Skip to main content
Gut Microbes logoLink to Gut Microbes
. 2014 Jun 12;5(3):419–429. doi: 10.4161/gmic.29417

The microbiota-gut-brain axis in functional gastrointestinal disorders

Giada De Palma 1, Stephen M Collins 1, Premysl Bercik 1,*
PMCID: PMC4153782  PMID: 24921926

Abstract

Functional gastrointestinal disorders (FGIDs) are highly prevalent and pose a significant burden on health care and society, and impact patients’ quality of life. FGIDs comprise a heterogeneous group of disorders, with unclear underlying pathophysiology. They are considered to result from the interaction of altered gut physiology and psychological factors via the gut-brain axis, where brain and gut symptoms are reciprocally influencing each other’s expression. Intestinal microbiota, as a part of the gut-brain axis, plays a central role in FGIDs. Patients with Irritable Bowel Syndrome, a prototype of FGIDs, display altered composition of the gut microbiota compared with healthy controls and benefit, at the gastrointestinal and psychological levels, from the use of probiotics and antibiotics. This review aims to recapitulate the available literature on FGIDs and microbiota-gut-brain axis.

Keywords: functional gastrointestinal disorders, IBS, gut-brain axis, microbiota, anxiety, depression

Functional Gastrointestinal Disorders

For the last few decades physicians have struggled to understand functional gastrointestinal disorders that escape the objective diagnosis of organic pathology and are characterized by non-structural symptoms that undermine patients’ quality of life. The following statement describes a functional gastrointestinal (GI) disorder: There is no evidence of an inflammatory, anatomic, metabolic, or neoplastic process that explains the patient’s symptoms.1 FGIDs rise from the combination of genetic2 and environmental factors, including exposure to infections, use of antibiotics, sexual or physical abuse, and also family influences on illness expression, that synergistically shape one’s psychological development and susceptibility to gut dysfunctions.1,3-6 Therefore a FGID is the clinical product of the interaction of altered gut physiology and psychological factors via the gut-brain axis, where brain and gut symptoms are reciprocally influencing each other’s expression. The communication between the “GI brain” (the enteric-nervous system; ENS), and the central nervous system (CNS) is key in the pathophysiology of FGIDs.7 The latest FGIDs classification is the Rome III criteria system, which groups FGIDs into 6 categories: esophageal, gastroduodenal, bowel, functional abdominal pain syndrome, biliary, and anorectal.1 In order to ascribe reported gut symptoms to FGID, the symptoms must have occurred for the first time ≥6 months before the patient presents to the physician practice and their presence had to be ≥3 days a month during the last 3 months.8 However the adoption of Rome III criteria is still matter of debate due to inadequate validation of the criteria and consequent low utilization,9 but also due to low sensitivity of the criteria to diagnose FGIDs and in particular IBS.10-12 FGIDs include irritable bowel syndrome (IBS), functional dyspepsia (FD), functional bloating, functional constipation, and functional diarrhea.1,13 For purposes of simplification this review will focus on IBS and FD.

IBS is the most common functional bowel disorder worldwide that affects between 7 to 10% of population.14 Its prevalence varies across the world according the diagnostic criteria (Manning, Rome II, Rome III, self-diagnosed), the population selected, the access to health care, and culture.15 IBS is one of the most common reasons of healthcare seeking with significant impacts on health care expenses,4,16 and the most studied FGID.15,17 IBS classifies into 4 different categories according to bowel habits and stool form using the Bristol Stool Scale:8 IBS-constipation (IBS-C), IBS-diarrhea (IBS-D), Mixed IBS, and Unsubtyped IBS.8 IBS affects patients across the lifespan but there is an overall strong female predominance.15 Men are more likely to suffer from IBS-D while women from IBS-C.18,19 Moreover, sex hormones are likely to affect GI function and the severity of IBS symptoms.19 IBS has been associated with abnormal gut motor function, enhanced visceral perception, abnormalities in central pain processing, and altered gut microbiota, besides psychosocial and genetic factors.

FD is the second most common FGID with a great impact on the quality of life of the patients,20,21 although it often remains unreported to physicians. Pathophysiological mechanisms underlying FD include delayed gastric emptying,13,20,22 impaired gastric accommodation to a meal, visceral hypersensitivity, and duodenal sensitivity to acids,20,23 as well as psychosocial and genetic factors.20

The Role of the CNS in Functional GI Disorders

Psychological and psychosocial factors are important in the understanding of the pathophysiology of FGIDs. Psychiatric disorders such as anxiety, depressive disorder, and neuroticism, are common comorbidities in patients with FGIDs.24-33 However, it is unclear whether the brain abnormalities drive the gut symptoms or the changes in the gut alter brain function through vagal and sympathetic afferents. A recent 12-year prospective study aimed at determining the role of the brain-gut mechanism in IBS and FD and concluded that the brain-gut pathway is bidirectional, as brain-gut and gut-brain dysfunctions both occur in FGIDs.34 FGID patients are also characterized by abnormalities in autonomic nervous system, neuroendocrine and immune functions, which are influenced by psychological distress35 in the model of emotional motor system (EMS), which reacts to interoceptive and exteroceptive stress. Specific brain structures involved in the EMS, including the anterior cingulate cortex (ACC), amygdala, hippocampus, hypothalamus, and periaqueductal gray, communicate to the gastrointestinal tract through the hypothalamus-pituitary-adrenal (HPA) axis, autonomic nervous system, the endogenous pain modulation system, and ascending aminergic pathways.36

An important player in the EMS is corticotrophin-releasing hormone (CRH) located in effector neurons of the paraventricular nucleus (PVN) of the hypothalamus, the amygdala, and the locus coeruleus complex that activate both the autonomic nervous system and HPA axis.36 Activation of the HPA axis followed by secretion of corticosteroid hormones from the adrenal cortex (i.e., cortisol in humans and corticosterone in rodents) is considered a physiological response to stress.37,38 Patients with FGID display dysregulation of the HPA-axis response to stress and changes in free cortisol secretion, which correlate with the gastrointestinal symptoms.39 Moreover, in response to a visceral stressor, IBS patients’ basal cortisol levels positively correlate with anxiety symptoms scores.40,41 HPA axis alterations and stress have also been related to abnormalities in gut motor function.42 Indeed, psychological stress appears to be a sensitive and specific predictor of symptoms in FD patients.31,43

Studies in animal models have shown that acute stress alters intestinal permeability through mechanisms involving CRH,44 while chronic stress induces low-grade inflammation and can lead to visceral hyperalgesia.45 Enhanced stress responsiveness has been implicated as a potential mechanism contributing to the pathophysiology of IBS, as stress reactivates previous enteric inflammation and enhances the response to subsequent inflammatory stimuli.46 Early life stress can permanently affect the development of the HPA-axis, contributing to altered visceral pain modulation, and behavioral changes associated with stress-related disorders.47 Corticotrophin-releasing factor (CRF) has thus been proposed as a possible mediator in IBS, as central CRF administration mimics acute stress-induced colonic responses and enhances colorectal distension-induced visceral pain, whereas peripheral CRF alters neuromotor gut function.48-50

Neuroimaging research has allowed for the investigation of underlying mechanisms of altered visceral perception in patients with IBS. Abnormal brain activation in response to visceral stimuli and dysregulation of the CNS has been found in FGIDs patients compared with healthy controls.51-54 However, it is unclear whether the reported abdominal pain reflects an abnormal afferent input to the brain, or central alterations in the signals from the gut or both. IBS patients have greater engagement of regions associated with emotional arousal and endogenous pain modulation, but similar activation of regions involved in processing of visceral afferent information, whereas controls have greater engagement of cognitive modulatory regions.52 Another study showed that upregulated emotional arousal circuitry and altered serotonergic modulation of this circuitry may play a role in centrally mediated visceral hypersensitivity in female patients with IBS.52,55 Indeed, these patients seem to present altered engagement of descending pain modulation systems that increases the excitability of the dorsal horn resulting in increased ascending input to brain regions processing interoceptive input.56 Inhibition of neurokinin-1 receptor, which is involved in augmented nociceptive response and behavioral and autonomic responses to stress, reduced central pain amplification during an acute experimental stimulus in women with IBS.57 A recent study has suggested that changes in gray matter density in regions involved in cognitive and/or evaluative functions are specifically observed in patients with IBS, whereas changes in other brain areas are associated with levels of anxiety and depression.58 These functional and gray matter abnormalities in IBS patients are also accompanied by white matter changes, which are possibly responsible for the emotional aspect of pain in IBS.59 Similarly, abnormalities in brain activity in response to visceral stimuli as well as during the resting state have been reported in patients with FD,60-65 and very recently abnormalities in white matter microstructure have been reported in patients with FD.66

Summary: FGIDs patients present with abnormalities in visceral perception, neuroendocrine and immune functions, which are influenced by psychological distress. Abnormal brain activation in response to visceral stimuli, or altered engagement of descending and ascending pathways, have been implicated in the pathophysiology of FGIDs,

The Gut Microbiota

The gut microbiota is a key player in determining gut health and function.67 The gut microbiota is composed mainly by bacteria but also by archaea, viruses, and protozoa that roughly reach 1014 cells, outnumbering the human cells in our bodies by a factor of ten.68 The human gut is rapidly colonized at birth and this ecosystem is under constant evolution until adult-like communities stabilize. The microbiota undergoes selective pressure from the host as well as from microbial competitors and once the ecosystem reaches homeostasis, some species will occur in high and others in low abundance.69-71 However, out of the numerous phyla described in the literature only 19 are present in the human GI tract,72 and five of them are predominant (Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, and Actinobacteria).72 Three genera have been used to determine the main “enterotypes” under which humans can be categorized (Bacteroides, Prevotella, and Ruminococcus)73,74; however, this categorization has recently become a matter of debate and the term “enterogradients” has been proposed instead, to describe bacterial communities with prevalence of Bacteroides or Prevotella.75 These autochthonous genera stably colonize the gastrointestinal tract and are present in a majority of individuals. Even though the gut microbiota still differs greatly between subjects in membership and community structure, the microbiomes appear largely functionally equivalent and necessary for the proper development of the host. Known functions of the gut microbiota include the conversion of non-digestible carbohydrates (dietary fiber) to short-chain fatty acids (SCFAs), transformation of bile acids, the provision of a barrier against pathogenic bacteria, and modulation of the innate and the adaptive immune systems (for review see ref. 69).69 The importance of the gut microbiota is highlighted by the increasing number of studies performed in germ-free animals, which demonstrate physiologic and metabolic abnormalities compared with conventional animals. Indeed, germ-free mice have an immature and deregulated immune system,76-80 with abnormal IgA production81,82 and decreased numbers of intestinal mast cells.83 Germ-free mice have also impaired capacity for harvesting energy from the diet.84 The absence of microbiota protects against diet-induced obesity85,86 and excessive energy storage in the liver and in the skeletal muscle.86 Interestingly, transplanting the microbiota from obese mice or mice fed high-fat diet induce the same donor phenotype in germ-free recipients,87-89 meaning that the gut microbiota plays a role in obesity and weight gain. Several studies showed that germ-free animals have an enlarged cecum reflecting abnormal gut motility,84,90,91 increased expression of genes encoding transporters throughout the gut,70 as well as altered perception of inflammatory pain.92

Summary: Gut microbiota has evolved with its host and plays a pivotal role in host’s physiology and homeostasis.

Gut Microbiota and Functional Gastrointestinal Disease

Alterations in the gut microbiota composition have been well described in several functional gastrointestinal disorders and are reviewed exhaustively in a recent report by the Rome Working Team.93 Multiple studies have shown differences in the composition of the gut microbiota between IBS patients and healthy controls.94-115 However, the results of these studies are inconsistent and no unique IBS bacterial signature and/or profile has been identified, due in part to different detection methods as well as different patient populations.94,116 A pair of recent studies confirmed that IBS is associated with a decrease in the stability and biodiversity of the gut microbiota.117,118 However there is not a clear consensus on what constitutes a healthy microbiota.

Post-infectious (PI) FGID represent a category within the general FGID classification.119,120 The occurrence of infectious gastroenteritis has been well documented by several studies in both IBS and FD, showing that the risk to develop a functional disorder is greater in exposed individuals.119,121,122 Some studies also reported the incidence of FGIDs after a viral infection.123,124 The underlying mechanisms involved in PI-FGIDs are still to be fully elucidated, although several studies have shown evidences of low grade “immune activation” in IBS patients.125 It has been proposed that transient inflammation could lead to subtle but permanent changes in the structure and function of the digestive system, such as in lymphocytes, mast cells, enterochromaffin (EC) cells, and enteric nerves, which, in turn, induce the symptoms.120 The microbiota is deeply perturbed at the site of the infection126 and it might act synergistically with ongoing inflammation and increased epithelial permeability, increasing the sensitivity to develop a FGID in prone individuals.127

Small intestinal bacterial overgrowth (SIBO) is another condition that has been associated with IBS and that may be responsible for symptom generation in some patients with IBS. SIBO is defined as a quantitative alteration of the small intestinal microbiota.128 Its role in IBS is controversial, partly as the scientific community has not reached a consensus on the detection method to use: the breath tests are not well validated and the jejunal aspirates are not always accurate.129-132 Bacterial overgrowth results in unusual fermentation with increases in gas production, abdominal bloating, malabsorption, abdominal pain, diarrhea, and abnormal gastrointestinal motility.133-135 It remains unclear whether SIBO is actually fundamental to the pathophysiology of IBS, or is just a complicating phenomenon. However, several studies suggested that treatment of SIBO with non-absorbable antibiotics improves gut symptoms in a proportion of patients with IBS.136-138

Summary: Patients with IBS have different composition of the gut microbiota but no unique bacterial profile has been identified. It is unclear whether this dysbiosis is a cause or a consequence of gut dysfunction.

Microbiota-Gut-Brain Axis

There is growing evidence that there is a complex interaction between the host and specific bacterial species or their metabolites. Striking examples are found in nature: Toxoplasma gondii, an obligate intracellular protozoal parasite is able to convert the natural fear of its intermediate host mice against cat urine into attraction, facilitating the transmission of the parasite from mice to its specific host, the cat.139,140 It has been shown that tachyzoites and bradyzoites (cysts) of Toxoplasma gondii impair neuronal function in a mouse model.141 Another example comes from clinical practice: laxatives and oral antibiotics are used to treat patients with hepatic encephalopathy, a disorder that likely results from the systemic accumulation of gut-derived neurotoxins in patients with impaired liver function and portosystemic shunting.142,143 The use of different antibiotics, on the other hand, has been reported to induce acute psychosis with symptoms resolved after cessation of antibiotics.144,145 Although controversial, there is some evidence of abnormal microbiota composition and partial improvement in symptoms after treatment with antibiotics in patients with late onset autism (for review see ref. 146).146 An association between Major Depressive Disorder and altered gut microbiota has been also suggested as carbohydrate malabsorption has been linked with increased risk to develop mental depression.147-150 The scientific community has begun to accept the concept that gut microbiota is implicated in brain autoimmunity in Multiple Sclerosis (for review see ref. 151).151 The commensal intestinal bacteria appear to be essential in triggering immune processes, leading to a relapsing-remitting autoimmune disease.152 Antibiotic treatment effectively reduces the severity of the disease in mouse model of experimental autoimmune encephalomyelitis (EAE) and germ-free mice are more resistant than conventional mice to develop EAE.153,154

Germ-free mice display abnormalities within the CNS with a dysregulated HPA stress response,155 altered level of brain-derived neurotrophic factor in the hippocampus,155-158 reduced anxiety-like behavior,157,158 altered expression of genes known to be involved in second messenger pathways and synaptic long-term potentiation,157 and altered tryptophan availability and metabolism.156 Non-absorbable antibiotic treatment in conventional BALB/c mice induced changes in intestinal microbiota composition, with increased levels of Firmicutes, phylum dominated by Lactobacillus species but including also some species of sulfate-reducing bacteria (SRB), and decreased levels of γ-Proteobacteria and Bacteroidetes.159 These microbial alterations were accompanied by increased levels of hippocampal BDNF and an autonomic-independent anxiolytic behavior in mice.159

Summary: Accumulating data suggest that gut microbiota influences CNS function and host's behavior. Underlying mechanisms are unclear but likely involve immune, humoral, and neural pathways.

Microbiota-Gut-Brain Interactions in FGIDs

A recent study has shown that IBS-C patients have higher numbers of SRB than healthy controls.96 These bacteria use lactate and H2 as substrates for H2S production, reducing availability of lactate for butyrate and propionate producing bacteria and increasing the levels of H2S in the gut.160 Interestingly, luminal H2S and NaHS (an H2S donor) have been reported to play pronociceptive roles in mouse colon, through activation of T-type Ca2+ channels,161 but also antinociceptive roles in a rodent model of visceral pain.162 Bacterial secretion of H2S has been also shown to alter the effectiveness of many clinically used antibiotics.163 Thus, it appears that H2S might affect visceral perception in patients with FGIDs; however the literature is controversial, and further studies are warranted in order to clarify H2S’ role in visceral nociception and inflammation in FGIDs. Several specific bacterial probiotic strains have been shown to improve symptoms severity and abdominal pain in IBS patients,164-182 although their mechanism of action remains unclear. There are some species that in clinical trials appear to be more effective than others, such as Bifidobacterium species (B.infantis 3564 and B.bifidum MIMBb75)172,176,178 and Lactobacillus species (Lactobacillus acidophilus-SDC 2012, 2013, L. paracasei B2106, L. plantarum 299V, and L. rhamnosus GG).164,170,171,175,177 These probiotics appear to be effective in reducing abdominal pain and discomfort in adults and in children (L. rhamnosus GG). Several studies have also suggested that combinations of different probiotic strains, such as VSL3# or mixtures of Bifidobacterium and Lactobacillus species, are able to decrease abdominal pain and discomfort in patients with IBS.166,180-183

As discussed previously, gut bacteria have been shown to affect depression- and anxiety-like behavior in animal models. The first study to suggest psychological benefits of a probiotic supplementation in human involved 132 healthy adults; a subset of the individuals with depressive symptoms at baseline appeared to improve their mood after consuming a L. casei fermented product.184 However, administration of this probiotic seemed to worsen their cognitive performance. Another study in healthy adults found that the combined supplementation with L. helveticus R0052 and B. longum R0175 for 30 days decreased scores for anxiety, depression, and psychological distress.185 Moreover, the same group subsequently reported improved well-being (anxiety, depression, and somatization) in those individuals who had the lowest urinary free cortisol.186 A recent study using fMRI has demonstrated that administration of probiotic mixture, containing B. lactis can affect brain regions concerned with the central processing of afferent signals from the gut, and reduce the impact of the brain regions involved in emotional arousal on the central processing of gut afferent signals.187 However, these studies were performed in healthy volunteers and its relevance to disease remains to be demonstrated.

Summary: Probiotics are widely used in FGIDs patients, either as single species or their mixtures. Probiotics appear to improve gut symptoms, but also affect anxiety, depression, and psychological distress.

Microbiota-Gut-Brain Axis in Animal Models of FGIDs

Animal models of functional bowel disorders have been used extensively to study effects of probiotics. L paracasei NCC2461 was found to improve post-infective neuromuscular dysfunction in mice.188 B.infantis 35624 reduced visceral sensitivity to colorectal distension in rats,189,190 likely through improvement in tissue inflammation. A similar effect was reported with B. lactis CNCM I-2494,103 and VSL3#.162,191 In another study, L paracasei NCC2461 reduced visceral perception via reduction of MPO activity and substance P.192 In parallel, L. acidophilus was shown to induce analgesic receptors193 which could contribute to visceral pain reduction. Other studies have shown barrier enhancing effects of several probiotics associated with normalization of visceral pain perception.103,191,194 Thus, the beneficial effects of probiotics in animal models of IBS demonstrate a variety of mechanisms and targets that may be strain dependent.

Maternal separation in rodents is widely used as a model of early life stress that mimics some of the features of IBS.195-197 Maternally separated mice display long-lasting hyperactivity of the HPA-axis,196,198,199 anxiety-like behavior,198,200-202 visceral hypersensitivity,203-205 and altered cholinergic activity in the gut,196 accompanied by increased intestinal permeability.196,201,203,206 The behavioral and physiological changes induced by early life stress are accompanied by altered gut colonization,205 and the use of probiotics ameliorates the detrimental effects of stress.162,207-209 Furthermore, the effects of maternal separation on anxiety and depression are absent in mice raised in germ-free conditions,210 suggesting that intestinal microbiota plays an important role in this animal model of IBS.

Infection models are commonly exploited to study the mechanisms responsible for generation of FGID symptoms. Chronic H. pylori infection in mice alters the gastric motility and increases visceral sensitivity, leads to abnormal feeding behavior and altered expression of pro-inflammatory cytokine TNF-α in the hypothalamus and regulatory peptide propiomelanocortin (POMC) in the arcuate nucleus.211,212 Citrobacter rodentium infection in mice has been used to mimic PI-IBS following bout of gastroenteritis by E. coli. C. rodentium results in a self-limiting colitis that induces chronic hyperexcitability of colonic dorsal root ganglia (DRG) neurons and hyperalgesia, a dominant feature of PI-IBS in humans.213-215 Combined with stress, C. rodentium infection results in increased intestinal permeability,216 increased levels of epinephrine and corticosterone, exaggerated neuronal excitability, and visceral hyperalgesia and/or allodynia.217 The ability of C. rodentium to colonize the intestine is significantly enhanced by stressor-induced changes in the microbiota.213 The initial phase of infection with C. rodentium also coincides with the development of anxiety-like behavior and the activation of vagal sensory neurons.218 Similarly, early phase of infection with Campylobacter jejuni, known to cause most of the food-borne gastroenteritis in humans,219 has been reported to induce anxiety-like behavior in mice, in the absence of immune response.220 The infection with C. jejuni activates viscerosensory pathways involved in identification and response to internal challenges, noradrenergic neurons and serotonergic neurons in different portions of the brain.221,222 Moreover, it affects central viscerosensory pathways that interface with stress-related and “defensive” network nuclei in the hypothalamic PVN, the amygdala, and the bed nucleus of the stria terminalis (BST), previously established as nodal points for the integration of psychological or processive stress with behavioral responses to potential threats or threatening situations.223 The above described effects of early infection appear to be purely neural in origin, however changes in behavior and brain biochemistry have been also observed in models of chronic low-grade colitis. Chronic infection with a non-invasive parasite, Trichuris muris, and mild chemically-induced colitis, induce anxiety-like behavior in mice and decreased levels of hippocampal BDNF expression224,225 via immune mediated mechanisms, including pro-inflammatory cytokines and altered tryptophan/kynurenine metabolism. Interestingly, probiotic B. longum, but not L. rhamnosus, was able to revert the abnormal behavior in both studies in a vagal-dependent manner.224,225 A recent study in healthy mice has demonstrated that administration of probiotic L. rhamnosus decreases anxiety and depression-like behaviors and alters expression of GABAergic receptors in the CNS, and this effect was also dependent on the integrity of the vagal nerve.47 Similarly, treatment with a probiotic combination of L. rhamnosus R0011 and L. helveticus was able to revert the memory impairment, accompanied by decreased BDNF levels in the hippocampus and c-fos expression, induced by C. rodentium infection.226

Summary: Animal models have been widely exploited to study the role of bacteria in pathophysiology of FGIDs and beneficial effects of probiotics, and have demonstrated a variety of mechanisms and targets that may be strain dependent.

Conclusions

Despite growing research on the microbiota-gut-brain axis, our knowledge of underlying mechanisms remains rather limited. It is unclear which pathways are involved in the communication between the intestinal microbiota or specific bacterial strains, the gut, and the brain, both in health and disease. Accumulating data suggest that, in a significant percentage of patients, the microbiota plays an important role in the genesis and maintenance of FGIDs. Probiotic supplementation appears to be of therapeutic value, although the clinical data to date remain controversial.227-229 This may be due to heterogeneity in underlying pathophysiological mechanisms, as well as the use of multiple probiotic bacteria with divergent mechanisms of action, as described in animal models, and which may not directly apply to the human condition. Further research should address whether specific probiotic treatment should be tailored to a particular host’s microbiota and whether the administration of a single strain is more effective than strain combinations.

Disclosure of Potential Conflicts of Interest

No potential conflict of interest was disclosed.

Acknowledgments

P.B. and S.M.C. received grant support from CIHR and Nestle. G.D.P. is recipient of CIHR-CAG Postdoctoral Fellowship; P.B. is recipient of the HHS Research Early Career Award.

Glossary

Abbreviations:

GI

gastrointestinal

FGIDs

functional gastrointestinal disorders

IBS

irritable bowel syndrome, FD, functional dyspepsia

IBS-C

IBS-constipation

IBS-D

IBS-diarrhea

ENS

enteric nervous system

HPA-axis

hypothalamus-pituitary-adrenal axis

CRH

corticotrophin-releasing hormone

CRF

corticotrophin-releasing factor

SIBO

small intestinal bowel overgrowth

PI-IBS

post-infectious IBS

10.4161/gmic.29417

References

  • 1.Drossman DA. The functional gastrointestinal disorders and the Rome III process. Gastroenterology. 2006;130:1377–90. doi: 10.1053/j.gastro.2006.03.008. [DOI] [PubMed] [Google Scholar]
  • 2.D’Amato M. Genes and functional GI disorders: from casual to causal relationship. Neurogastroenterol Motil. 2013;25:638–49. doi: 10.1111/nmo.12173. [DOI] [PubMed] [Google Scholar]
  • 3.Drossman DA. Abuse, trauma, and GI illness: is there a link? Am J Gastroenterol. 2011;106:14–25. doi: 10.1038/ajg.2010.453. [DOI] [PubMed] [Google Scholar]
  • 4.Levy RL, Olden KW, Naliboff BD, Bradley LA, Francisconi C, Drossman DA, Creed F. Psychosocial aspects of the functional gastrointestinal disorders. Gastroenterology. 2006;130:1447–58. doi: 10.1053/j.gastro.2005.11.057. [DOI] [PubMed] [Google Scholar]
  • 5.Geeraerts B, Van Oudenhove L, Fischler B, Vandenberghe J, Caenepeel P, Janssens J, Tack J. Influence of abuse history on gastric sensorimotor function in functional dyspepsia. Neurogastroenterol Motil. 2009;21:33–41. doi: 10.1111/j.1365-2982.2008.01178.x. [DOI] [PubMed] [Google Scholar]
  • 6.Villarreal AA, Aberger FJ, Benrud R, Gundrum JD. Use of broad-spectrum antibiotics and the development of irritable bowel syndrome. WMJ. 2012;111:17–20. [PubMed] [Google Scholar]
  • 7.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]
  • 8.Longstreth GF, Thompson WG, Chey WD, Houghton LA, Mearin F, Spiller RC. Functional bowel disorders. Gastroenterology. 2006;130:1480–91. doi: 10.1053/j.gastro.2005.11.061. [DOI] [PubMed] [Google Scholar]
  • 9.Dang J, Ardila-Hani A, Amichai MM, Chua K, Pimentel M. Systematic review of diagnostic criteria for IBS demonstrates poor validity and utilization of Rome III. Neurogastroenterol Motil. 2012;24:853–e397. doi: 10.1111/j.1365-2982.2012.01943.x. [DOI] [PubMed] [Google Scholar]
  • 10.Ford AC, Bercik P, Morgan DG, Bolino C, Pintos-Sanchez MI, Moayyedi P. The Rome III criteria for the diagnosis of functional dyspepsia in secondary care are not superior to previous definitions. Gastroenterology. 2014;146:932–40, quiz e14-5. doi: 10.1053/j.gastro.2014.01.014. [DOI] [PubMed] [Google Scholar]
  • 11.Ford AC, Bercik P, Morgan DG, Bolino C, Pintos-Sanchez MI, Moayyedi P. Characteristics of functional bowel disorder patients: a cross-sectional survey using the Rome III criteria. Aliment Pharmacol Ther. 2014;39:312–21. doi: 10.1111/apt.12573. [DOI] [PubMed] [Google Scholar]
  • 12.Ford AC, Bercik P, Morgan DG, Bolino C, Pintos-Sanchez MI, Moayyedi P. Validation of the Rome III criteria for the diagnosis of irritable bowel syndrome in secondary care. Gastroenterology. 2013;145:1262–70, e1. doi: 10.1053/j.gastro.2013.08.048. [DOI] [PubMed] [Google Scholar]
  • 13.Tack J, Talley NJ, Camilleri M, Holtmann G, Hu P, Malagelada JR, Stanghellini V. Functional gastroduodenal disorders. Gastroenterology. 2006;130:1466–79. doi: 10.1053/j.gastro.2005.11.059. [DOI] [PubMed] [Google Scholar]
  • 14.Spiegel BM. The burden of IBS: looking at metrics. Curr Gastroenterol Rep. 2009;11:265–9. doi: 10.1007/s11894-009-0039-x. [DOI] [PubMed] [Google Scholar]
  • 15.Quigley EM, Abdel-Hamid H, Barbara G, Bhatia SJ, Boeckxstaens G, De Giorgio R, Delvaux M, Drossman DA, Foxx-Orenstein AE, Guarner F, et al. A global perspective on irritable bowel syndrome: a consensus statement of the World Gastroenterology Organisation Summit Task Force on irritable bowel syndrome. J Clin Gastroenterol. 2012;46:356–66. doi: 10.1097/MCG.0b013e318247157c. [DOI] [PubMed] [Google Scholar]
  • 16.Koloski NA, Talley NJ, Boyce PM. Epidemiology and health care seeking in the functional GI disorders: a population-based study. Am J Gastroenterol. 2002;97:2290–9. doi: 10.1111/j.1572-0241.2002.05783.x. [DOI] [PubMed] [Google Scholar]
  • 17.Drossman DA, Chang L, Bellamy N, Gallo-Torres HE, Lembo A, Mearin F, Norton NJ, Whorwell P. Severity in irritable bowel syndrome: a Rome Foundation Working Team report. Am J Gastroenterol. 2011;106:1749–59, quiz 1760. doi: 10.1038/ajg.2011.201. [DOI] [PubMed] [Google Scholar]
  • 18.Lovell RM, Ford AC. Effect of gender on prevalence of irritable bowel syndrome in the community: systematic review and meta-analysis. Am J Gastroenterol. 2012;107:991–1000. doi: 10.1038/ajg.2012.131. [DOI] [PubMed] [Google Scholar]
  • 19.Adeyemo MA, Spiegel BM, Chang L. Meta-analysis: do irritable bowel syndrome symptoms vary between men and women? Aliment Pharmacol Ther. 2010;32:738–55. doi: 10.1111/j.1365-2036.2010.04409.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Oustamanolakis P, Tack J. Dyspepsia: organic versus functional. J Clin Gastroenterol. 2012;46:175–90. doi: 10.1097/MCG.0b013e318241b335. [DOI] [PubMed] [Google Scholar]
  • 21.Aro P, Talley NJ, Agréus L, Johansson SE, Bolling-Sternevald E, Storskrubb T, Ronkainen J. Functional dyspepsia impairs quality of life in the adult population. Aliment Pharmacol Ther. 2011;33:1215–24. doi: 10.1111/j.1365-2036.2011.04640.x. [DOI] [PubMed] [Google Scholar]
  • 22.Sarnelli G, Caenepeel P, Geypens B, Janssens J, Tack J. Symptoms associated with impaired gastric emptying of solids and liquids in functional dyspepsia. Am J Gastroenterol. 2003;98:783–8. doi: 10.1111/j.1572-0241.2003.07389.x. [DOI] [PubMed] [Google Scholar]
  • 23.Tack J, Janssen P. Gastroduodenal motility. Curr Opin Gastroenterol. 2010;26:647–55. doi: 10.1097/MOG.0b013e32833ece1e. [DOI] [PubMed] [Google Scholar]
  • 24.Van Oudenhove L, Aziz Q. The role of psychosocial factors and psychiatric disorders in functional dyspepsia. Nat Rev Gastroenterol Hepatol. 2013;10:158–67. doi: 10.1038/nrgastro.2013.10. [DOI] [PubMed] [Google Scholar]
  • 25.Wu JC. Psychological Co-morbidity in Functional Gastrointestinal Disorders: Epidemiology, Mechanisms and Management. J Neurogastroenterol Motil. 2012;18:13–8. doi: 10.5056/jnm.2012.18.1.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Clauwaert N, Jones MP, Holvoet L, Vandenberghe J, Vos R, Tack J, Van Oudenhove L. Associations between gastric sensorimotor function, depression, somatization, and symptom-based subgroups in functional gastroduodenal disorders: are all symptoms equal? Neurogastroenterol Motil. 2012;24:1088–e565. doi: 10.1111/j.1365-2982.2012.01985.x. [DOI] [PubMed] [Google Scholar]
  • 27.Kindt S, Van Oudenhove L, Mispelon L, Caenepeel P, Arts J, Tack J. Longitudinal and cross-sectional factors associated with long-term clinical course in functional dyspepsia: a 5-year follow-up study. Am J Gastroenterol. 2011;106:340–8. doi: 10.1038/ajg.2010.406. [DOI] [PubMed] [Google Scholar]
  • 28.Tse AW, Lai LH, Lee CC, Tsoi KK, Wong VW, Chan Y, Sung JJ, Chan FK, Wu JC. Validation of Self-administrated Questionnaire for Psychiatric Disorders in Patients with Functional Dyspepsia. J Neurogastroenterol Motil. 2010;16:52–60. doi: 10.5056/jnm.2010.16.1.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Van Oudenhove L, Vandenberghe J, Geeraerts B, Vos R, Persoons P, Fischler B, Demyttenaere K, Tack J. Determinants of symptoms in functional dyspepsia: gastric sensorimotor function, psychosocial factors or somatisation? Gut. 2008;57:1666–73. doi: 10.1136/gut.2008.158162. [DOI] [PubMed] [Google Scholar]
  • 30.Karling P, Danielsson A, Adolfsson R, Norrback KF. No difference in symptoms of irritable bowel syndrome between healthy subjects and patients with recurrent depression in remission. Neurogastroenterol Motil. 2007;19:896–904. doi: 10.1111/j.1365-2982.2007.00967.x. [DOI] [PubMed] [Google Scholar]
  • 31.Locke GR, 3rd, Weaver AL, Melton LJ, 3rd, Talley NJ. Psychosocial factors are linked to functional gastrointestinal disorders: a population based nested case-control study. Am J Gastroenterol. 2004;99:350–7. doi: 10.1111/j.1572-0241.2004.04043.x. [DOI] [PubMed] [Google Scholar]
  • 32.Lydiard RB, Fossey MD, Marsh W, Ballenger JC. Prevalence of psychiatric disorders in patients with irritable bowel syndrome. Psychosomatics. 1993;34:229–34. doi: 10.1016/S0033-3182(93)71884-8. [DOI] [PubMed] [Google Scholar]
  • 33.Talley NJ, Fung LH, Gilligan IJ, McNeil D, Piper DW. Association of anxiety, neuroticism, and depression with dyspepsia of unknown cause. A case-control study. Gastroenterology. 1986;90:886–92. doi: 10.1016/0016-5085(86)90864-4. [DOI] [PubMed] [Google Scholar]
  • 34.Koloski NA, Jones M, Kalantar J, Weltman M, Zaguirre J, Talley NJ. The brain--gut pathway in functional gastrointestinal disorders is bidirectional: a 12-year prospective population-based study. Gut. 2012;61:1284–90. doi: 10.1136/gutjnl-2011-300474. [DOI] [PubMed] [Google Scholar]
  • 35.Stasi C, Rosselli M, Bellini M, Laffi G, Milani S. Altered neuro-endocrine-immune pathways in the irritable bowel syndrome: the top-down and the bottom-up model. J Gastroenterol. 2012;47:1177–85. doi: 10.1007/s00535-012-0627-7. [DOI] [PubMed] [Google Scholar]
  • 36.Mayer EA. The neurobiology of stress and gastrointestinal disease. Gut. 2000;47:861–9. doi: 10.1136/gut.47.6.861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Joëls M. Impact of glucocorticoids on brain function: relevance for mood disorders. Psychoneuroendocrinology. 2011;36:406–14. doi: 10.1016/j.psyneuen.2010.03.004. [DOI] [PubMed] [Google Scholar]
  • 38.Kirschbaum C, Hellhammer DH. Salivary cortisol in psychoneuroendocrine research: recent developments and applications. Psychoneuroendocrinology. 1994;19:313–33. doi: 10.1016/0306-4530(94)90013-2. [DOI] [PubMed] [Google Scholar]
  • 39.Ehlert U, Nater UM, Böhmelt A. High and low unstimulated salivary cortisol levels correspond to different symptoms of functional gastrointestinal disorders. J Psychosom Res. 2005;59:7–10. doi: 10.1016/j.jpsychores.2005.03.005. [DOI] [PubMed] [Google Scholar]
  • 40.Chang L, Sundaresh S, Elliott J, Anton PA, Baldi P, Licudine A, Mayer M, Vuong T, Hirano M, Naliboff BD, et al. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis in irritable bowel syndrome. Neurogastroenterol Motil. 2009;21:149–59. doi: 10.1111/j.1365-2982.2008.01171.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mawdsley JE, Rampton DS. Psychological stress in IBD: new insights into pathogenic and therapeutic implications. Gut. 2005;54:1481–91. doi: 10.1136/gut.2005.064261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Taché Y, Bonaz B. Corticotropin-releasing factor receptors and stress-related alterations of gut motor function. J Clin Invest. 2007;117:33–40. doi: 10.1172/JCI30085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.De la Roca-Chiapas JM, Solís-Ortiz S, Fajardo-Araujo M, Sosa M, Córdova-Fraga T, Rosa-Zarate A, JM DlR-C Stress profile, coping style, anxiety, depression, and gastric emptying as predictors of functional dyspepsia: a case-control study. J Psychosom Res. 2010;68:73–81. doi: 10.1016/j.jpsychores.2009.05.013. [DOI] [PubMed] [Google Scholar]
  • 44.Saunders PR, Santos J, Hanssen NP, Yates D, Groot JA, Perdue MH. Physical and psychological stress in rats enhances colonic epithelial permeability via peripheral CRH. Dig Dis Sci. 2002;47:208–15. doi: 10.1023/A:1013204612762. [DOI] [PubMed] [Google Scholar]
  • 45.Bradesi S, Schwetz I, Ennes HS, Lamy CM, Ohning G, Fanselow M, Pothoulakis C, McRoberts JA, Mayer EA. Repeated exposure to water avoidance stress in rats: a new model for sustained visceral hyperalgesia. Am J Physiol Gastrointest Liver Physiol. 2005;289:G42–53. doi: 10.1152/ajpgi.00500.2004. [DOI] [PubMed] [Google Scholar]
  • 46.Collins SM. Stress and the Gastrointestinal Tract IV. Modulation of intestinal inflammation by stress: basic mechanisms and clinical relevance. Am J Physiol Gastrointest Liver Physiol. 2001;280:G315–8. doi: 10.1152/ajpgi.2001.280.3.G315. [DOI] [PubMed] [Google Scholar]
  • 47.Bravo JA, Dinan TG, Cryan JF. Alterations in the central CRF system of two different rat models of comorbid depression and functional gastrointestinal disorders. Int J Neuropsychopharmacol. 2011;14:666–83. doi: 10.1017/S1461145710000994. [DOI] [PubMed] [Google Scholar]
  • 48.Park AJ, Collins J, Blennerhassett PA, Ghia JE, Verdu EF, Bercik P, Collins SM. Altered colonic function and microbiota profile in a mouse model of chronic depression. Neurogastroenterol Motil. 2013;25:733–e575. doi: 10.1111/nmo.12153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Martinez V, Taché Y. CRF1 receptors as a therapeutic target for irritable bowel syndrome. Curr Pharm Des. 2006;12:4071–88. doi: 10.2174/138161206778743637. [DOI] [PubMed] [Google Scholar]
  • 50.Sagami Y, Shimada Y, Tayama J, Nomura T, Satake M, Endo Y, Shoji T, Karahashi K, Hongo M, Fukudo S. Effect of a corticotropin releasing hormone receptor antagonist on colonic sensory and motor function in patients with irritable bowel syndrome. Gut. 2004;53:958–64. doi: 10.1136/gut.2003.018911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tillisch K, Labus JS. Advances in imaging the brain-gut axis: functional gastrointestinal disorders. Gastroenterology. 2011;140:407–11, e1. doi: 10.1053/j.gastro.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tillisch K, Mayer EA, Labus JS. Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome. Gastroenterology. 2011;140:91–100. doi: 10.1053/j.gastro.2010.07.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Van Oudenhove L, Coen SJ, Aziz Q. Functional brain imaging of gastrointestinal sensation in health and disease. World J Gastroenterol. 2007;13:3438–45. doi: 10.3748/wjg.v13.i25.3438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Derbyshire SW. A systematic review of neuroimaging data during visceral stimulation. Am J Gastroenterol. 2003;98:12–20. doi: 10.1111/j.1572-0241.2003.07168.x. [DOI] [PubMed] [Google Scholar]
  • 55.Labus JS, Mayer EA, Jarcho J, Kilpatrick LA, Kilkens TO, Evers EA, Backes WH, Brummer RJ, van Nieuwenhoven MA. Acute tryptophan depletion alters the effective connectivity of emotional arousal circuitry during visceral stimuli in healthy women. Gut. 2011;60:1196–203. doi: 10.1136/gut.2010.213447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Larsson MB, Tillisch K, Craig AD, Engström M, Labus J, Naliboff B, Lundberg P, Ström M, Mayer EA, Walter SA. Brain responses to visceral stimuli reflect visceral sensitivity thresholds in patients with irritable bowel syndrome. Gastroenterology. 2012;142:463–72, e3. doi: 10.1053/j.gastro.2011.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tillisch K, Labus J, Nam B, Bueller J, Smith S, Suyenobu B, Siffert J, McKelvy J, Naliboff B, Mayer E. Neurokinin-1-receptor antagonism decreases anxiety and emotional arousal circuit response to noxious visceral distension in women with irritable bowel syndrome: a pilot study. Aliment Pharmacol Ther. 2012;35:360–7. doi: 10.1111/j.1365-2036.2011.04958.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Seminowicz DA, Labus JS, Bueller JA, Tillisch K, Naliboff BD, Bushnell MC, Mayer EA. Regional gray matter density changes in brains of patients with irritable bowel syndrome. Gastroenterology. 2010;139:48–57, e2. doi: 10.1053/j.gastro.2010.03.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chen JY, Blankstein U, Diamant NE, Davis KD. White matter abnormalities in irritable bowel syndrome and relation to individual factors. Brain Res. 2011;1392:121–31. doi: 10.1016/j.brainres.2011.03.069. [DOI] [PubMed] [Google Scholar]
  • 60.Zhou G, Liu P, Wang J, Wen H, Zhu M, Zhao R, von Deneen KM, Zeng F, Liang F, Gong Q, et al. Fractional amplitude of low-frequency fluctuation changes in functional dyspepsia: a resting-state fMRI study. Magn Reson Imaging. 2013;31:996–1000. doi: 10.1016/j.mri.2013.03.019. [DOI] [PubMed] [Google Scholar]
  • 61.Zhou G, Liu P, Zeng F, Yuan K, Yu D, von Deneen KM, Liang F, Qin W, Tian J. Increased interhemispheric resting-state functional connectivity in functional dyspepsia: a pilot study. NMR Biomed. 2012;26:410–41. doi: 10.1002/nbm.2878. [DOI] [PubMed] [Google Scholar]
  • 62.Zeng F, Qin W, Liang F, Liu J, Tang Y, Liu X, Yuan K, Yu S, Song W, Liu M, et al. Abnormal resting brain activity in patients with functional dyspepsia is related to symptom severity. Gastroenterology. 2011;141:499–506. doi: 10.1053/j.gastro.2011.05.003. [DOI] [PubMed] [Google Scholar]
  • 63.Van Oudenhove L, Vandenberghe J, Dupont P, Geeraerts B, Vos R, Dirix S, Bormans G, Vanderghinste D, Van Laere K, Demyttenaere K, et al. Abnormal regional brain activity during rest and (anticipated) gastric distension in functional dyspepsia and the role of anxiety: a H(2)(15)O-PET study. Am J Gastroenterol. 2010;105:913–24. doi: 10.1038/ajg.2010.39. [DOI] [PubMed] [Google Scholar]
  • 64.Van Oudenhove L, Vandenberghe J, Dupont P, Geeraerts B, Vos R, Dirix S, Van Laere K, Bormans G, Vanderghinste D, Demyttenaere K, et al. Regional brain activity in functional dyspepsia: a H(2)(15)O-PET study on the role of gastric sensitivity and abuse history. Gastroenterology. 2010;139:36–47. doi: 10.1053/j.gastro.2010.04.015. [DOI] [PubMed] [Google Scholar]
  • 65.Vandenberghe J, Dupont P, Van Oudenhove L, Bormans G, Demyttenaere K, Fischler B, Geeraerts B, Janssens J, Tack J. Regional cerebral blood flow during gastric balloon distention in functional dyspepsia. Gastroenterology. 2007;132:1684–93. doi: 10.1053/j.gastro.2007.03.037. [DOI] [PubMed] [Google Scholar]
  • 66.Zhou G, Qin W, Zeng F, Liu P, Yang X, von Deneen KM, Gong Q, Liang F, Tian J. White-matter microstructural changes in functional dyspepsia: a diffusion tensor imaging study. Am J Gastroenterol. 2013;108:260–9. doi: 10.1038/ajg.2012.405. [DOI] [PubMed] [Google Scholar]
  • 67.Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol. 2013;14:685–90. doi: 10.1038/ni.2608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90:859–904. doi: 10.1152/physrev.00045.2009. [DOI] [PubMed] [Google Scholar]
  • 69.Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7. doi: 10.1126/science.1223813. [DOI] [PubMed] [Google Scholar]
  • 70.Bäckhed F. Programming of host metabolism by the gut microbiota. Ann Nutr Metab. 2011;58(Suppl 2):44–52. doi: 10.1159/000328042. [DOI] [PubMed] [Google Scholar]
  • 71.Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6:e280. doi: 10.1371/journal.pbio.0060280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Stearns JC, Lynch MD, Senadheera DB, Tenenbaum HC, Goldberg MB, Cvitkovitch DG, Croitoru K, Moreno-Hagelsieb G, Neufeld JD. Bacterial biogeography of the human digestive tract. Sci Rep. 2011;1:170. doi: 10.1038/srep00170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, et al. MetaHIT Consortium Enterotypes of the human gut microbiome. Nature. 2011;473:174–80. doi: 10.1038/nature09944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zoetendal EG, Rajilic-Stojanovic M, de Vos WM. High-throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut. 2008;57:1605–15. doi: 10.1136/gut.2007.133603. [DOI] [PubMed] [Google Scholar]
  • 75.Jeffery IB, Claesson MJ, O’Toole PW, Shanahan F. Categorization of the gut microbiota: enterotypes or gradients? Nat Rev Microbiol. 2012;10:591–2. doi: 10.1038/nrmicro2859. [DOI] [PubMed] [Google Scholar]
  • 76.Hansen CH, Nielsen DS, Kverka M, Zakostelska Z, Klimesova K, Hudcovic T, Tlaskalova-Hogenova H, Hansen AK. Patterns of early gut colonization shape future immune responses of the host. PLoS One. 2012;7:e34043. doi: 10.1371/journal.pone.0034043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, Glickman JN, Siebert R, Baron RM, Kasper DL, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science. 2012;336:489–93. doi: 10.1126/science.1219328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hapfelmeier S, Lawson MA, Slack E, Kirundi JK, Stoel M, Heikenwalder M, Cahenzli J, Velykoredko Y, Balmer ML, Endt K, et al. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science. 2010;328:1705–9. doi: 10.1126/science.1188454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Slack E, Hapfelmeier S, Stecher B, Velykoredko Y, Stoel M, Lawson MA, Geuking MB, Beutler B, Tedder TF, Hardt WD, et al. Innate and adaptive immunity cooperate flexibly to maintain host-microbiota mutualism. Science. 2009;325:617–20. doi: 10.1126/science.1172747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Geuking MB, Cahenzli J, Lawson MA, Ng DC, Slack E, Hapfelmeier S, McCoy KD, Macpherson AJ. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity. 2011;34:794–806. doi: 10.1016/j.immuni.2011.03.021. [DOI] [PubMed] [Google Scholar]
  • 81.Benveniste J, Lespinats G, Adam C, Salomon JC. Immunoglobulins in intact, immunized, and contaminated axenic mice: study of serum IgA. J Immunol. 1971;107:1647–55. [PubMed] [Google Scholar]
  • 82.Macpherson AJ, Geuking MB, McCoy KD. Homeland security: IgA immunity at the frontiers of the body. Trends Immunol. 2012;33:160–7. doi: 10.1016/j.it.2012.02.002. [DOI] [PubMed] [Google Scholar]
  • 83.Kunii J, Takahashi K, Kasakura K, Tsuda M, Nakano K, Hosono A, Kaminogawa S. Commensal bacteria promote migration of mast cells into the intestine. Immunobiology. 2011;216:692–7. doi: 10.1016/j.imbio.2010.10.007. [DOI] [PubMed] [Google Scholar]
  • 84.Wostmann BS. The germfree animal in nutritional studies. Annu Rev Nutr. 1981;1:257–79. doi: 10.1146/annurev.nu.01.070181.001353. [DOI] [PubMed] [Google Scholar]
  • 85.Rabot S, Membrez M, Bruneau A, Gérard P, Harach T, Moser M, Raymond F, Mansourian R, Chou CJ. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J. 2010;24:4948–59. doi: 10.1096/fj.10-164921. [DOI] [PubMed] [Google Scholar]
  • 86.Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A. 2007;104:979–84. doi: 10.1073/pnas.0605374104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–23. doi: 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
  • 89.Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1:ra14. doi: 10.1126/scitranslmed.3000322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gustafsson BE, Midtvedt T, Strandberg K. Effects of microbial contamination on the cecum enlargement of germfree rats. Scand J Gastroenterol. 1970;5:309–14. [PubMed] [Google Scholar]
  • 91.Abrams GD, Bishop JE. Effect of the normal microbial flora on gastrointestinal motility. Proc Soc Exp Biol Med. 1967;126:301–4. doi: 10.3181/00379727-126-32430. [DOI] [PubMed] [Google Scholar]
  • 92.Amaral FA, Sachs D, Costa VV, Fagundes CT, Cisalpino D, Cunha TM, Ferreira SH, Cunha FQ, Silva TA, Nicoli JR, et al. Commensal microbiota is fundamental for the development of inflammatory pain. Proc Natl Acad Sci U S A. 2008;105:2193–7. doi: 10.1073/pnas.0711891105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Simrén M, Barbara G, Flint HJ, Spiegel BM, Spiller RC, Vanner S, Verdu EF, Whorwell PJ, Zoetendal EG, Rome Foundation Committee Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut. 2013;62:159–76. doi: 10.1136/gutjnl-2012-302167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.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]
  • 95.Carroll IM, Chang YH, Park J, Sartor RB, Ringel Y. Luminal and mucosal-associated intestinal microbiota in patients with diarrhea-predominant irritable bowel syndrome. Gut Pathog. 2010;2:19. doi: 10.1186/1757-4749-2-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.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]
  • 97.Codling C, O’Mahony L, Shanahan F, Quigley EM, Marchesi JR. A molecular analysis of fecal and mucosal bacterial communities in irritable bowel syndrome. Dig Dis Sci. 2010;55:392–7. doi: 10.1007/s10620-009-0934-x. [DOI] [PubMed] [Google Scholar]
  • 98.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]
  • 99.Ponnusamy K, Choi JN, Kim J, Lee SY, Lee CH. Microbial community and metabolomic comparison of irritable bowel syndrome faeces. J Med Microbiol. 2011;60:817–27. doi: 10.1099/jmm.0.028126-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Silk DB, Davis A, Vulevic J, Tzortzis G, Gibson GR. Clinical trial: the effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome. Aliment Pharmacol Ther. 2009;29:508–18. doi: 10.1111/j.1365-2036.2008.03911.x. [DOI] [PubMed] [Google Scholar]
  • 101.Carroll IM, Ringel-Kulka T, Keku TO, Chang YH, Packey CD, Sartor RB, Ringel Y. Molecular analysis of the luminal- and mucosal-associated intestinal microbiota in diarrhea-predominant irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2011;301:G799–807. doi: 10.1152/ajpgi.00154.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Carroll IM, Ringel-Kulka T, Siddle JP, Ringel Y. Alterations in composition and diversity of the intestinal microbiota in patients with diarrhea-predominant irritable bowel syndrome. Neurogastroenterol Motil. 2012;24:521–30, e248. doi: 10.1111/j.1365-2982.2012.01891.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.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]
  • 104.Kerckhoffs AP, Ben-Amor K, Samsom M, van der Rest ME, de Vogel J, Knol J, Akkermans LM. Molecular analysis of faecal and duodenal samples reveals significantly higher prevalence and numbers of Pseudomonas aeruginosa in irritable bowel syndrome. J Med Microbiol. 2011;60:236–45. doi: 10.1099/jmm.0.022848-0. [DOI] [PubMed] [Google Scholar]
  • 105.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]
  • 106.Noor SO, Ridgway K, Scovell L, Kemsley EK, Lund EK, Jamieson C, Johnson IT, Narbad A. Ulcerative colitis and irritable bowel patients exhibit distinct abnormalities of the gut microbiota. BMC Gastroenterol. 2010;10:134. doi: 10.1186/1471-230X-10-134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Saulnier DM, Riehle K, Mistretta TA, Diaz MA, Mandal D, Raza S, Weidler EM, Qin X, Coarfa C, Milosavljevic A, et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology. 2011;141:1782–91. doi: 10.1053/j.gastro.2011.06.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.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]
  • 109.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]
  • 110.Krogius-Kurikka L, Lyra A, Malinen E, Aarnikunnas J, Tuimala J, Paulin L, Mäkivuokko H, Kajander K, Palva A. Microbial community analysis reveals high level phylogenetic alterations in the overall gastrointestinal microbiota of diarrhoea-predominant irritable bowel syndrome sufferers. BMC Gastroenterol. 2009;9:95. doi: 10.1186/1471-230X-9-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Lyra A, Rinttilä T, Nikkilä J, Krogius-Kurikka L, Kajander K, Malinen E, Mättö J, Mäkelä L, Palva A. Diarrhoea-predominant irritable bowel syndrome distinguishable by 16S rRNA gene phylotype quantification. World J Gastroenterol. 2009;15:5936–45. doi: 10.3748/wjg.15.5936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.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]
  • 113.Maukonen J, Satokari R, Mättö J, Söderlund H, Mattila-Sandholm T, Saarela M. Prevalence and temporal stability of selected clostridial groups in irritable bowel syndrome in relation to predominant faecal bacteria. J Med Microbiol. 2006;55:625–33. doi: 10.1099/jmm.0.46134-0. [DOI] [PubMed] [Google Scholar]
  • 114.Balsari A, Ceccarelli A, Dubini F, Fesce E, Poli G. The fecal microbial population in the irritable bowel syndrome. Microbiologica. 1982;5:185–94. [PubMed] [Google Scholar]
  • 115.Si JM, Yu YC, Fan YJ, Chen SJ. Intestinal microecology and quality of life in irritable bowel syndrome patients. World J Gastroenterol. 2004;10:1802–5. doi: 10.3748/wjg.v10.i12.1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Parkes GC, Rayment NB, Hudspith BN, Petrovska L, Lomer MC, Brostoff J, Whelan K, Sanderson JD. Distinct microbial populations exist in the mucosa-associated microbiota of sub-groups of irritable bowel syndrome. Neurogastroenterol Motil. 2012;24:31–9. doi: 10.1111/j.1365-2982.2011.01803.x. [DOI] [PubMed] [Google Scholar]
  • 117.Durbán A, Abellán JJ, Jiménez-Hernández N, Artacho A, Garrigues V, Ortiz V, Ponce J, Latorre A, Moya A. Instability of the faecal microbiota in diarrhoea-predominant irritable bowel syndrome. FEMS Microbiol Ecol. 2013;86:581–9. doi: 10.1111/1574-6941.12184. [DOI] [PubMed] [Google Scholar]
  • 118.Durbán A, Abellán JJ, Jiménez-Hernández N, Salgado P, Ponce M, Ponce J, Garrigues V, Latorre A, Moya A. Structural alterations of faecal and mucosa-associated bacterial communities in irritable bowel syndrome. Environ Microbiol Rep. 2012;4:242–7. doi: 10.1111/j.1758-2229.2012.00327.x. [DOI] [PubMed] [Google Scholar]
  • 119.Mearin F. Postinfectious functional gastrointestinal disorders. J Clin Gastroenterol. 2011;45(Suppl):S102–5. doi: 10.1097/MCG.0b013e31821fbf58. [DOI] [PubMed] [Google Scholar]
  • 120.Mearin F. Editorial: From the acute infection to the chronic disorder “Don’t worry it’s just a viral gastroenteritis”. Am J Gastroenterol. 2012;107:900–1. doi: 10.1038/ajg.2012.105. [DOI] [PubMed] [Google Scholar]
  • 121.Ford AC, Thabane M, Collins SM, Moayyedi P, Garg AX, Clark WF, Marshall JK. Prevalence of uninvestigated dyspepsia 8 years after a large waterborne outbreak of bacterial dysentery: a cohort study. Gastroenterology. 2010;138:1727–36, quiz e12. doi: 10.1053/j.gastro.2010.01.043. [DOI] [PubMed] [Google Scholar]
  • 122.Thabane M, Kottachchi DT, Marshall JK. Systematic review and meta-analysis: The incidence and prognosis of post-infectious irritable bowel syndrome. Aliment Pharmacol Ther. 2007;26:535–44. doi: 10.1111/j.1365-2036.2007.03399.x. [DOI] [PubMed] [Google Scholar]
  • 123.Marshall JK, Thabane M, Borgaonkar MR, James C. Postinfectious irritable bowel syndrome after a food-borne outbreak of acute gastroenteritis attributed to a viral pathogen. Clin Gastroenterol Hepatol. 2007;5:457–60. doi: 10.1016/j.cgh.2006.11.025. [DOI] [PubMed] [Google Scholar]
  • 124.Zanini B, Ricci C, Bandera F, Caselani F, Magni A, Laronga AM, Lanzini A, San Felice del Benaco Study Investigators Incidence of post-infectious irritable bowel syndrome and functional intestinal disorders following a water-borne viral gastroenteritis outbreak. Am J Gastroenterol. 2012;107:891–9. doi: 10.1038/ajg.2012.102. [DOI] [PubMed] [Google Scholar]
  • 125.Spiller R, Lam C. An Update on Post-infectious Irritable Bowel Syndrome: Role of Genetics, Immune Activation, Serotonin and Altered Microbiome. J Neurogastroenterol Motil. 2012;18:258–68. doi: 10.5056/jnm.2012.18.3.258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Lupp C, Robertson ML, Wickham ME, Sekirov I, Champion OL, Gaynor EC, Finlay BB. Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell Host Microbe. 2007;2:204. doi: 10.1016/j.chom.2007.08.002. [DOI] [PubMed] [Google Scholar]
  • 127.Thabane M, Marshall JK. Post-infectious irritable bowel syndrome. World J Gastroenterol. 2009;15:3591–6. doi: 10.3748/wjg.15.3591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sachdev AH, Pimentel M. Gastrointestinal bacterial overgrowth: pathogenesis and clinical significance. Ther Adv Chronic Dis. 2013;4:223–31. doi: 10.1177/2040622313496126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Spiegel BM. Questioning the bacterial overgrowth hypothesis of irritable bowel syndrome: an epidemiologic and evolutionary perspective. Clin Gastroenterol Hepatol. 2011;9:461–9, quiz e59. doi: 10.1016/j.cgh.2011.02.030. [DOI] [PubMed] [Google Scholar]
  • 130.Ghoshal UC. How to interpret hydrogen breath tests. J Neurogastroenterol Motil. 2011;17:312–7. doi: 10.5056/jnm.2011.17.3.312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Khoshini R, Dai SC, Lezcano S, Pimentel M. A systematic review of diagnostic tests for small intestinal bacterial overgrowth. Dig Dis Sci. 2008;53:1443–54. doi: 10.1007/s10620-007-0065-1. [DOI] [PubMed] [Google Scholar]
  • 132.Pimentel M. Evaluating a bacterial hypothesis in IBS using a modification of Koch’s postulates: part 1. Am J Gastroenterol. 2010;105:718–21. doi: 10.1038/ajg.2009.678. [DOI] [PubMed] [Google Scholar]
  • 133.Dukowicz AC, Lacy BE, Levine GM. Small intestinal bacterial overgrowth: a comprehensive review. Gastroenterol Hepatol (N Y) 2007;3:112–22. [PMC free article] [PubMed] [Google Scholar]
  • 134.Pyleris E, Giamarellos-Bourboulis EJ, Tzivras D, Koussoulas V, Barbatzas C, Pimentel M. The prevalence of overgrowth by aerobic bacteria in the small intestine by small bowel culture: relationship with irritable bowel syndrome. Dig Dis Sci. 2012;57:1321–9. doi: 10.1007/s10620-012-2033-7. [DOI] [PubMed] [Google Scholar]
  • 135.Ford AC, Spiegel BM, Talley NJ, Moayyedi P. Small intestinal bacterial overgrowth in irritable bowel syndrome: systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2009;7:1279–86. doi: 10.1016/j.cgh.2009.06.031. [DOI] [PubMed] [Google Scholar]
  • 136.Chey WD, Maneerattaporn M, Saad R. Pharmacologic and complementary and alternative medicine therapies for irritable bowel syndrome. Gut Liver. 2011;5:253–66. doi: 10.5009/gnl.2011.5.3.253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.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]
  • 138.Pimentel M. An evidence-based treatment algorithm for IBS based on a bacterial/SIBO hypothesis: Part 2. Am J Gastroenterol. 2010;105:1227–30. doi: 10.1038/ajg.2010.125. [DOI] [PubMed] [Google Scholar]
  • 139.Vyas A, Kim SK, Giacomini N, Boothroyd JC, Sapolsky RM. Behavioral changes induced by Toxoplasma infection of rodents are highly specific to aversion of cat odors. Proc Natl Acad Sci U S A. 2007;104:6442–7. doi: 10.1073/pnas.0608310104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Vyas A, Kim SK, Sapolsky RM. The effects of toxoplasma infection on rodent behavior are dependent on dose of the stimulus. Neuroscience. 2007;148:342–8. doi: 10.1016/j.neuroscience.2007.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Haroon F, Händel U, Angenstein F, Goldschmidt J, Kreutzmann P, Lison H, Fischer KD, Scheich H, Wetzel W, Schlüter D, et al. Toxoplasma gondii actively inhibits neuronal function in chronically infected mice. PLoS One. 2012;7:e35516. doi: 10.1371/journal.pone.0035516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bass NM. Review article: the current pharmacological therapies for hepatic encephalopathy. Aliment Pharmacol Ther. 2007;25(Suppl 1):23–31. doi: 10.1111/j.1746-6342.2006.03218.x. [DOI] [PubMed] [Google Scholar]
  • 143.Bass NM, Mullen KD, Sanyal A, Poordad F, Neff G, Leevy CB, Sigal S, Sheikh MY, Beavers K, Frederick T, et al. Rifaximin treatment in hepatic encephalopathy. N Engl J Med. 2010;362:1071–81. doi: 10.1056/NEJMoa0907893. [DOI] [PubMed] [Google Scholar]
  • 144.Sternbach H, State R. Antibiotics: neuropsychiatric effects and psychotropic interactions. Harv Rev Psychiatry. 1997;5:214–26. doi: 10.3109/10673229709000304. [DOI] [PubMed] [Google Scholar]
  • 145.Mehdi S. Antibiotic-induced psychosis: a link to D-alanine? Med Hypotheses. 2010;75:676–7. doi: 10.1016/j.mehy.2010.07.021. [DOI] [PubMed] [Google Scholar]
  • 146.Mulle JG, Sharp WG, Cubells JF. The gut microbiome: a new frontier in autism research. Curr Psychiatry Rep. 2013;15:337. doi: 10.1007/s11920-012-0337-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ledochowski M, Sperner-Unterweger B, Fuchs D. Lactose malabsorption is associated with early signs of mental depression in females: a preliminary report. Dig Dis Sci. 1998;43:2513–7. doi: 10.1023/A:1026654820461. [DOI] [PubMed] [Google Scholar]
  • 148.Ledochowski M, Sperner-Unterweger B, Widner B, Fuchs D. Fructose malabsorption is associated with early signs of mental depression. Eur J Med Res. 1998;3:295–8. [PubMed] [Google Scholar]
  • 149.Ledochowski M, Widner B, Murr C, Sperner-Unterweger B, Fuchs D. Fructose malabsorption is associated with decreased plasma tryptophan. Scand J Gastroenterol. 2001;36:367–71. doi: 10.1080/003655201300051135. [DOI] [PubMed] [Google Scholar]
  • 150.Ledochowski M, Widner B, Sperner-Unterweger B, Propst T, Vogel W, Fuchs D. Carbohydrate malabsorption syndromes and early signs of mental depression in females. Dig Dis Sci. 2000;45:1255–9. doi: 10.1023/A:1005527230346. [DOI] [PubMed] [Google Scholar]
  • 151.Berer K, Krishnamoorthy G. Commensal gut flora and brain autoimmunity: a love or hate affair? Acta Neuropathol. 2012;123:639–51. doi: 10.1007/s00401-012-0949-9. [DOI] [PubMed] [Google Scholar]
  • 152.Berer K, Mues M, Koutrolos M, Rasbi ZA, Boziki M, Johner C, Wekerle H, Krishnamoorthy G. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature. 2011;479:538–41. doi: 10.1038/nature10554. [DOI] [PubMed] [Google Scholar]
  • 153.Ochoa-Repáraz J, Mielcarz DW, Begum-Haque S, Kasper LH. Gut, bugs, and brain: role of commensal bacteria in the control of central nervous system disease. Ann Neurol. 2011;69:240–7. doi: 10.1002/ana.22344. [DOI] [PubMed] [Google Scholar]
  • 154.Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4615–22. doi: 10.1073/pnas.1000082107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, Kubo C, Koga Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558:263–75. doi: 10.1113/jphysiol.2004.063388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, Dinan TG, Cryan JF. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry. 2013;18:666–73. doi: 10.1038/mp.2012.77. [DOI] [PubMed] [Google Scholar]
  • 157.Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, Hibberd ML, Forssberg H, Pettersson S. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011;108:3047–52. doi: 10.1073/pnas.1010529108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil. 2011;23:255–64, e119. doi: 10.1111/j.1365-2982.2010.01620.x. [DOI] [PubMed] [Google Scholar]
  • 159.Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y, Blennerhassett P, Macri J, McCoy KD, et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141:599–609, e1-3. doi: 10.1053/j.gastro.2011.04.052. [DOI] [PubMed] [Google Scholar]
  • 160.Gibson GR, Macfarlane GT, Cummings JH. Sulphate reducing bacteria and hydrogen metabolism in the human large intestine. Gut. 1993;34:437–9. doi: 10.1136/gut.34.4.437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.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]
  • 162.Distrutti E, Cipriani S, Mencarelli A, Renga B, Fiorucci S. Probiotics VSL#3 protect against development of visceral pain in murine model of irritable bowel syndrome. PLoS One. 2013;8:e63893. doi: 10.1371/journal.pone.0063893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Shatalin K, Shatalina E, Mironov A, Nudler E. H2S: a universal defense against antibiotics in bacteria. Science. 2011;334:986–90. doi: 10.1126/science.1209855. [DOI] [PubMed] [Google Scholar]
  • 164.Andriulli A, Neri M, Loguercio C, Terreni N, Merla A, Cardarella MP, Federico A, Chilovi F, Milandri GL, De Bona M, et al. Clinical trial on the efficacy of a new symbiotic formulation, Flortec, in patients with irritable bowel syndrome: a multicenter, randomized study. J Clin Gastroenterol. 2008;42(Suppl 3 Pt 2):S218–23. doi: 10.1097/MCG.0b013e31817fadd6. [DOI] [PubMed] [Google Scholar]
  • 165.Bausserman M, Michail S. The use of Lactobacillus GG in irritable bowel syndrome in children: a double-blind randomized control trial. J Pediatr. 2005;147:197–201. doi: 10.1016/j.jpeds.2005.05.015. [DOI] [PubMed] [Google Scholar]
  • 166.Cui S, Hu Y. Multistrain probiotic preparation significantly reduces symptoms of irritable bowel syndrome in a double-blind placebo-controlled study. Int J Clin Exp Med. 2012;5:238–44. [PMC free article] [PubMed] [Google Scholar]
  • 167.Diop L, Guillou S, Durand H. Probiotic food supplement reduces stress-induced gastrointestinal symptoms in volunteers: a double-blind, placebo-controlled, randomized trial. Nutr Res. 2008;28:1–5. doi: 10.1016/j.nutres.2007.10.001. [DOI] [PubMed] [Google Scholar]
  • 168.Enck P, Zimmermann K, Menke G, Klosterhalfen S. Randomized controlled treatment trial of irritable bowel syndrome with a probiotic E.-coli preparation (DSM17252) compared to placebo. Z Gastroenterol. 2009;47:209–14. doi: 10.1055/s-2008-1027702. [DOI] [PubMed] [Google Scholar]
  • 169.Enck P, Zimmermann K, Menke G, Müller-Lissner S, Martens U, Klosterhalfen S. A mixture of Escherichia coli (DSM 17252) and Enterococcus faecalis (DSM 16440) for treatment of the irritable bowel syndrome--a randomized controlled trial with primary care physicians. Neurogastroenterol Motil. 2008;20:1103–9. doi: 10.1111/j.1365-2982.2008.01156.x. [DOI] [PubMed] [Google Scholar]
  • 170.Francavilla R, Miniello V, Magistà AM, De Canio A, Bucci N, Gagliardi F, Lionetti E, Castellaneta S, Polimeno L, Peccarisi L, et al. A randomized controlled trial of Lactobacillus GG in children with functional abdominal pain. Pediatrics. 2010;126:e1445–52. doi: 10.1542/peds.2010-0467. [DOI] [PubMed] [Google Scholar]
  • 171.Gawrońska A, Dziechciarz P, Horvath A, Szajewska H. A randomized double-blind placebo-controlled trial of Lactobacillus GG for abdominal pain disorders in children. Aliment Pharmacol Ther. 2007;25:177–84. doi: 10.1111/j.1365-2036.2006.03175.x. [DOI] [PubMed] [Google Scholar]
  • 172.Guglielmetti S, Mora D, Gschwender M, Popp K. Randomised clinical trial: Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and improves quality of life--a double-blind, placebo-controlled study. Aliment Pharmacol Ther. 2011;33:1123–32. doi: 10.1111/j.1365-2036.2011.04633.x. [DOI] [PubMed] [Google Scholar]
  • 173.Horvath A, Dziechciarz P, Szajewska H. Meta-analysis: Lactobacillus rhamnosus GG for abdominal pain-related functional gastrointestinal disorders in childhood. Aliment Pharmacol Ther. 2011;33:1302–10. doi: 10.1111/j.1365-2036.2011.04665.x. [DOI] [PubMed] [Google Scholar]
  • 174.Hun L. Bacillus coagulans significantly improved abdominal pain and bloating in patients with IBS. Postgrad Med. 2009;121:119–24. doi: 10.3810/pgm.2009.03.1984. [DOI] [PubMed] [Google Scholar]
  • 175.Niedzielin K, Kordecki H, Birkenfeld B. A controlled, double-blind, randomized study on the efficacy of Lactobacillus plantarum 299V in patients with irritable bowel syndrome. Eur J Gastroenterol Hepatol. 2001;13:1143–7. doi: 10.1097/00042737-200110000-00004. [DOI] [PubMed] [Google Scholar]
  • 176.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]
  • 177.Sinn DH, Song JH, Kim HJ, Lee JH, Son HJ, Chang DK, Kim YH, Kim JJ, Rhee JC, Rhee PL. Therapeutic effect of Lactobacillus acidophilus-SDC 2012, 2013 in patients with irritable bowel syndrome. Dig Dis Sci. 2008;53:2714–8. doi: 10.1007/s10620-007-0196-4. [DOI] [PubMed] [Google Scholar]
  • 178.Whorwell PJ, Altringer L, Morel J, Bond Y, Charbonneau D, O’Mahony L, Kiely B, Shanahan F, Quigley EM. Efficacy of an encapsulated probiotic Bifidobacterium infantis 35624 in women with irritable bowel syndrome. Am J Gastroenterol. 2006;101:1581–90. doi: 10.1111/j.1572-0241.2006.00734.x. [DOI] [PubMed] [Google Scholar]
  • 179.Williams EA, Stimpson J, Wang D, Plummer S, Garaiova I, Barker ME, Corfe BM. Clinical trial: a multistrain probiotic preparation significantly reduces symptoms of irritable bowel syndrome in a double-blind placebo-controlled study. Aliment Pharmacol Ther. 2009;29:97–103. doi: 10.1111/j.1365-2036.2008.03848.x. [DOI] [PubMed] [Google Scholar]
  • 180.Yoon JS, Sohn W, Lee OY, Lee SP, Lee KN, Jun DW, Lee HL, Yoon BC, Choi HS, Chung WS, et al. Effect of multispecies probiotics on irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. J Gastroenterol Hepatol. 2014;29:52–9. doi: 10.1111/jgh.12322. [DOI] [PubMed] [Google Scholar]
  • 181.Ki Cha B, Mun Jung S, Hwan Choi C, Song ID, Woong Lee H, Joon Kim H, Hyuk J, Kyung Chang S, Kim K, Chung WS, et al. The effect of a multispecies probiotic mixture on the symptoms and fecal microbiota in diarrhea-dominant irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. J Clin Gastroenterol. 2012;46:220–7. doi: 10.1097/MCG.0b013e31823712b1. [DOI] [PubMed] [Google Scholar]
  • 182.Michail S, Kenche H. Gut microbiota is not modified by Randomized, Double-blind, Placebo-controlled Trial of VSL#3 in Diarrhea-predominant Irritable Bowel Syndrome. Probiotics Antimicrob Proteins. 2011;3:1–7. doi: 10.1007/s12602-010-9059-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Guandalini S, Magazzù G, Chiaro A, La Balestra V, Di Nardo G, Gopalan S, Sibal A, Romano C, Canani RB, Lionetti P, et al. VSL#3 improves symptoms in children with irritable bowel syndrome: a multicenter, randomized, placebo-controlled, double-blind, crossover study. J Pediatr Gastroenterol Nutr. 2010;51:24–30. doi: 10.1097/MPG.0b013e3181ca4d95. [DOI] [PubMed] [Google Scholar]
  • 184.Benton D, Williams C, Brown A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur J Clin Nutr. 2007;61:355–61. doi: 10.1038/sj.ejcn.1602546. [DOI] [PubMed] [Google Scholar]
  • 185.Messaoudi M, Violle N, Bisson JF, Desor D, Javelot H, Rougeot C. Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes. 2011;2:256–61. doi: 10.4161/gmic.2.4.16108. [DOI] [PubMed] [Google Scholar]
  • 186.Messaoudi M, Lalonde R, Violle N, Javelot H, Desor D, Nejdi A, Bisson JF, Rougeot C, Pichelin M, Cazaubiel M, et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr. 2011;105:755–64. doi: 10.1017/S0007114510004319. [DOI] [PubMed] [Google Scholar]
  • 187.Tillisch K, Labus J, Kilpatrick L, Jiang Z, Stains J, Ebrat B, Guyonnet D, Legrain-Raspaud S, Trotin B, Naliboff B, et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology. 2013;144:1394–401, e1-4. doi: 10.1053/j.gastro.2013.02.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Verdú EF, Bercík P, Bergonzelli GE, Huang XX, Blennerhasset P, Rochat F, Fiaux M, Mansourian R, Corthésy-Theulaz I, Collins SM. Lactobacillus paracasei normalizes muscle hypercontractility in a murine model of postinfective gut dysfunction. Gastroenterology. 2004;127:826–37. doi: 10.1053/j.gastro.2004.06.007. [DOI] [PubMed] [Google Scholar]
  • 189.Johnson AC, Greenwood-Van Meerveld B, McRorie J. Effects of Bifidobacterium infantis 35624 on post-inflammatory visceral hypersensitivity in the rat. Dig Dis Sci. 2011;56:3179–86. doi: 10.1007/s10620-011-1730-y. [DOI] [PubMed] [Google Scholar]
  • 190.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]
  • 191.Dai C, Guandalini S, Zhao DH, Jiang M. Antinociceptive effect of VSL#3 on visceral hypersensitivity in a rat model of irritable bowel syndrome: a possible action through nitric oxide pathway and enhance barrier function. Mol Cell Biochem. 2012;362:43–53. doi: 10.1007/s11010-011-1126-5. [DOI] [PubMed] [Google Scholar]
  • 192.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]
  • 193.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]
  • 194.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]
  • 195.Barreau F, Ferrier L, Fioramonti J, Bueno L. New insights in the etiology and pathophysiology of irritable bowel syndrome: contribution of neonatal stress models. Pediatr Res. 2007;62:240–5. doi: 10.1203/PDR.0b013e3180db2949. [DOI] [PubMed] [Google Scholar]
  • 196.Gareau MG, Jury J, Perdue MH. Neonatal maternal separation of rat pups results in abnormal cholinergic regulation of epithelial permeability. Am J Physiol Gastrointest Liver Physiol. 2007;293:G198–203. doi: 10.1152/ajpgi.00392.2006. [DOI] [PubMed] [Google Scholar]
  • 197.O’Mahony SM, Hyland NP, Dinan TG, Cryan JF. Maternal separation as a model of brain-gut axis dysfunction. Psychopharmacology (Berl) 2011;214:71–88. doi: 10.1007/s00213-010-2010-9. [DOI] [PubMed] [Google Scholar]
  • 198.Lippmann M, Bress A, Nemeroff CB, Plotsky PM, Monteggia LM. Long-term behavioural and molecular alterations associated with maternal separation in rats. Eur J Neurosci. 2007;25:3091–8. doi: 10.1111/j.1460-9568.2007.05522.x. [DOI] [PubMed] [Google Scholar]
  • 199.Ladd CO, Huot RL, Thrivikraman KV, Nemeroff CB, Meaney MJ, Plotsky PM. Long-term behavioral and neuroendocrine adaptations to adverse early experience. Prog Brain Res. 2000;122:81–103. doi: 10.1016/S0079-6123(08)62132-9. [DOI] [PubMed] [Google Scholar]
  • 200.Li M, Xue X, Shao S, Shao F, Wang W. Cognitive, emotional and neurochemical effects of repeated maternal separation in adolescent rats. Brain Res. 2013;1518:82–90. doi: 10.1016/j.brainres.2013.04.026. [DOI] [PubMed] [Google Scholar]
  • 201.Söderholm JD, Yates DA, Gareau MG, Yang PC, MacQueen G, Perdue MH. Neonatal maternal separation predisposes adult rats to colonic barrier dysfunction in response to mild stress. Am J Physiol Gastrointest Liver Physiol. 2002;283:G1257–63. doi: 10.1152/ajpgi.00314.2002. [DOI] [PubMed] [Google Scholar]
  • 202.Varghese AK, Verdú EF, Bercik P, Khan WI, Blennerhassett PA, Szechtman H, Collins SM. Antidepressants attenuate increased susceptibility to colitis in a murine model of depression. Gastroenterology. 2006;130:1743–53. doi: 10.1053/j.gastro.2006.02.007. [DOI] [PubMed] [Google Scholar]
  • 203.Barreau F, Ferrier L, Fioramonti J, Bueno L. Neonatal maternal deprivation triggers long term alterations in colonic epithelial barrier and mucosal immunity in rats. Gut. 2004;53:501–6. doi: 10.1136/gut.2003.024174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Moloney RD, O’Leary OF, Felice D, Bettler B, Dinan TG, Cryan JF. Early-life stress induces visceral hypersensitivity in mice. Neurosci Lett. 2012;512:99–102. doi: 10.1016/j.neulet.2012.01.066. [DOI] [PubMed] [Google Scholar]
  • 205.O’Mahony SM, Marchesi JR, Scully P, Codling C, Ceolho AM, Quigley EM, Cryan JF, Dinan TG. Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry. 2009;65:263–7. doi: 10.1016/j.biopsych.2008.06.026. [DOI] [PubMed] [Google Scholar]
  • 206.Oines E, Murison R, Mrdalj J, Grønli J, Milde AM. Neonatal maternal separation in male rats increases intestinal permeability and affects behavior after chronic social stress. Physiol Behav. 2012;105:1058–66. doi: 10.1016/j.physbeh.2011.11.024. [DOI] [PubMed] [Google Scholar]
  • 207.Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience. 2010;170:1179–88. doi: 10.1016/j.neuroscience.2010.08.005. [DOI] [PubMed] [Google Scholar]
  • 208.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]
  • 209.Eutamene H, Bueno L. Role of probiotics in correcting abnormalities of colonic flora induced by stress. Gut. 2007;56:1495–7. doi: 10.1136/gut.2007.124040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Giada De Palma PB. Jun Lu,Amber J.Park,Vivek Philip,Manuel A.Silva,Elena F.Verdu,Stephen M.Collins,Premysl Bercik. Su1990 The Role of Microbiota in the Maternal Separation Model of Depression. Gastroenterology. 2012;142:pS554. [Google Scholar]
  • 211.Bercík P, De Giorgio R, Blennerhassett P, Verdú EF, Barbara G, Collins SM. Immune-mediated neural dysfunction in a murine model of chronic Helicobacter pylori infection. Gastroenterology. 2002;123:1205–15. doi: 10.1053/gast.2002.36024. [DOI] [PubMed] [Google Scholar]
  • 212.Bercik P, Verdú EF, Foster JA, Lu J, Scharringa A, Kean I, Wang L, Blennerhassett P, Collins SM. Role of gut-brain axis in persistent abnormal feeding behavior in mice following eradication of Helicobacter pylori infection. Am J Physiol Regul Integr Comp Physiol. 2009;296:R587–94. doi: 10.1152/ajpregu.90752.2008. [DOI] [PubMed] [Google Scholar]
  • 213.Bailey MT, Dowd SE, Parry NM, Galley JD, Schauer DB, Lyte M. Stressor exposure disrupts commensal microbial populations in the intestines and leads to increased colonization by Citrobacter rodentium. Infect Immun. 2010;78:1509–19. doi: 10.1128/IAI.00862-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Ibeakanma C, Miranda-Morales M, Richards M, Bautista-Cruz F, Martin N, Hurlbut D, Vanner S. Citrobacter rodentium colitis evokes post-infectious hyperexcitability of mouse nociceptive colonic dorsal root ganglion neurons. J Physiol. 2009;587:3505–21. doi: 10.1113/jphysiol.2009.169110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Mundy R, MacDonald TT, Dougan G, Frankel G, Wiles S. Citrobacter rodentium of mice and man. Cell Microbiol. 2005;7:1697–706. doi: 10.1111/j.1462-5822.2005.00625.x. [DOI] [PubMed] [Google Scholar]
  • 216.Mackos AR, Eubank TD, Parry NM, Bailey MT. Probiotic Lactobacillus reuteri attenuates the stressor-enhanced severity of Citrobacter rodentium infection. Infect Immun. 2013;81:3253–63. doi: 10.1128/IAI.00278-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Ibeakanma C, Ochoa-Cortes F, Miranda-Morales M, McDonald T, Spreadbury I, Cenac N, Cattaruzza F, Hurlbut D, Vanner S, Bunnett N, et al. Brain-gut interactions increase peripheral nociceptive signaling in mice with postinfectious irritable bowel syndrome. Gastroenterology. 2011;141:2098–108, e5. doi: 10.1053/j.gastro.2011.08.006. [DOI] [PubMed] [Google Scholar]
  • 218.Lyte M, Li W, Opitz N, Gaykema RP, Goehler LE. Induction of anxiety-like behavior in mice during the initial stages of infection with the agent of murine colonic hyperplasia Citrobacter rodentium. Physiol Behav. 2006;89:350–7. doi: 10.1016/j.physbeh.2006.06.019. [DOI] [PubMed] [Google Scholar]
  • 219.Riddle MS, Gutierrez RL, Verdu EF, Porter CK. The chronic gastrointestinal consequences associated with campylobacter. Curr Gastroenterol Rep. 2012;14:395–405. doi: 10.1007/s11894-012-0278-0. [DOI] [PubMed] [Google Scholar]
  • 220.Lyte M, Varcoe JJ, Bailey MT. Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiol Behav. 1998;65:63–8. doi: 10.1016/S0031-9384(98)00145-0. [DOI] [PubMed] [Google Scholar]
  • 221.Gaykema RP, Goehler LE, Lyte M. Brain response to cecal infection with Campylobacter jejuni: analysis with Fos immunohistochemistry. Brain Behav Immun. 2004;18:238–45. doi: 10.1016/j.bbi.2003.08.002. [DOI] [PubMed] [Google Scholar]
  • 222.Goehler LE, Gaykema RP, Opitz N, Reddaway R, Badr N, Lyte M. Activation in vagal afferents and central autonomic pathways: early responses to intestinal infection with Campylobacter jejuni. Brain Behav Immun. 2005;19:334–44. doi: 10.1016/j.bbi.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 223.Goehler LE, Park SM, Opitz N, Lyte M, Gaykema RP. Campylobacter jejuni infection increases anxiety-like behavior in the holeboard: possible anatomical substrates for viscerosensory modulation of exploratory behavior. Brain Behav Immun. 2008;22:354–66. doi: 10.1016/j.bbi.2007.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, Deng Y, Blennerhassett PA, Fahnestock M, Moine D, et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterol Motil. 2011;23:1132–9. doi: 10.1111/j.1365-2982.2011.01796.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Bercik P, Verdu EF, Foster JA, Macri J, Potter M, Huang X, Malinowski P, Jackson W, Blennerhassett P, Neufeld KA, et al. Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology. 2010;139:2102–12, e1. doi: 10.1053/j.gastro.2010.06.063. [DOI] [PubMed] [Google Scholar]
  • 226.Gareau MG, Wine E, Rodrigues DM, Cho JH, Whary MT, Philpott DJ, Macqueen G, Sherman PM. Bacterial infection causes stress-induced memory dysfunction in mice. Gut. 2011;60:307–17. doi: 10.1136/gut.2009.202515. [DOI] [PubMed] [Google Scholar]
  • 227.Moayyedi P, Ford AC, Talley NJ, Cremonini F, Foxx-Orenstein AE, Brandt LJ, Quigley EM. The efficacy of probiotics in the treatment of irritable bowel syndrome: a systematic review. Gut. 2010;59:325–32. doi: 10.1136/gut.2008.167270. [DOI] [PubMed] [Google Scholar]
  • 228.Ortiz-Lucas M, Tobías A, Saz P, Sebastián JJ. Effect of probiotic species on irritable bowel syndrome symptoms: A bring up to date meta-analysis. Rev Esp Enferm Dig. 2013;105:19–36. doi: 10.4321/S1130-01082013000100005. [DOI] [PubMed] [Google Scholar]
  • 229.Whelan K, Quigley EM. Probiotics in the management of irritable bowel syndrome and inflammatory bowel disease. Curr Opin Gastroenterol. 2013;29:184–9. doi: 10.1097/MOG.0b013e32835d7bba. [DOI] [PubMed] [Google Scholar]

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

RESOURCES