of a Th1 profile) characterizing the IBS patients before treatment, suggesting that the antinociceptrive effect of this strain is linked to a decrease of the IBS inflammatory tone.86 The beneficial effects of probiotic isolated strains on stress-induced gastrointestinal disturbances can be extended to treatments with fermented dietary products containing probiotics. Accordingly, it has been recently shown that treatment with a fermented milk containing B lactis CNCM I-2494 and yogurt strains reduced stress-induced visceral hypersensitivity by normalizing intestinal epithelial barrier via a synergistic interplay between the different probiotic strains and/or metabolites contained in this product.87

Conclusions

The understanding of the role of microbiota in the bidirectional cross talk between the gut and the brain is definitively a new and fascinating research field. As such, the mechanisms of action involved in the visceral pain modulation and even more generally in the central nervous system influences by the microbiota are not yet clearly established. Many questions at different levels need answers. For instance, (1) regarding the long lasting effects of neonatal stress in the visceral sensitivity and gut epithelial barrier integrity, a new challenge may be the determination of the gut’s bacteria role in this persisting imprinting. (2) If disturbances of the gut microbiota are observed in several pathologies it is not known if they are cause or consequences of the pathology. Further, in our opinion the functional characterization approach in the IBS dysbiosis may be more informative, at least from a mechanistic point of view, than phylogenetic characterization (3) if modulation of the gut microbiota by probiotics or antimicrobial compounds leads to observable effects there is need to understand the specificity of each agent in the precise effect and to decipher the molecular basis in the dialog with the gut and beyond the gut.

Disclosure of Potential Conflicts of Interest

No potential conflict of interest was disclosed.

10.4161/gmic.29796

References

  • 1.Guarner F. The intestinal flora in inflammatory bowel disease: normal or abnormal? Curr Opin Gastroenterol. 2005;21:414–8. [PubMed] [Google Scholar]
  • 2.Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. MetaHIT Consortium A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65. doi: 10.1038/nature08821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mondot S, de Wouters T, Doré J, Lepage P. The human gut microbiome and its dysfunctions. Dig Dis. 2013;31:278–85. doi: 10.1159/000354678. [DOI] [PubMed] [Google Scholar]
  • 4.Manichanh C, Borruel N, Casellas F, Guarner F. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol. 2012;9:599–608. doi: 10.1038/nrgastro.2012.152. [DOI] [PubMed] [Google Scholar]
  • 5.Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán LG, Gratadoux JJ, Blugeon S, Bridonneau C, Furet JP, Corthier G, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008;105:16731–6. doi: 10.1073/pnas.0804812105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Jeffery IB, O’Toole PW, Öhman L, Claesson MJ, Deane J, Quigley EM, Simrén M. An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut. 2012;61:997–1006. doi: 10.1136/gutjnl-2011-301501. [DOI] [PubMed] [Google Scholar]
  • 7.Kassinen A, Krogius-Kurikka L, Mäkivuokko H, Rinttilä T, Paulin L, Corander J, Malinen E, Apajalahti J, Palva A. The fecal microbiota of irritable bowel syndrome patients differs significantly from that of healthy subjects. Gastroenterology. 2007;133:24–33. doi: 10.1053/j.gastro.2007.04.005. [DOI] [PubMed] [Google Scholar]
  • 8.Kerckhoffs AP, Samsom M, van der Rest ME, de Vogel J, Knol J, Ben-Amor K, Akkermans LM. Lower Bifidobacteria counts in both duodenal mucosa-associated and fecal microbiota in irritable bowel syndrome patients. World J Gastroenterol. 2009;15:2887–92. doi: 10.3748/wjg.15.2887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Malinen E, Rinttilä T, Kajander K, Mättö J, Kassinen A, Krogius L, Saarela M, Korpela R, Palva A. Analysis of the fecal microbiota of irritable bowel syndrome patients and healthy controls with real-time PCR. Am J Gastroenterol. 2005;100:373–82. doi: 10.1111/j.1572-0241.2005.40312.x. [DOI] [PubMed] [Google Scholar]
  • 10.Mättö J, Maunuksela L, Kajander K, Palva A, Korpela R, Kassinen A, Saarela M. Composition and temporal stability of gastrointestinal microbiota in irritable bowel syndrome--a longitudinal study in IBS and control subjects. FEMS Immunol Med Microbiol. 2005;43:213–22. doi: 10.1016/j.femsim.2004.08.009. [DOI] [PubMed] [Google Scholar]
  • 11.Rajilić-Stojanović M, Biagi E, Heilig HG, Kajander K, Kekkonen RA, Tims S, de Vos WM. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011;141:1792–801. doi: 10.1053/j.gastro.2011.07.043. [DOI] [PubMed] [Google Scholar]
  • 12.Chassard C, Dapoigny M, Scott KP, Crouzet L, Del’homme C, Marquet P, Martin JC, Pickering G, Ardid D, Eschalier A, et al. Functional dysbiosis within the gut microbiota of patients with constipated-irritable bowel syndrome. Aliment Pharmacol Ther. 2012;35:828–38. doi: 10.1111/j.1365-2036.2012.05007.x. [DOI] [PubMed] [Google Scholar]
  • 13.Meyrat P, Safroneeva E, Schoepfer AM. Rifaximin treatment for the irritable bowel syndrome with a positive lactulose hydrogen breath test improves symptoms for at least 3 months. Aliment Pharmacol Ther. 2012;36:1084–93. doi: 10.1111/apt.12087. [DOI] [PubMed] [Google Scholar]
  • 14.O’Mahony L, McCarthy J, Kelly P, Hurley G, Luo F, Chen K, O’Sullivan GC, Kiely B, Collins JK, Shanahan F, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology. 2005;128:541–51. doi: 10.1053/j.gastro.2004.11.050. [DOI] [PubMed] [Google Scholar]
  • 15.Saulnier DM, Ringel Y, Heyman MB, Foster JA, Bercik P, Shulman RJ, Versalovic J, Verdu EF, Dinan TG, Hecht G, et al. The intestinal microbiome, probiotics and prebiotics in neurogastroenterology. Gut Microbes. 2013;4:17–27. doi: 10.4161/gmic.22973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mayer EA. Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci. 2011;12:453–66. doi: 10.1038/nrn3071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Saito YA, Schoenfeld P, Locke GR., 3rd The epidemiology of irritable bowel syndrome in North America: a systematic review. Am J Gastroenterol. 2002;97:1910–5. doi: 10.1111/j.1572-0241.2002.05913.x. [DOI] [PubMed] [Google Scholar]
  • 18.Hungin AP, Chang L, Locke GR, Dennis EH, Barghout V. Irritable bowel syndrome in the United States: prevalence, symptom patterns and impact. Aliment Pharmacol Ther. 2005;21:1365–75. doi: 10.1111/j.1365-2036.2005.02463.x. [DOI] [PubMed] [Google Scholar]
  • 19.Robinson DR, Gebhart GF. Inside information: the unique features of visceral sensation. Mol Interv. 2008;8:242–53. doi: 10.1124/mi.8.5.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ritchie JA, Ardran GM, Truelove SC. Observations on experimentally induced colonic pain. Gut. 1972;13:841–7. [PubMed] [Google Scholar]
  • 21.van Wanrooij SJ, Wouters MM, Van Oudenhove L, Vanbrabant W, Mondelaers S, Kollmann P, Kreutz F, Schemann M, Boeckxstaens GE. Sensitivity testing in irritable bowel syndrome with rectal capsaicin stimulations: role of TRPV1 upregulation and sensitization in visceral hypersensitivity? Am J Gastroenterol. 2014;109:99–109. doi: 10.1038/ajg.2013.371. [DOI] [PubMed] [Google Scholar]
  • 22.Palsson OS, Baggish JS, Turner MJ, Whitehead WE. IBS patients show frequent fluctuations between loose/watery and hard/lumpy stools: implications for treatment. Am J Gastroenterol. 2012;107:286–95. doi: 10.1038/ajg.2011.358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gasbarrini A, Lauritano EC, Garcovich M, Sparano L, Gasbarrini G. New insights into the pathophysiology of IBS: intestinal microflora, gas production and gut motility. Eur Rev Med Pharmacol Sci. 2008;12(Suppl 1):111–7. [PubMed] [Google Scholar]
  • 24.Mayer EA. Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci. 2011;12:453–66. doi: 10.1038/nrn3071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Barbara G, Cremon C, De Giorgio R, Dothel G, Zecchi L, Bellacosa L, Carini G, Stanghellini V, Corinaldesi R. Mechanisms underlying visceral hypersensitivity in irritable bowel syndrome. Curr Gastroenterol Rep. 2011;13:308–15. doi: 10.1007/s11894-011-0195-7. [DOI] [PubMed] [Google Scholar]
  • 26.Mayer EA. Emerging disease model for functional gastrointestinal disorders. Am J Med. 1999;107(5A):12S–9S. doi: 10.1016/S0002-9343(99)00277-6. [DOI] [PubMed] [Google Scholar]
  • 27.Kellow JE, Azpiroz F, Delvaux M, Gebhart GF, Mertz HR, Quigley EM, Smout AJ. Applied principles of neurogastroenterology: physiology/motility sensation. Gastroenterology. 2006;130:1412–20. doi: 10.1053/j.gastro.2005.08.061. [DOI] [PubMed] [Google Scholar]
  • 28.Grundy D, Al-Chaer ED, Aziz Q, Collins SM, Ke M, Taché Y, Wood JD. Fundamentals of neurogastroenterology: basic science. Gastroenterology. 2006;130:1391–411. doi: 10.1053/j.gastro.2005.11.060. [DOI] [PubMed] [Google Scholar]
  • 29.Feng B, La JH, Schwartz ES, Gebhart GF. Irritable bowel syndrome: methods, mechanisms, and pathophysiology. Neural and neuro-immune mechanisms of visceral hypersensitivity in irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2012;302:G1085–98. doi: 10.1152/ajpgi.00542.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hertig VL, Cain KC, Jarrett ME, Burr RL, Heitkemper MM. Daily stress and gastrointestinal symptoms in women with irritable bowel syndrome. Nurs Res. 2007;56:399–406. doi: 10.1097/01.NNR.0000299855.60053.88. [DOI] [PubMed] [Google Scholar]
  • 31.Chaudhary NA, Truelove SC. Human colonic motility. A comparative study of normal subjects, patients with ulcerative colitis, and patients with the irritable colon syndrome. Gastroenterology. 1968;54:777–8. [PubMed] [Google Scholar]
  • 32.Spiller RC, Jenkins D, Thornley JP, Hebden JM, Wright T, Skinner M, Neal KR. Increased rectal mucosal enteroendocrine cells, T lymphocytes, and increased gut permeability following acute Campylobacter enteritis and in post-dysenteric irritable bowel syndrome. Gut. 2000;47:804–11. doi: 10.1136/gut.47.6.804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Marshall JK, Thabane M, Garg AX, Clark W, Meddings J, Collins SM, WEL Investigators Intestinal permeability in patients with irritable bowel syndrome after a waterborne outbreak of acute gastroenteritis in Walkerton, Ontario. Aliment Pharmacol Ther. 2004;20:1317–22. doi: 10.1111/j.1365-2036.2004.02284.x. [DOI] [PubMed] [Google Scholar]
  • 34.Villani AC, Lemire M, Thabane M, Belisle A, Geneau G, Garg AX, Clark WF, Moayyedi P, Collins SM, Franchimont D, et al. Genetic risk factors for post-infectious irritable bowel syndrome following a waterborne outbreak of gastroenteritis. Gastroenterology. 2010;138:1502–13. doi: 10.1053/j.gastro.2009.12.049. [DOI] [PubMed] [Google Scholar]
  • 35.Spiller R, Garsed K. Postinfectious irritable bowel syndrome. Gastroenterology. 2009;136:1979–88. doi: 10.1053/j.gastro.2009.02.074. [DOI] [PubMed] [Google Scholar]
  • 36.Jalanka-Tuovinen J, Salojärvi J, Salonen A, Immonen O, Garsed K, Kelly FM, Zaitoun A, Palva A, Spiller RC, de Vos WM. Faecal microbiota composition and host-microbe cross-talk following gastroenteritis and in postinfectious irritable bowel syndrome. Gut. 2013 doi: 10.1136/gutjnl-2013-305994. In press. [DOI] [PubMed] [Google Scholar]
  • 37.Schoepfer AM, Schaffer T, Seibold-Schmid B, Müller S, Seibold F. Antibodies to flagellin indicate reactivity to bacterial antigens in IBS patients. Neurogastroenterol Motil. 2008;20:1110–8. doi: 10.1111/j.1365-2982.2008.01166.x. [DOI] [PubMed] [Google Scholar]
  • 38.Matsunami M, Tarui T, Mitani K, Nagasawa K, Fukushima O, Okubo K, Yoshida S, Takemura M, Kawabata A. Luminal hydrogen sulfide plays a pronociceptive role in mouse colon. Gut. 2009;58:751–61. doi: 10.1136/gut.2007.144543. [DOI] [PubMed] [Google Scholar]
  • 39.King TS, Elia M, Hunter JO. Abnormal colonic fermentation in irritable bowel syndrome. Lancet. 1998;352:1187–9. doi: 10.1016/S0140-6736(98)02146-1. [DOI] [PubMed] [Google Scholar]
  • 40.Lin HC. Small intestinal bacterial overgrowth: a framework for understanding irritable bowel syndrome. JAMA. 2004;292:852–8. doi: 10.1001/jama.292.7.852. [DOI] [PubMed] [Google Scholar]
  • 41.Pimentel M, Chow EJ, Lin HC. Eradication of small intestinal bacterial overgrowth reduces symptoms of irritable bowel syndrome. Am J Gastroenterol. 2000;95:3503–6. doi: 10.1111/j.1572-0241.2000.03368.x. [DOI] [PubMed] [Google Scholar]
  • 42.Pimentel M, Chatterjee S, Chow EJ, Park S, Kong Y. Neomycin improves constipation-predominant irritable bowel syndrome in a fashion that is dependent on the presence of methane gas: subanalysis of a double-blind randomized controlled study. Dig Dis Sci. 2006;51:1297–301. doi: 10.1007/s10620-006-9104-6. [DOI] [PubMed] [Google Scholar]
  • 43.Pimentel M, Chow EJ, Lin HC. Normalization of lactulose breath testing correlates with symptom improvement in irritable bowel syndrome. a double-blind, randomized, placebo-controlled study. Am J Gastroenterol. 2003;98:412–9. doi: 10.1111/j.1572-0241.2003.07234.x. [DOI] [PubMed] [Google Scholar]
  • 44.Ward KM, Rounthwaite FJ. Neomycin ototoxicity. Ann Otol Rhinol Laryngol. 1978;87:211–5. doi: 10.1177/000348947808700211. [DOI] [PubMed] [Google Scholar]
  • 45.Yang J, Lee HR, Low K, Chatterjee S, Pimentel M. Rifaximin versus other antibiotics in the primary treatment and retreatment of bacterial overgrowth in IBS. Dig Dis Sci. 2008;53:169–74. doi: 10.1007/s10620-007-9839-8. [DOI] [PubMed] [Google Scholar]
  • 46.Pimentel M, Lembo A, Chey WD, Zakko S, Ringel Y, Yu J, Mareya SM, Shaw AL, Bortey E, Forbes WP, TARGET Study Group Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med. 2011;364:22–32. doi: 10.1056/NEJMoa1004409. [DOI] [PubMed] [Google Scholar]
  • 47.Meyrat P, Safroneeva E, Schoepfer AM. Rifaximin treatment for the irritable bowel syndrome with a positive lactulose hydrogen breath test improves symptoms for at least 3 months. Aliment Pharmacol Ther. 2012;36:1084–93. doi: 10.1111/apt.12087. [DOI] [PubMed] [Google Scholar]
  • 48.Tana C, Umesaki Y, Imaoka A, Handa T, Kanazawa M, Fukudo S. Altered profiles of intestinal microbiota and organic acids may be the origin of symptoms in irritable bowel syndrome. Neurogastroenterol Motil. 2010;22:512–9, e114-5. doi: 10.1111/j.1365-2982.2009.01427.x. [DOI] [PubMed] [Google Scholar]
  • 49.Treem WR, Ahsan N, Kastoff G, Hyams JS. Fecal short-chain fatty acids in patients with diarrhea-predominant irritable bowel syndrome: in vitro studies of carbohydrate fermentation. J Pediatr Gastroenterol Nutr. 1996;23:280–6. doi: 10.1097/00005176-199610000-00013. [DOI] [PubMed] [Google Scholar]
  • 50.Vanhoutvin SA, Troost FJ, Kilkens TO, Lindsey PJ, Hamer HM, Jonkers DM, Venema K, Brummer RJ. The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol Motil. 2009;21:952–e76. doi: 10.1111/j.1365-2982.2009.01324.x. [DOI] [PubMed] [Google Scholar]
  • 51.Koide A, Yamaguchi T, Odaka T, Koyama H, Tsuyuguchi T, Kitahara H, Ohto M, Saisho H. Quantitative analysis of bowel gas using plain abdominal radiograph in patients with irritable bowel syndrome. Am J Gastroenterol. 2000;95:1735–41. doi: 10.1111/j.1572-0241.2000.02189.x. [DOI] [PubMed] [Google Scholar]
  • 52.Crouzet L, Gaultier E, Del’Homme C, Cartier C, Delmas E, Dapoigny M, Fioramonti J, Bernalier-Donadille A. The hypersensitivity to colonic distension of IBS patients can be transferred to rats through their fecal microbiota. Neurogastroenterol Motil. 2013;25:e272–82. doi: 10.1111/nmo.12103. [DOI] [PubMed] [Google Scholar]
  • 53.Ford AC, Talley NJ. Mucosal inflammation as a potential etiological factor in irritable bowel syndrome: a systematic review. J Gastroenterol. 2011;46:421–31. doi: 10.1007/s00535-011-0379-9. [DOI] [PubMed] [Google Scholar]
  • 54.Cervero F, Jänig W. Visceral nociceptors: a new world order? Trends Neurosci. 1992;15:374–8. doi: 10.1016/0166-2236(92)90182-8. [DOI] [PubMed] [Google Scholar]
  • 55.Verdú EF, Bercik P, Verma-Gandhu M, Huang XX, Blennerhassett P, Jackson W, Mao Y, Wang L, Rochat F, Collins SM. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut. 2006;55:182–90. doi: 10.1136/gut.2005.066100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.O’Sullivan M, Clayton N, Breslin NP, Harman I, Bountra C, McLaren A, O’Morain CA. Increased mast cells in the irritable bowel syndrome. Neurogastroenterol Motil. 2000;12:449–57. doi: 10.1046/j.1365-2982.2000.00221.x. [DOI] [PubMed] [Google Scholar]
  • 57.Weston AP, Biddle WL, Bhatia PS, Miner PB., Jr. Terminal ileal mucosal mast cells in irritable bowel syndrome. Dig Dis Sci. 1993;38:1590–5. doi: 10.1007/BF01303164. [DOI] [PubMed] [Google Scholar]
  • 58.Langhorst J, Junge A, Rueffer A, Wehkamp J, Foell D, Michalsen A, Musial F, Dobos GJ. Elevated human beta-defensin-2 levels indicate an activation of the innate immune system in patients with irritable bowel syndrome. Am J Gastroenterol. 2009;104:404–10. doi: 10.1038/ajg.2008.86. [DOI] [PubMed] [Google Scholar]
  • 59.Collins SM. The immunomodulation of enteric neuromuscular function: implications for motility and inflammatory disorders. Gastroenterology. 1996;111:1683–99. doi: 10.1016/S0016-5085(96)70034-3. [DOI] [PubMed] [Google Scholar]
  • 60.Barbara G, Stanghellini V, De Giorgio R, Cremon C, Cottrell GS, Santini D, Pasquinelli G, Morselli-Labate AM, Grady EF, Bunnett NW, et al. Activated mast cells in proximity to colonic nerves correlate with abdominal pain in irritable bowel syndrome. Gastroenterology. 2004;126:693–702. doi: 10.1053/j.gastro.2003.11.055. [DOI] [PubMed] [Google Scholar]
  • 61.Buhner S, Li Q, Vignali S, Barbara G, De Giorgio R, Stanghellini V, Cremon C, Zeller F, Langer R, Daniel H, et al. Activation of human enteric neurons by supernatants of colonic biopsy specimens from patients with irritable bowel syndrome. Gastroenterology. 2009;137:1425–34. doi: 10.1053/j.gastro.2009.07.005. [DOI] [PubMed] [Google Scholar]
  • 62.Macfarlane GT, Allison C, Gibson SA, Cummings JH. Contribution of the microflora to proteolysis in the human large intestine. J Appl Bacteriol. 1988;64:37–46. doi: 10.1111/j.1365-2672.1988.tb02427.x. [DOI] [PubMed] [Google Scholar]
  • 63.Róka R, Rosztóczy A, Leveque M, Izbéki F, Nagy F, Molnár T, Lonovics J, Garcia-Villar R, Fioramonti J, Wittmann T, et al. A pilot study of fecal serine-protease activity: a pathophysiologic factor in diarrhea-predominant irritable bowel syndrome. Clin Gastroenterol Hepatol. 2007;5:550–5. doi: 10.1016/j.cgh.2006.12.004. [DOI] [PubMed] [Google Scholar]
  • 64.Gecse K, Róka R, Ferrier L, Leveque M, Eutamene H, Cartier C, Ait-Belgnaoui A, Rosztóczy A, Izbéki F, Fioramonti J, et al. Increased faecal serine protease activity in diarrhoeic IBS patients: a colonic lumenal factor impairing colonic permeability and sensitivity. Gut. 2008;57:591–9. doi: 10.1136/gut.2007.140210. [DOI] [PubMed] [Google Scholar]
  • 65.Annaházi A, Ferrier L, Bézirard V, Lévêque M, Eutamène H, Ait-Belgnaoui A, Coëffier M, Ducrotté P, Róka R, Inczefi O, et al. Luminal cysteine-proteases degrade colonic tight junction structure and are responsible for abdominal pain in constipation-predominant IBS. Am J Gastroenterol. 2013;108:1322–31. doi: 10.1038/ajg.2013.152. [DOI] [PubMed] [Google Scholar]
  • 66.Piche T, Barbara G, Aubert P, Bruley des Varannes S, Dainese R, Nano JL, Cremon C, Stanghellini V, De Giorgio R, Galmiche JP, et al. Impaired intestinal barrier integrity in the colon of patients with irritable bowel syndrome: involvement of soluble mediators. Gut. 2009;58:196–201. doi: 10.1136/gut.2007.140806. [DOI] [PubMed] [Google Scholar]
  • 67.Zhou Q, Zhang B, Verne GN. Intestinal membrane permeability and hypersensitivity in the irritable bowel syndrome. Pain. 2009;146:41–6. doi: 10.1016/j.pain.2009.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Martínez C, Lobo B, Pigrau M, Ramos L, González-Castro AM, Alonso C, Guilarte M, Guilá M, de Torres I, Azpiroz F, et al. Diarrhoea-predominant irritable bowel syndrome: an organic disorder with structural abnormalities in the jejunal epithelial barrier. Gut. 2013;62:1160–8. doi: 10.1136/gutjnl-2012-302093. [DOI] [PubMed] [Google Scholar]
  • 69.Martínez C, Vicario M, Ramos L, Lobo B, Mosquera JL, Alonso C, Sánchez A, Guilarte M, Antolín M, de Torres I, et al. The jejunum of diarrhea-predominant irritable bowel syndrome shows molecular alterations in the tight junction signaling pathway that are associated with mucosal pathobiology and clinical manifestations. Am J Gastroenterol. 2012;107:736–46. doi: 10.1038/ajg.2011.472. [DOI] [PubMed] [Google Scholar]
  • 70.Ait-Belgnaoui A, Bradesi S, Fioramonti J, Theodorou V, Bueno L. Acute stress-induced hypersensitivity to colonic distension depends upon increase in paracellular permeability: role of myosin light chain kinase. Pain. 2005;113:141–7. doi: 10.1016/j.pain.2004.10.002. [DOI] [PubMed] [Google Scholar]
  • 71.FAO/WHO Working Group Guidelines for the evaluation of probiotics in food. London, Ontario, Canada Abvailable at ftp://ftp.fao.org/es/esn/food/wgreport2.pdf
  • 72.Hungin AP, Mulligan C, Pot B, Whorwell P, Agréus L, Fracasso P, Lionis C, Mendive J, Philippart de Foy JM, Rubin G, et al. European Society for Primary Care Gastroenterology. Systematic review: probiotics in the management of lower gastrointestinal symptoms in clinical practice–an evidence-based international guide. Aliment Pharmacol Ther. 2013;38:864–86. doi: 10.1111/apt.12460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Vahabnezhad E, Mochon AB, Wozniak LJ, Ziring DA. Lactobacillus bacteremia associated with probiotic use in a pediatric patient with ulcerative colitis. J Clin Gastroenterol. 2013;47:437–9. doi: 10.1097/MCG.0b013e318279abf0. [DOI] [PubMed] [Google Scholar]
  • 74.Theodorakopoulou M, Perros E, Giamarellos-Bourboulis EJ, Dimopoulos G. Controversies in the management of the critically ill: the role of probiotics. Int J Antimicrob Agents. 2013;42(Suppl):S41–4. doi: 10.1016/j.ijantimicag.2013.04.010. [DOI] [PubMed] [Google Scholar]
  • 75.Morrow LE, Gogineni V, Malesker MA. Probiotic, prebiotic, and synbiotic use in critically ill patients. Curr Opin Crit Care. 2012;18:186–91. doi: 10.1097/MCC.0b013e3283514b17. [DOI] [PubMed] [Google Scholar]
  • 76.Kamiya T, Wang L, Forsythe P, Goettsche G, Mao Y, Wang Y, Tougas G, Bienenstock J. Inhibitory effects of Lactobacillus reuteri on visceral pain induced by colorectal distension in Sprague-Dawley rats. Gut. 2006;55:191–6. doi: 10.1136/gut.2005.070987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rousseaux C, Thuru X, Gelot A, Barnich N, Neut C, Dubuquoy L, Dubuquoy C, Merour E, Geboes K, Chamaillard M, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med. 2007;13:35–7. doi: 10.1038/nm1521. [DOI] [PubMed] [Google Scholar]
  • 78.Larauche M, Mulak A, Taché Y. Stress-related alterations of visceral sensation: animal models for irritable bowel syndrome study. J Neurogastroenterol Motil. 2011;17:213–34. doi: 10.5056/jnm.2011.17.3.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gareau MG, Silva MA, Perdue MH. Pathophysiological mechanisms of stress-induced intestinal damage. Curr Mol Med. 2008;8:274–81. doi: 10.2174/156652408784533760. [DOI] [PubMed] [Google Scholar]
  • 80.Zareie M, Johnson-Henry K, Jury J, Yang PC, Ngan BY, McKay DM, Soderholm JD, Perdue MH, Sherman PM. Probiotics prevent bacterial translocation and improve intestinal barrier function in rats following chronic psychological stress. Gut. 2006;55:1553–60. doi: 10.1136/gut.2005.080739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gareau MG, Jury J, MacQueen G, Sherman PM, Perdue MH. Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut. 2007;56:1522–8. doi: 10.1136/gut.2006.117176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Ait-Belgnaoui A, Han W, Lamine F, Eutamene H, Fioramonti J, Bueno L, Theodorou V. Lactobacillus farciminis treatment suppresses stress induced visceral hypersensitivity: a possible action through interaction with epithelial cell cytoskeleton contraction. Gut. 2006;55:1090–4. doi: 10.1136/gut.2005.084194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ait-Belgnaoui A, Eutamene H, Houdeau E, Bueno L, Fioramonti J, Theodorou V. Lactobacillus farciminis treatment attenuates stress-induced overexpression of Fos protein in spinal and supraspinal sites after colorectal distension in rats. Neurogastroenterol Motil. 2009;21:567–73, e18-9. doi: 10.1111/j.1365-2982.2009.01280.x. [DOI] [PubMed] [Google Scholar]
  • 84.Eutamene H, Lamine F, Chabo C, Theodorou V, Rochat F, Bergonzelli GE, Corthésy-Theulaz I, Fioramonti J, Bueno L. Synergy between Lactobacillus paracasei and its bacterial products to counteract stress-induced gut permeability and sensitivity increase in rats. J Nutr. 2007;137:1901–7. doi: 10.1093/jn/137.8.1901. [DOI] [PubMed] [Google Scholar]
  • 85.McKernan DP, Fitzgerald P, Dinan TG, Cryan JF. The probiotic Bifidobacterium infantis 35624 displays visceral antinociceptive effects in the rat. Neurogastroenterol Motil. 2010;22:1029–35, e268. doi: 10.1111/j.1365-2982.2010.01520.x. [DOI] [PubMed] [Google Scholar]
  • 86.O’Mahony L, McCarthy J, Kelly P, Hurley G, Luo F, Chen K, O’Sullivan GC, Kiely B, Collins JK, Shanahan F, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology. 2005;128:541–51. doi: 10.1053/j.gastro.2004.11.050. [DOI] [PubMed] [Google Scholar]
  • 87.Agostini S, Goubern M, Tondereau V, Salvador-Cartier C, Bezirard V, Lévèque M, Keränen H, Theodorou V, Bourdu-Naturel S, Goupil-Feuillerat N, et al. A marketed fermented dairy product containing Bifidobacterium lactis CNCM I-2494 suppresses gut hypersensitivity and colonic barrier disruption induced by acute stress in rats. Neurogastroenterol Motil. 2012;24:376–e172. doi: 10.1111/j.1365-2982.2011.01865.x. [DOI] [PubMed] [Google Scholar]

Articles from Gut Microbes are provided here courtesy of Taylor & Francis

RESOURCES