Abstract
Background
Functional dyspepsia (FD) is a common and debilitating gastrointestinal disorder attributed to altered gut‐brain interactions. While the etiology of FD remains unknown, emerging research suggests the mechanisms are likely multifactorial and heterogenous among patient subgroups. Small bowel motor disturbances, visceral hypersensitivity, chronic microinflammation, and increased intestinal tract permeability have all been linked to the pathogenesis of FD. Recently, alterations to the gut microbiome have also been implicated to play an important role in the disease. Changes to the duodenal microbiota may either trigger or be a consequence of immune and neuronal disturbances observed in the disease, but the mechanisms of influence of small intestinal flora on gastrointestinal function and symptomatology are unknown.
Purpose
This review summarizes and synthesizes the literature on the link between the microbiota, low‐grade inflammatory changes in the duodenum and FD. This review is not intended to provide a complete overview of FD or the small intestinal microbiota, but instead outline some of the key conceptual advances in understanding the interactions between altered gastrointestinal bacterial communities; dietary factors; host immune activation; and stimulation of the gut–brain axes in patients with FD versus controls. Current and emerging treatment approaches such as dietary interventions and antibiotic or probiotic use that have demonstrated symptom benefits for patients are reviewed, and their role in modulating the host–microbiota is discussed. Finally, suggested opportunities for diagnostic and therapeutic improvements for patients with this condition are presented.
Keywords: disorders of gut–brain interaction, dysbiosis, functional dyspepsia, functional gastrointestinal disorders, intestinal mucosa, microbiota, small intestinal bacterial overgrowth
There is accumulating evidence that microbial colonization of the small intestine plays a role in the pathophysiology of disorders of gut–brain interaction. Functional dyspepsia is characterised in a major subset by low grade duodenal inflammation (eosinophils and mast cells), duodenal dysbiosis and altered mucosal barrier dysfunction. While low‐grade inflammation might be the consequence of microbial dysbiosis, the inflammation may play a key role in sensory dysfunction. This disease process resulting in the manifestation of dyspeptic symptoms is likely modified by environmental factors most notably dietary factors or medications.

1. INTRODUCTION
Functional dyspepsia (FD) is a complex and heterogenous disorder characterized by chronic or recurrent upper abdominal pain or discomfort including postprandial fullness and early satiety not explained by structural or biochemical abnormalities identified in the routine clinical setting. 1 Similar to the irritable bowel syndrome (IBS), FD is a disorder of gut–brain interaction (DGBI). 2 These disorders were previously known as functional gastrointestinal disorders (FGID) 3 and likely involve several disease mechanisms including disrupted normal sensorimotor function of the gastrointestinal tract, but with essentially normal endoscopic findings. 3 , 4 , 5
FD is common with a pooled worldwide prevalence of approximately 5%–7%, 6 with variation across geographical regions 6 , 7 ; however, treatment options are currently limited to chronic symptom relief, 8 , 9 contributing to a high disease burden with an impaired quality of life, 6 , 10 and financial hardship. 11 , 12 , 13 Thus, there remains a significant need for further research on the pathophysiology of FD to guide improved diagnostics and therapy for patients.
Based upon Rome IV criteria, FD is stratified into two subtypes: epigastric pain syndrome (EPS), with epigastric pain and/or epigastric burning; and postprandial distress syndrome (PDS), with postprandial fullness and/or early satiety. 3 While different risk factors have been identified for each FD subtype, 14 , 15 overlap is seen in approximately 30–60% of cases. 16 , 17 , 18 Individuals’ symptoms may also change over time. 19 Moreover, many patients with FD have concomitant IBS 20 , 21 or gastroesophageal reflux disease (GERD). 22 , 23 These diseases overlap more than expected by chance and are part of a “spectrum” of gastrointestinal diseases with most likely a similar underlying pathogenesis. 3 , 8 , 22
The pathophysiology of FD is likely multifactorial. 24 Increased intestinal permeability, 25 , 26 gastric hypersensitivity, 27 , 28 gastric motor disturbances such as delayed gastric emptying and fundic dysaccommodation, 27 , 28 H. pylori infection, 29 , 30 abnormalities of bile acid metabolism, 31 dietary, environmental, psychosocial, microbiota, and atopic, allergic, and autoimmune changes have all been implicated in the disease process. 32 Emerging evidence shows that microscopic upper gut alterations may exist in some patient subsets 33 and that the pathophysiology of FD is likely due to alterations in duodenal microbiota, which interact with the immune system to trigger symptoms 8 (Figure 1). Here, we review the recent literature on the duodenal microbiota changes in FD and host immunological, dietary and psychological factors, and outline future directions for mechanistic research in this complex condition.
FIGURE 1.

Proposed disease model for the pathogenesis of functional dyspepsia. 1. Antigen presentation to the small intestinal mucosa, including food macromolecules or microbial antigens. 2. Activation of eosinophils and mast cells through an immune cascade. 3. Local nerve sensitization and systemic immune activation leading to symptomatology. 4. Maintenance of a low‐grade state of inflammation through bidirectional gut–brain and brain–gut pathways [Image created in BioRender]
1.1. The small intestinal microbiome in FD
There is growing interest in the microbiome of the small intestine in FD. The gastrointestinal tract has a large surface and represents one of the major interfaces between the human body and the external environment, with a microbiota made up of over 1000 commensal bacterial species. 34 , 35 Many factors influence microbial composition including diet, environmental exposures, and medications, leading to high interindividual variation. 36 The microbiome plays a role in gut homeostasis and in particular the integrity of the epithelial barrier or the modulation of the mucosal immune system pathways, including tolerance to commensal microorganisms and digested food antigens in the lumen. 37 , 38 , 39 Furthermore, metabolites of the microbiota, most notably, tryptophan, short‐chain fatty acids, and bile salts, interact with a number of pathways in the host. 40 The main two phyla of the gut microbiota are gram‐negative Bacteroidetes and gram‐positive Firmicutes. 41 While extensive physiological microbiota variation and compartmentalization exists along the length of the gastrointestinal tract, 42 , 43 , 44 this may be disturbed in disease states. 45 , 46 , 47 The small bowel is characterized by higher acidic, oxygen, and antimicrobial peptide levels, and these factors and a singular tightly packed mucosal layer and phasic propulsion at the ileum, limit bacterial density compared with the colon. 44
The small intestine is dominated by the phyla Firmicutes, Proteobacteria, and Actinobacteria, with fewer Bacteroidetes, 48 since fast‐growing facultative anaerobes tolerate this environment and can metabolize the simple carbohydrates available. 44 The taxonomy of these is visualized in Figure 2. Moreover, the microbial compositions of the luminal microbiota and the mucosa‐associated microbiota (MAM) are taxonomically and functionally distinct from each other. 44 , 49 The MAM is posited to have a more direct role in the pathogenesis of gastrointestinal diseases due to its proximity to the epithelium. 42 , 50 The mucosal layer acts as a barrier against pathogenic microbes, preventing translocation into host tissue 51 ; hence, the bacteria that can penetrate this layer and in the MAM may have a high potential to induce pro‐inflammatory gene expression in the epithelium. 44 , 52 Furthermore, the duodenal MAM is again taxonomically distinct from other areas of the gastrointestinal tract, 48 dominated by Streptococcus, and lower levels of Prevotella, Veillonella, and Neisseria species. 53
FIGURE 2.

Taxonomy of the small intestinal microbiota across the four main bacterial phyla. Pictured are key intestinal microbial species from phylum–class–genus
Microbial “dysbiosis” is defined as alterations in the composition, density, and function of the intestinal microbes that regulate immune and metabolic homeostasis. 54 Gastrointestinal dysbiosis, especially a decrease in bacterial diversity, is an increasingly recognized feature of multiple chronic noncommunicable diseases such as obesity, liver disease, cardiometabolic conditions, type 2 diabetes mellitus, and malnutrition. 55 It is suggested that microbial dysregulation may enable the expansion of opportunistic “pathobionts”—commensals with pathological potential, 54 activating a disease process 56 such as what is increasingly hypothesized to be a feature of IBS and FD. 54 , 57 , 58
1.2. Small intestinal bacterial overgrowth and FD
DGBIs have been linked to small intestinal bacterial overgrowth (SIBO), 59 , 60 , 61 , 62 , 63 , 64 a clinical condition that is characterized by the presence of excessive and/or abnormal type of microbes in the small intestine. 65 It is currently understood that SIBO involves bacteria that produce hydrogen, while patients that are also colonized with methanogenic archaea, anaerobic organisms producing methane, have an overgrowth termed “intestinal methanogen overgrowth” (IMO), as distinct from SIBO. 65 SIBO is traditionally diagnosed by >105 organisms per mL of jejunal aspirate, 66 or by a positive hydrogen breath test after peroral glucose or lactulose challenge. 65 However, methane breath tests are now used for IMO diagnosis, and the organism cutoff value is debatable, and a lower threshold of ≥103 cfu ml−1 cutoff has been proposed. 67 It should be noted that jejunal aspirates are a limited sampling tool for the small intestinal microbiota due to risk of contamination and difficulties sampling hard‐to‐access areas like blind loops 48 , 66 , 67 , 68 and because the sampled luminal fluid may not represent the mucosal microbial composition. 69 Breath tests lack sensitivity and specificity confounded by intestinal transit, 66 , 70 , 71 and heterogenous methodology. 67 , 71
Sequencing of the duodenum has identified microbial alterations in SIBO, including a lower relative abundance of Firmicutes and a higher abundance of Proteobacteria, both positively associated with decreased alpha diversity. 72 These changes also correlated with symptom severity such as bloating, 72 likely as a result of increased microbial fermentation by the imbalanced over‐abundant community. 73 , 74 SIBO has been shown to be increased in chronic gastrointestinal diseases irrespective of the diagnostic method used. 62 , 75 , 76 , 77 Specifically, the prevalence and risk of SIBO are both increased significantly in chronic uninvestigated dyspepsia and FD compared with controls. 78 A recent study demonstrated elevated duodenal biopsy bacterial loads in DGBI patients compared with controls and increases in breath positivity in some patients, but no association between breath test positivity and duodenal microbial load. 79 Patients diagnosed with SIBO may receive antibiotic treatment such as with the nonabsorbable antibiotic rifaximin, 80 which also provides symptom relief in FD patients. 81 Interestingly, a recent systematic review demonstrated that treatment with rifaximin in combination with fiber, 82 , 83 Lactobacilli and Bifidobacteria probiotics 84 or mesalazine, 85 , 86 augmented the eradication rate of SIBO, further indicating that the condition is likely not underpinned simply by an increased microbial load alone, but also by an element of dysbiosis and possibly inflammation. 80 It has been postulated that SIBO may not be a distinct disease but a pathogenic mechanism underlying the development of FD. 60 The link between SIBO and DGBIs remains controversial, as the small intestinal microbial alterations in symptomatic patients do not always correspond with a positive SIBO diagnosis. 87
1.3. Small intestinal microbial signatures in FD
Recent studies have characterized the duodenal MAM of FD patients to a bacterial genus level. An Australian study first reported the increased relative abundance of Streptococcus in patients with FD compared with matched controls, albeit nonsignificant (which may have been because of the small sample size), and a notable inverse relationship between Streptococcus abundance and that of the anaerobic genera Prevotella, Veillonella, and Actinomyces, which were significantly reduced in the FD patients. Moreover, a higher bacterial load observed in FD patients negatively correlated with bacterial diversity and reported quality of life scores using the Nepean Dyspepsia Index and positively correlated with the severity of upper gastrointestinal meal symptoms. 88 A Japanese study identified an increase in Streptococcus in all sites of the upper gut in an FD cohort that again correlated with patient symptoms scores. 89 Furthermore, the reported beta diversity of the duodenal MAM was significantly different between patients and controls, while the alpha diversity remained unchanged, indicating a more complex microbiota structural change may be involved in the disease rather than changes in relative abundance of particular genera alone. 89 Dyspeptic patients also demonstrated increased anaerobic metabolism in the gastric microbial community in conjunction with increased Pseudoclavibacter and Tannerella, increased Veillonella, Cohnella, Sporolactobacillus, Propionigenium in saliva, and a higher duodenal prevalence of Rothia, Clostridium, Haemophilus, and Actinobacillus species. 90 A recent study demonstrated decreased duodenal mucosal Neisseria and Porphyromonas abundance in FD patients and controls prior to treatment with PPIs, but no differences in microbial load. 91 These findings are presented in Table 1. These bacterial shifts reported in FD patients are typical of the oral microbiota, and one study has confirmed that the healthy duodenum is taxonomically similar to the oral cavity. 42 Importantly, a recent Australian publication describes a new Streptococcus salivarius strain isolated from an FD duodenal biopsy. 92 Overall, this literature confirms that distinct duodenal microbial changes in both microbial load and diversity do occur in patients with FD.
TABLE 1.
Microbial changes in functional dyspepsia patients compared with controls as reported in published literature
| Microbial change | Location | Number of FD patients | Number of controls | Citation |
|---|---|---|---|---|
| Increased relative abundance of Streptococcus | Duodenal biopsies | 9 FD patients | 9 matched endoscopy‐negative controls | Zhong et al. 88 |
| Mucosal brush sample of all sites in upper gut | 11 FD patients | 7 healthy controls | Fukui et al. 89 | |
| Streptococcus abundance positively correlated with severe upper gastrointestinal symptoms | Mucosal brush samples of all sites in upper gut | 11 FD patients | 7 healthy controls | Fukui et al. 89 |
| Lower abundance of Prevotella, Veillonella and Actinomyces | Duodenal biopsies | 9 FD patients | 9 matched endoscopy‐negative controls | Zhong et al. 88 |
| Inverse relationship between Streptococcus and Prevotella, Veillonella and Actinomyces | ||||
| Higher bacterial load negatively correlated with bacterial diversity | ||||
| Higher bacterial load and lower bacterial diversity correlated with quality of life scores | ||||
| Reduced beta diversity | Mucosal brush samples of all sites in upper gut | 11 FD patients | 7 healthy controls | Fukui et al. 89 |
| Unchanged alpha diversity | ||||
| Increased Firmicutes | ||||
| Increased Pseudoclavibacter and Tannerella, increased Veillonella, Cohnella, Sporolactobacillus, Propionigenium in saliva | Gastric and duodenal mucosal brush samples and saliva samples | 25 symptomatic patients with dyspepsia, dysphagia and reflux | 11 patients with achalasia | Cervantes et al. 90 |
| Increased duodenal Rothia, Clostridium, Haemophilus, and Actinobacillus | ||||
| Increased gastric Pseudoclavibacter and Tannerella | ||||
| Lower abundance of Prevotella | Gastric fluid (via nasogastric tube) | 44 FD patients | 44 healthy controls | Nakae et al. 101 |
| Higher abundance of Bifidobacterium and Clostridium | ||||
| Prevotella inversely associated with PDS symptom severity | ||||
| Lower interindividual bacterial diversity | ||||
| Predominance of Bacteroidetes over Proteobacteria | Gastric fluid (via nasogastric tube) | 24 FD patients | 21 matched healthy controls | Igarashi et al. 102 |
| Absence of Acidobacteria | ||||
| Higher Escherichia, Shigella and Bifidobacterium longum | ||||
| Lower Neisseria and Porphyromonas abundance | Mucosal brush samples of duodenum | 28 PPI‐naive FD patients | 30 healthy controls | Wauters et al. 103 |
Abbreviation: FD, functional dyspepsia.
1.4. Effects of antibiotics and probiotics on the microbiome in FD
A pathogenic role of the microbiota in FD is supported by responses to drugs that alter the microbiome (Table 1). Rifaximin is a broad‐spectrum antibiotic with gram‐positive, gram‐negative, aerobic and anaerobic coverage, a high intraluminal bioavailability in the gastrointestinal tract, and minimal systemic adverse effects. 93 It was superior to placebo FD treatment in one randomized trial, wherein relief of dyspeptic symptoms was reported in 79% of patients after taking rifaximin, compared to 47% in the placebo group at 8 weeks. 81 Moreover, the clinical benefits of rifaximin in FD do not appear to be explained by the overlap with IBS. 20 There is some evidence that its efficacy in IBS is due to rifaximin altering the intestinal gut microbiota, 94 and in other gastrointestinal diseases, it has been shown to preserve colonic flora, increasing the abundance of Lactobacilli and Bifidobacteria 95 in a “eubiotic” effect of positively modulating the gut microbiota, favoring the growth of beneficial bacteria altering the overall composition. 96 However, these microbial changes with treatment need to be studied in the duodenum in FD cohorts.
Conversely, previous antibiotic treatments are a risk factor for developing FD, 97 , 98 and it is well known that broad‐spectrum antibiotic use can result in long‐term reductions in commensal species. 55 Probiotics may have a therapeutic role in FD due to their modulation of gastrointestinal flora, and three studies have shown postprandial fullness and bloating symptom improvements after administration of probiotic Lactobacillus gasseri OLL2716 for twelve weeks in H. pylori‐positive and negative dyspeptic patients. 99 , 100 , 101 Symptom resolution was demonstrated in 35.5% of H. pylori‐negative FD patients compared to 17% with placebo. 100 A lower abundance of Prevotella and a higher abundance of Bifidobacterium and Clostridium were seen in the gastric fluid of the FD cohort prior to treatment, but post‐treatment, FD patients showed decreased gastric fluid volume possibly signifying more efficient gastric emptying, and a gastric fluid microbiota resembling the healthy controls pre‐treatment. Interestingly, Prevotella abundance specifically was inversely associated with postprandial distress levels, wherein an increase in this genus post‐treatment was related to symptom alleviation. 101 Lactobacillus gasseri OLL271 was also shown to “restore” the abnormal gastric fluid microbiota of patients with FD. The gastric fluid of FD patients prior to twelve weeks of probiotics showed a predominance of Bacteroidetes over Proteobacteria (24.2/4.9% compared to 13.5/29.5% for controls) and an absence of Acidobacteria (0.2% compared to 3.8% in controls) and post‐probiotic treatment, these proportions shifted to reflect a microbiota composition similar to healthy volunteers. 102 Additionally, while small intestinal bacterial data were not collected, 8 weeks of treatment with Bacillus coagulans MY01 and Bacillus subtilis MY02 demonstrated a 28% response rate improvement over placebo in FD patients. 103 These findings offer preliminary evidence that patient symptoms are related to abundances of certain genera in the upper gut microbiota, which can be modified by therapeutic antibiotic and probiotic agents. Despite these noted specific changes, further research on larger patient cohorts across more diverse geographical regions is needed to characterize the microbiome alterations in the disease, especially in the duodenum.
Proton pump inhibitors (PPIs) also modulate the microbiome, and patients with DGBIs using PPIs have been shown to have a higher bacterial load in the duodenum compared with non‐PPI users, 79 and there an increased risk of SIBO in PPI users has been observed. 104 Studies of the fecal microbiota suggest that PPIs are associated with decreased bacterial diversity, 105 , 106 and a shift toward a community more closely resembling that of the oral microbiota, such as increased Streptococci, 105 , 106 Enterococcus and potentially pathogenic species of Escherichia coli, 105 and decreased Faecalibacterium. 107 Some duodenal microbiota changes at a family level have been observed between PPI users and non‐PPI users, 108 but little is known about how PPIs impact the duodenal MAM in FD. A recent preliminary study using duodenal brushings found significant MAM variation and decreased diversity from baseline after PPI use in FD patients and controls. 91 While the healthy duodenum is taxonomically similar to the oral microbiota, 42 it is of interest to understand whether PPIs reduce the abundance of species specific to the duodenum, resulting in increased similarity to oral bacterial communities. Furthermore, PPIs are a current treatment for FD and gastroesophageal reflux disease (GERD), 109 and oral microbial species have been linked to other gastrointestinal inflammatory diseases, including GERD. 110 , 111 Thus, there is likely to be a complex interrelationship between microbial load, diversity, and PPI use in the small intestine of patients with FD.
1.5. Immune activation and the gastrointestinal microbiome in FD
1.5.1. The microbial antigen or “postinfectious” hypothesis of FD
The microbiota’s significance in gastrointestinal inflammation and FD symptomatology is highlighted in cases of postinfectious FD (PI‐FD). The prevalence of PI‐FD is approximately 10%, with ~2.5 times increased likelihood of development at 6‐month post‐acute gastroenteritis exposure (OR = 2.54, 95% CI 1.76–3.65, p < 0.05). 112 PI‐FD is currently understood to be a product of residual dysfunctional immune activation 113 , 114 ; or from permanent gastrointestinal damage from transient inflammation 27 driven by either microbial displacement or antibiotic treatment. 32 Previously, persistent microscopic duodenitis 114 and higher proportions of eosinophils and mast cells activating in close range (<5 nM) of nerve fibers in the gut have been noted in PI‐FD patients. 115 Postinfectious gastrointestinal symptoms have been reported up to 10 years after an intestinal infection, 116 and those exposed to gastroenteritis were more likely to develop new‐onset dyspepsia 117 and IBS, with relative risks of 5.2 (95% CI 2.7–9.8, p < 0.05) and 7.8 times greater (95% CI 3.1–19.7, p < 0.05), respectively, at 1‐year follow‐up. 118
Moreover, while H. pylori infection is now classified as a separate entity, 3 , 119 its role in the pathogenesis of FD is under scrutiny. 30 , 32 , 70 Higher oesophageal and gastric eosinophils 29 , 120 , 121 , 122 and altered gastric microbiotas including lower bacterial richness have been found in H. pylori‐positive FD patients compared to those not infected. 123 Some H. pylori‐positive patients have decreased Firmicutes, Bacteroidetes, Actinobacteria, 123 , 124 and Fusobacteria, 123 and an increase in Proteobacteria. 124 Moreover, the benefit of long‐term symptom relief after H. pylori eradication therapy in FD has been postulated to come not from the resolution of the infection, but possibly from the antibiotics’ effect on the upper gut microbiota. 125 , 126 , 127 Nonetheless, this supports the role of therapeutics targeting the microbiota in symptom alleviation for FD patients.
1.5.2. Mucosal barrier disruption, inflammation, and dysbiosis
From infancy, an individual’s microbiome matures in conjunction with their immune system. 128 A disruption in the triad of epithelial gut barrier integrity, the gut microbiome, and the immune system, such as intestinal dysbiosis, may lead to pathology and an inflammatory process. Impaired intestinal mucosal barrier integrity has been reported in FD and may relate to microbiome disruption, leading to a conceptualization of the disorder involving a “leaky gut.” 129 A relationship between the extent of increased duodenal mucosal permeability and the severity of low‐grade duodenal inflammation has been reported in FD patients; however, the mechanism by which the microbiome influences this process is still unknown.
Increased immune mediators and cell populations and abnormally decreased protein expression at epithelial intercellular have been seen in FD patients. 25 , 26 , 130 Immune mediator interleukin‐1β disrupts the function of epithelial cells, 131 and in FD patients, increased expression was correlated inversely with duodenal epithelial integrity. 26 Inflammasomes have a crucial role in intestinal mucosa homeostasis as NOD‐like receptor 6 (NLRP6) deficiency leads to defective intestinal goblet cells and a compromised mucosal barrier, which abrogates effective clearance of enteric pathogens. 132 This leaves patients vulnerable to infection and intestinal dysbiosis 133 , 134 and consequential inflammation. 135 In FD patients, duodenal NLRP6 was significantly reduced and correlated with decreased duodenal corticotropin‐releasing hormone (CRH) receptor‐2 expression compared with controls. 136 This indicates that the upregulation of NLRP6 may lead to mucosal disruption and immune activation in FD.
Eosinophils and mast cells in the proximal small intestine are increased in FD and associated with eosinophil degranulation. 137 These cells may be stimulated after barrier disruption, leading to mucosal nerve activation and visceral hypersensitivity, or alternatively, eosinophil degranulation may induce barrier disruption. 138 , 139 The involvement of these cells in FD is supported by patients’ responses to therapeutics such as montelukast, 140 , 141 histamine H1/H2 antagonists, 142 and budesonide. 143 Moreover, emerging evidence indicates that PPIs may have therapeutic value through anti‐inflammatory actions 144 , 145 such as reducing duodenal eosinophils and improve mucosal barrier dysfunction. 146 , 147
Other documented immune changes in FD include increased duodenal intraepithelial lymphocytes in a subset 148 and circulating TNF‐α, IL‐1β, and IL‐10 cytokines in patients compared with controls. 149 Moreover, higher proportions of gut‐homing (positive to CD4, α4 β7 integrins, CCR9‐positive) T cells have also been observed and correlated with gastrointestinal symptoms in patients compared with controls, potentially due to localizing immune recruitment in the small intestine in FD. 149 Notably, increased duodenal populations of CD68‐ and CCR2‐positive cells in PI‐FD patients compared with non‐PI‐FD patients and controls have been reported, suggesting a potential cellular response is triggered by bacterial antigens in this subtype. 114 Moreover, upregulated antimicrobial pathways involving toll‐like receptors 150 , 151 and β‐defensin 2 152 have been observed in other DGBI patients and controls, but these have not yet been studied in FD.
Interestingly, animal studies have demonstrated a relationship between microbial communities including Clostridium and Bacteroides and increased regulatory T cell populations. 153 , 154 , 155 While there is emerging evidence for links between barrier integrity, specific microbial changes, and immune activation, associations between specific microbial communities and immune signatures in FD patients are areas for further study.
1.5.3. Gastrointestinal microbial and immunological relationships with dietary factors in FD
Over 80% of patients with FD report meal‐related gastrointestinal symptoms and alleviation with dietary modification. 14 , 156 Dietary factors are known to play a role in gut symptomatology and may represent a link between food antigens, immune cell activation, and microbiome changes. The potential mechanisms of dietary antigens in this condition have recently been reviewed. 157 , 158 The relationship between digested food, mucosal barrier dysregulation, and hypersensitivity responses is complex. Digested nutrients activate neurons in the small intestine, such as submucosal sensory neurons, and myenteric motor neurons, and the specific site of neuronal activation is nutrient‐specific. 159 FD patients’ increased sensory responses to glucose challenge, 159 lipids, 160 and capsaicin 161 are suggested to be linked to barrier dysfunction and gastric acid hypersensitivity, but the role of the intestinal microbiota in this process is unclear. Notably, patient symptoms after a standardized nutrient challenge were significantly correlated with an increased bacterial load dominated by Streptococcus and Prevotella and reduced Actinomyces. 88 Moreover, this increased bacterial load was negatively correlated with bacterial diversity. 88 This suggests an association between meal‐related symptoms in FD and both microbial load and diversity changes. Interestingly, avoidance of highly processed, “inflammatory” foods minimized the risk of dyspepsia onset, 162 prevented colonic inflammation, 163 , 164 and is associated with beneficial fecal microbial changes. 165 Furthermore, fecal microbiota transplant in IBS patients exhibited gastrointestinal and psychological symptom relief benefits, 166 and associations with increased short‐chain fatty acid butyrate, a fiber metabolite produced by gut microbes, 167 , 168 which has been linked to nociception. 169 , 170 However, these associations are still conjectural and have not yet been investigated in the small intestine of patients with FD.
Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) have been speculated to influence FD symptom onset. FODMAPs are carbohydrates that are poorly absorbed by the small intestine, and their incomplete digestion by the small intestinal microbiota may lead to increased fatty acid, gas production, and increased water transport in the colon leading to bloating, pain, and diarrhea in FD and IBS. 171 Patients with IBS have higher reported levels of known microbial fermentation by‐products compared with controls, 172 and changes of the microbiota may at least partly explain the clinical effects of low‐FODMAP diets in patients with DGBI symptoms. 173 , 174 , 175 , 176 Interestingly, high FODMAPs lead to increased gastrointestinal symptoms, higher levels of anger, and a decreased positive affect in IBS patients compared with controls, 177 indicating gastrointestinal dysfunction is more complex than just macromolecule metabolism and is intertwined with psychological processes. However, a randomized trial of a low‐FODMAP diet in FD was negative in terms of symptom improvement versus a standard diet, although PDS symptoms improved more on a low‐FODMAP diet on a post hoc analysis. 178
FD symptoms may be directly or indirectly influenced by lipid intake interacting with the intestinal microbiota, bile acid signaling, or both. Fatty foods have been associated with FD symptoms, but the mechanism of this is unclear. 156 Dietary fat intake is an independent risk factor for increased gastrointestinal permeability, and most lipids are absorbed in the first 20 cm of the small intestine. 179 , 180 Lipids are a microbial substrate, and the most potent nutrient modulators of gastrointestinal motility and distention through hormones such as cholecystokinin (CCK), released from enteroendocrine cells in a vago‐vagal loop with the brain to regulate satiety. 181 While yet to be studied in FD, a high‐fat diet is associated with a pro‐inflammatory immune cascade involving a transition to barrier‐disrupting hydrophobic bile acids and changes to the intestinal microbiota. 182 , 183 An overall reduced bacterial load and diversity in the gut has been seen 182 and increased Firmicutes and reduced Bacteroidetes 182 (with a predominance of Bacteriodes and lower Prevotella 183 ). This abnormal, higher Firmicutes/Bacteroidetes ratio is associated with disturbances in the intestinal mucosa 184 and may be a hallmark of metabolic pathology 185 ; however, the latter remains controversial. Interestingly, these bacterial changes are consistent with those that have been observed independently of dietary fat in FD patients (Table 1).
The relationship between microbial translocation, intestinal mucosal permeability, and bile salts has been investigated in FD. The microbiota alter the bile acid pool composition through biochemical transformation, such as partial dihydroxylation. 186 Bile salt composition, such as lower concentrations of hydroxylated bile salts, has been associated with increased duodenal permeability 31 and slowed gastric emptying in FD. 187 While remediation by administration of an anti‐inflammatory, hydrophilic bile salt was observed, 31 the influence of bacterial translocation across the lumen on bile salt composition is unclear in FD. 31 Further analysis of the gut microbiota in FD patients and how it modulates nutrient and bile metabolism, gastrointestinal symptom onset, and intestinal function is needed to understand the pathogenesis of this condition.
1.5.4. Gut–brain and brain–gut axes in FD
The immune activation and dysbiosis in the pathogenesis of FD may be explained by a relationship with psychological symptoms through the bidirectional brain–gut and gut–brain axes. The brain can influence the microbiota indirectly via gut motility, secretions, and intestinal permeability, or directly through the release of signaling molecules into the lumen from lamina propria cells. Microbes can conversely communicate with the nervous system by direct stimulation of cells in the lamina propria by microbial metabolites, signaling molecules, and hormones to stimulate vagal pathways and contribute to reward processing, pain, sleep, mood, and cognition. 188
FD was originally conceptualized as psychosomatic, 189 and patients often have extraintestinal symptoms and comorbidities. 190 , 191 , 192 , 193 , 194 FD patients have a higher prevalence of psychiatric comorbidities such as depression and anxiety compared with non‐dyspeptic controls, 195 , 196 and patients may be able to be classified based on their gastrointestinal symptoms and concomitant psychological burden. 197 Baseline anxiety is an independent predictor of persistent GI symptoms and DGBI onset, 198 , 199 but the order of onset of psychiatric conditions and gastrointestinal symptoms may vary between patients. One third of patients have psychological conditions that precede their DGBI diagnosis, 200 while many patients experience psychological disturbances after DGBI onset. 16 , 200 , 201 , 202 , 203 Moreover, those with an FD or IBS diagnosis but no psychological comorbidities at baseline had higher levels of psychological distress at 12‐month follow‐up. 199 Notably, several brain regions in patients with FD demonstrate both white and gray matter anomalies, including the frontal and somatosensory cortices, hippocampus, amygdala, internal capsule, and corpus callosum, suggesting both abnormal central processing (brain to gut) and overactive visceral signaling and pain modulation (gut to brain). 204 , 205 , 206 , 207 , 208 , 209
Chronic stress is a risk factor for FD and IBS, posited to be mediated by the gut–brain axis and hypothalamic–pituitary–adrenal (HPA) axis. 210 Stress may increase intestinal permeability and potentiate the uptake of noxious agents, 211 dysregulate motility, 212 and lead to visceral hypersensitivity 213 and an activated inflammatory state. 214 Anxiety and depression may contribute to the systemic immune activation seen in FD, 215 as CRH released by the hypothalamus during stress is also produced by peripheral inflammatory cells and increases gastrointestinal permeability. 216 Moreover, it is suggested that eosinophils and possibly mast cells may alter neural structure and function, and this sensitization of the enteric nervous system may be exacerbated by pre‐existing psychological problems. 217 Interestingly, anxiety in IBS patients is linked to increased mast cells in the rectum, 218 while gastric mast cell density has been shown to relate to somatization, depression, and anxiety in pediatric FD cases. 219
The intestinal microbiota have a defining impact on the nervous, neuroendocrine, and metabolic systems as outlined in several comprehensive reviews. 188 , 220 , 221 , 222 Microbial factors have now been identified in a number of psychiatric diseases, 40 , 223 , 224 and it is known that gut microbes produce their own neuroactive molecules, potentially having a modifiable impact on brain signaling. 55 This is demonstrated by healthy female adults exhibiting reduced reactive midbrain neural activity in response to a negative emotional attention task after ingestion of a fermented probiotic product containing Bifidobacterium lactis DN‐173 010 for four weeks. 225 This same probiotic product led to a reduction in gastrointestinal symptoms in undiagnosed subjects 226 and improved symptom severity, abdominal transit, and distension in IBS patients. 227 Moreover, Bifidobacterium longum supplementation reduced limbic reactivity and depressive scores in IBS patients. 228 These results may be a product of vagal nerve signaling, as murine and human studies have demonstrated vagal nerve‐mediated anxiolytic effects of Lactobacillus 229 , 230 and Bifidobacterium 230 , 231 and increased neural plasticity gene expression, linked to stress circuitry. 232 Furthermore, other bacterial strains have been linked to changes to serotonin metabolism in the brain stem, 233 inhibited pain sensation from visceral distension 234 , 235 and expression of endogenous opioid and cannabinoid receptors by gut epithelium. 236 These findings exemplify that the intestinal microbiota have a complex relationship with neuroendocrine pathways in the brain–gut and gut–brain axes and likely contribute to both psychological symptoms and gastrointestinal discomfort in FD.
2. LIMITATIONS OF MICROBIAL ANALYSIS IN DGBIs
Analyzing the role of the intestinal microbiome in FD is complicated by difficulties in studying the gut microbes themselves. This is partly due to wide human microbiota heterogeneity between individuals, 237 , 238 and the hostile and dynamic environment of the proximal small intestine wherein microbes have fastidious growth requirements. 36 , 239 , 240 , 241 Culture‐independent methods of analysis remain the mainstay of gastrointestinal microbiological research such as metagenomics and 16S rRNA sequencing. 45 , 242 , 243 However, few metagenomic studies exist due to the emerging technology and high cost, 45 , 243 while the distinct variable rRNA gene regions targeted in different 16S sequencing protocols may influence microbial diversity results and exclude viral and fungal analysis. 244 , 245 Furthermore, the small bowel is difficult to study due to limitations with current sampling and storage methods. 245 The assessment of MAM in the duodenum requires the analysis of endoscopic biopsies as they adequately capture bacteria deep in the mucosa more effectively than other techniques. 44 , 246 However, current biopsy techniques pose a risk of cross‐contamination of biopsy samples, 247 so the more widespread use of novel techniques such as the Brisbane Aseptic Biopsy Device (BABD) that minimizes cross‐contamination from luminal contents, is key for representative analysis of the MAM. 53 Moreover, the heterogenous aggregation of microbial communities along the gastrointestinal tract means may require considered mucosal biopsy technique to obtain samples representative of FD’s disease process and accurately capture any subtle microbial changes. 248 , 249 , 250 Multifarious factors influencing mucosal microbial diversity contribute to extensive variation between individuals, revealing a need for further study to better understand the normal microbiome across a variety of populations. 36 , 251
3. CONCLUSION
There is growing evidence that the microbial colonization of the small intestine plays a role in the pathophysiology of disorders of gut–brain interaction. FD has been shown to have component of gastrointestinal dysbiosis and altered mucosal barrier dysfunction. While low‐grade inflammation might be the consequence of microbial dysbiosis, the inflammation may play a role in sensory dysfunction. This disease process resulting in the manifestation of dyspeptic symptoms is likely modified by environmental factors such as dietary factors or medications. The recent observations require further research to appropriately delineate cause and consequence and explore interventions that allow individualized treatments targeting the causes for symptoms. While the understanding of FD continues to improve, further analysis of the small intestinal microbiome in conjunction with immune cell activation levels and function is required to clarify the pathophysiological mechanisms of this debilitating condition to guide further diagnostic and curative therapeutic innovations.
AUTHOR CONTRIBUTIONS
GB formulated the idea, concept and design, contributed to data extraction, data interpretation, and drafting of the manuscript. ECH reviewed the final manuscript. SK contributed to concept and design, drafting of the manuscript, and review of final manuscript. AS reviewed the final manuscript. MMW contributed to concept and design, drafting of the manuscript, and review of final manuscript. GH reviewed the final manuscript. NJT contributed to concept and design, drafting of the manuscript, and review of final manuscript.
DISCLOSURES
Dr Nicholas J. Talley reports personal fees from Allakos, from Aviro Health, from Antara Life Sciences, from Arlyx from Bayer, from Danone, from Planet Innovation, from Takeda, from Viscera Labs, from twoXAR, from Viscera Labs, from Dr Falk Pharma, from Censa, from Cadila Pharmaceuticals, from Progenity Inc, from Sanofi‐aventis, from Glutagen, from ARENA Pharmaceuticals, from IsoThrive, from BluMaiden, from RosePharma, from Intrinsic Medicine, from OzSage, non‐financial support from HVN National Science Challenge NZ, outside the submitted work; In addition, NJT has a patent Biomarkers of IBS licensed (#12735358.9‐1405/2710383) and (#12735358.9‐1405/2710384), a patent Licensing Questionnaires Talley Bowel Disease Questionnaire licensed to Mayo/Talley, a patent Nestec European Patent licensed, and a patent Singapore Provisional Patent NTU Ref: TD/129/17 “Microbiota Modulation Of BDNF Tissue Repair Pathway” issued and copyright Nepean Dyspepsia Index (NDI) 1998 and Editorial: Medical Journal of Australia (Editor in Chief), Up to Date (Section Editor), Precision and Future Medicine, Sungkyunkwan University School of Medicine, South Korea, Med (Journal of Cell Press). NJT participates Committees: Australian Medical Council (AMC) Council Member (2016–2019), MBS Review Taskforce (2016–2020), NHMRC Principal Committee, Research Committee (2016–2021), Asia Pacific Association of Medical Journal Editors (APAME) (current), GESA Board Member (2017–2019). NJT Misc: Avant Foundation (judging of research grants) (2019). NJT community and patient advocacy groups: Advisory Board, IFFGD (International Foundation for Functional GI Disorders). NJT acknowledges funding from the National Health and Medical Research Council (NHMRC) for the Centre for Research Excellence in Digestive Health. NJT holds an NHMRC Investigator grant. Dr Simon Keely reports grants and research support from the Cancer Institute NSW (Career Development Fellowship), from the National Health and Medical Research Council (Centre of Research Excellence, Project and Ideas Grant), from Viscera Labs, from Gossamer Bio, from Anatara Lifesciences, and from Microba (Funded research and consultancy). Dr Gerald Holtmann is on the advisory boards Australian Biotherapeutics, Glutagen, Bayer and received research support from Bayer, Abbott, Pfizer, Janssen, Takeda, and Allergan. He serves on the Boards of the West Moreton Hospital and Health Service, Queensland, UQ Healthcare, Brisbane and the Gastro‐Liga, Germany. He has a patent for the Brisbane aseptic biopsy device and serves as Editor of the Gastro‐Liga Newsletter. Other co‐authors: None.
GUARANTOR OR ARTICLE
Professor Nicholas J Talley.
WRITING ASSISTANCE
None.
ACKNOWLEDGMENTS
The authors would like to acknowledge our librarians, Ms Debbie Booth and Ms Angela Smith who assisted with the literature search. Open access publishing facilitated by The University of Newcastle, as part of the Wiley ‐ The University of Newcastle agreement via the Council of Australian University Librarians. WOA Institution: The University of Newcastle Blended DEAL: CAUL 2022
Brown G, Hoedt EC, Keely S, et al. Role of the duodenal microbiota in functional dyspepsia. Neurogastroenterology & Motility. 2022;34:e14372. doi: 10.1111/nmo.14372
Funding information
National Health and Medical Research Council (APP1084544) and Centre for Research Excellence (APP170993)
REFERENCES
- 1. Mostafa R. Rome III: The functional gastrointestinal disorders, third edition, 2006. World Journal of Gastroenterology. 2008;14(13):2124. doi: 10.3748/wjg.14.2124 [DOI] [Google Scholar]
- 2. Drossman DA, Hasler WL. Rome IV‐functional GI disorders: disorders of gut‐brain interaction. Gastroenterology. 2016;150(6):1257–1261. [DOI] [PubMed] [Google Scholar]
- 3. Stanghellini V, Chan FKL, Hasler WL, et al. Gastroduodenal disorders. Gastroenterology. 2016;1(6):1380–1392. [DOI] [PubMed] [Google Scholar]
- 4. McLaughlin JT. How should we classify and treat patients with functional gastrointestinal disorders? Therap Adv Gastroenterol. 2008;1(3):153–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Drossman DA. The functional gastrointestinal disorders and the Rome III process. Gastroenterology. 2006;130(5):1377–1390. [DOI] [PubMed] [Google Scholar]
- 6. Sperber AD, Bangdiwala SI, Drossman DA, et al. Worldwide prevalence and burden of functional gastrointestinal disorders, results of Rome foundation global study. Gastroenterology. 2021;160(1):99–114.e113. [DOI] [PubMed] [Google Scholar]
- 7. Ford AC, Marwaha A, Sood R, Moayyedi P. Global prevalence of, and risk factors for, uninvestigated dyspepsia: a meta‐analysis. Gut. 2015;64(7):1049–1057. [DOI] [PubMed] [Google Scholar]
- 8. Talley NJ, Ford AC. Functional dyspepsia. N Engl J Med. 2015;373(19):1853–1863. [DOI] [PubMed] [Google Scholar]
- 9. Talley NJ, Weaver AL, Zinsmeister AR. Impact of functional dyspepsia on quality of life. Dig Dis Sci. 1995;40(3):584–589. [DOI] [PubMed] [Google Scholar]
- 10. Koloski NA, Talley NJ, Boyce PM. The impact of functional gastrointestinal disorders on quality of life. Am J Gastroenterol. 2000;95(1):67–71. [DOI] [PubMed] [Google Scholar]
- 11. Brook RA, Kleinman NL, Choung RS, Melkonian AK, Smeeding JE, Talley NJ. Functional dyspepsia impacts absenteeism and direct and indirect costs. Clin Gastroenterol Hepatol. 2010;8(6):498–503. [DOI] [PubMed] [Google Scholar]
- 12. Sander GB, Mazzoleni LE, Francesconi CFDM, et al. Influence of organic and functional dyspepsia on work productivity: the HEROES‐DIP study. Value Health. 2011;14(5 Suppl 1):S126–129. [DOI] [PubMed] [Google Scholar]
- 13. Lacy BE, Weiser KT, Kennedy AT, Crowell MD, Talley NJ. Functional dyspepsia: the economic impact to patients. Aliment Pharmacol Ther. 2013;38(2):170–177. [DOI] [PubMed] [Google Scholar]
- 14. Aziz I, Palsson OS, Törnblom H, Sperber AD, Whitehead WE, Simrén M. Epidemiology, clinical characteristics, and associations for symptom‐based Rome IV functional dyspepsia in adults in the USA, Canada, and the UK: a cross‐sectional population‐based study. Lancet Gastroenterol Hepatol. 2018;3(4):252–262. [DOI] [PubMed] [Google Scholar]
- 15. Fang Y‐J, Liou J‐M, Chen C‐C, et al. Distinct aetiopathogenesis in subgroups of functional dyspepsia according to the Rome III criteria. Gut. 2015;64(10):1517–1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Carbone F, Holvoet L, Tack J. Rome III functional dyspepsia subdivision in PDS and EPS: recognizing postprandial symptoms reduces overlap. Neurogastroenterol Motil. 2015;27(8):1069–1074. [DOI] [PubMed] [Google Scholar]
- 17. Vakil N, Halling K, Ohlsson L, Wernersson B. Symptom overlap between postprandial distress and epigastric pain syndromes of the Rome III dyspepsia classification. Am J Gastroenterol. 2013;108(5):767–774. [DOI] [PubMed] [Google Scholar]
- 18. Carbone F, Vandenberghe A, Holvoet L, Vanuytsel T, Tack JF. The Impact of Rome IV Criteria on Functional Dyspepsia Subgroups in Secondary Care. Gastroenterology. 2017;152(5):S304. doi: 10.1016/s0016-5085(17)31305-7 [DOI] [Google Scholar]
- 19. Baker G, Fraser RJ, Young G. Subtypes of functional dyspepsia. World J Gastroenterol. 2006;12(17):2667–2671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Shah A, Gurusamy SR, Hansen T, et al. Concomitant irritable bowel syndrome does not influence the response to antimicrobial therapy in patients with functional dyspepsia. Dig Dis Sci. 2021. doi: 10.1007/s10620-021-07149-1 [DOI] [PubMed] [Google Scholar]
- 21. von Wulffen M, Talley NJ, Hammer J, et al. Overlap of irritable bowel syndrome and functional dyspepsia in the clinical setting: prevalence and risk factors. Dig Dis Sci. 2019;64(2):480–486. [DOI] [PubMed] [Google Scholar]
- 22. Rasmussen S, Jensen TH, Henriksen SL, et al. Overlap of symptoms of gastroesophageal reflux disease, dyspepsia and irritable bowel syndrome in the general population. Scand J Gastroenterol. 2015;50(2):162–169. [DOI] [PubMed] [Google Scholar]
- 23. Kaji M, Fujiwara Y, Shiba M, et al. Prevalence of overlaps between GERD, FD and IBS and impact on health‐related quality of life. J Gastroenterol Hepatol. 2010;25(6):1151–1156. [DOI] [PubMed] [Google Scholar]
- 24. Walker MM, Potter MD, Talley NJ. Tangible pathologies in functional dyspepsia. Best Pract Res Clin Gastroenterol. 2019;40–41:101650 (no pagination). [DOI] [PubMed] [Google Scholar]
- 25. Vanheel H, Vicario M, Vanuytsel T, et al. Impaired duodenal mucosal integrity and low‐grade inflammation in functional dyspepsia. Gut. 2014;63(2):262–271. [DOI] [PubMed] [Google Scholar]
- 26. Komori K, Ihara E, Minoda Y, et al. The altered mucosal barrier function in the duodenum plays a role in the pathogenesis of functional dyspepsia. Dig Dis Sci. 2019;64(11):3228–3239. [DOI] [PubMed] [Google Scholar]
- 27. Vanheel H, Farré R. Changes in gastrointestinal tract function and structure in functional dyspepsia. Nat Rev Gastroenterol Hepatol. 2013;10(3):142–149. [DOI] [PubMed] [Google Scholar]
- 28. 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(4):783–788. [DOI] [PubMed] [Google Scholar]
- 29. Ashorn M, Ruuska T, Karikoski R, Välipakka J, Mäki M. Gastric mucosal cell densities in Helicobacter pylori‐positive and ‐negative dyspeptic children and healthy controls. J Pediatr Gastroenterol Nutr. 1994;18(2):146–151. [DOI] [PubMed] [Google Scholar]
- 30. Zullo A, Hassan C, De Francesco V, et al. Helicobacter pylori and functional dyspepsia: an unsolved issue? World J Gastroenterol. 2014;20(27):8957–8963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Beeckmans D, Farré R, Riethorst D, et al. Relationship between bile salts, bacterial translocation, and duodenal mucosal integrity in functional dyspepsia. Neurogastroenterol Motil. 2020;32(5):e13788. [DOI] [PubMed] [Google Scholar]
- 32. Walker MM, Potter MD, Talley NJ. Tangible pathologies in functional dyspepsia. Best Pract Res Clin Gastroenterol. 2019;40–41:101650. [DOI] [PubMed] [Google Scholar]
- 33. Lee KJ, Tack J. Duodenal implications in the pathophysiology of functional dyspepsia. J Neurogastroenterol Motil. 2010;16(3):251–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Sears CL. A dynamic partnership: celebrating our gut flora. Anaerobe. 2005;11(5):247–251. [DOI] [PubMed] [Google Scholar]
- 36. Guarner F, Malagelada J‐R. Gut flora in health and disease. Lancet. 2003;361(9356):512–519. [DOI] [PubMed] [Google Scholar]
- 37. Zoetendal EG, Rajilic‐Stojanovic M, de Vos WM. High‐throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut. 2008;57(11):1605–1615. [DOI] [PubMed] [Google Scholar]
- 38. Yang X, Xie L, Li Y, Wei C. More than 9,000,000 unique genes in human gut bacterial community: estimating gene numbers inside a human body. PLoS One. 2009;4(6):e6074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. 2017;474(11):1823–1836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Spichak S, Bastiaanssen TFS, Berding K, et al. Mining microbes for mental health: determining the role of microbial metabolic pathways in human brain health and disease. Neurosci Biobehav Rev. 2021;125:698–761. [DOI] [PubMed] [Google Scholar]
- 41. Malys MK, Campbell L, Malys N. Symbiotic and antibiotic interactions between gut commensal microbiota and host immune system. Medicina. 2015;51(2):69–75. [DOI] [PubMed] [Google Scholar]
- 42. Kashiwagi S, Naito Y, Inoue R, et al. Mucosa‐associated microbiota in the gastrointestinal tract of healthy Japanese subjects. Digestion. 2020;101(2):107–120. [DOI] [PubMed] [Google Scholar]
- 43. Quigley EMM. Gut bacteria in health and disease. Gastroenterol Hepatol (N Y). 2013;9(9):560–569. [PMC free article] [PubMed] [Google Scholar]
- 44. Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol. 2015;14(1):20–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Zhang S, Cao X, Huang H. Sampling strategies for three‐dimensional spatial community structures in IBD microbiota research. Front Cell Infect Microbiol. 2017;7(FEB):51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Sommer F, Nookaew I, Sommer N, Fogelstrand P, Backhed F. Site‐specific programming of the host epithelial transcriptome by the gut microbiota. Genome Biol. 2015;1:62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Moor AE, Harnik Y, Ben‐Moshe S, et al. Spatial reconstruction of single enterocytes uncovers broad zonation along the intestinal villus axis. Cell. 2018;175(4):1156–1167.e1115. [DOI] [PubMed] [Google Scholar]
- 48. Leite GGS, Weitsman S, Parodi G, et al. Mapping the segmental microbiomes in the human small bowel in comparison with stool: a REIMAGINE study. Dig Dis Sci. 2020;65(9):2595–2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Ringel Y, Maharshak N, Ringel‐Kulka T, Wolber EA, Sartor RB, Carroll IM. High throughput sequencing reveals distinct microbial populations within the mucosal and luminal niches in healthy individuals. Gut Microbes. 2015;6(3):173–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Nishino K, Nishida A, Inoue R, et al. Analysis of endoscopic brush samples identified mucosa‐associated dysbiosis in inflammatory bowel disease. J Gastroenterol. 2018;53(1):95–106. [DOI] [PubMed] [Google Scholar]
- 51. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157(1):121–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Chassaing B, Gewirtz AT. Identification of inner mucus‐associated bacteria by laser capture microdissection. Cell Mol Gastroenterol Hepatol. 2019;1(1):157–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Shanahan ER, Zhong L, Talley NJ, Morrison M, Holtmann G. Characterisation of the gastrointestinal mucosa‐associated microbiota: a novel technique to prevent cross‐contamination during endoscopic procedures. Aliment Pharmacol Ther. 2016;43(11):1186–1196. [DOI] [PubMed] [Google Scholar]
- 54. Chow J, Tang H, Mazmanian SK. Pathobionts of the gastrointestinal microbiota and inflammatory disease. Curr Opin Immunol. 2011;23(4):473–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71. [DOI] [PubMed] [Google Scholar]
- 56. Keely S, Walker MM, Marks E, Talley NJ. Immune dysregulation in the functional gastrointestinal disorders. Eur J Clin Invest. 2015;1(12):1350–1359. [DOI] [PubMed] [Google Scholar]
- 57. Keely S, Feighery L, Campion DP, O'Brien L, Brayden DJ, Baird AW. Chloride‐led disruption of the intestinal mucous layer impedes Salmonella invasion: evidence for an 'enteric tear' mechanism. Cell Physiol Biochem. 2011;28(4):743–752. [DOI] [PubMed] [Google Scholar]
- 58. Bonfrate L, Tack J, Grattagliano I, Cuomo R, Portincasa P. Microbiota in health and irritable bowel syndrome: current knowledge, perspectives and therapeutic options. Scand J Gastroenterol. 2013;48(9):995–1009. [DOI] [PubMed] [Google Scholar]
- 59. Shimura S, Ishimura N, Mikami H, et al. Small intestinal bacterial overgrowth in patients with refractory functional gastrointestinal disorders. Neurogastroenterol Motil. 2016;22(1):60–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tziatzios G, Giamarellos‐Bourboulis EJ, Papanikolaou IS, Pimentel M, Dimitriadis GD, Triantafyllou K. Is small intestinal bacterial overgrowth involved in the pathogenesis of functional dyspepsia? Med Hypotheses. 2017;1:26–32. [DOI] [PubMed] [Google Scholar]
- 61. Hutyra T, Iwanczak B, Pytrus T, Krzesiek E. Assessment of small intestinal bacterial overgrowth in functional disorders of the alimentary canal in children. Adv Clin Exp Med. 2009;18(5):493–500. [Google Scholar]
- 62. Ford AC, Spiegel BMR, Talley NJ, Moayyedi P. Small intestinal bacterial overgrowth in irritable bowel syndrome: systematic review and meta‐analysis. Clin Gastroenterol Hepatol. 2009;7(12):1279–1286. [DOI] [PubMed] [Google Scholar]
- 63. Ghoshal UC, Shukla R, Ghoshal U. Small intestinal bacterial overgrowth and irritable bowel syndrome: a bridge between functional organic dichotomy. Gut Liv. 2017;11(2):196–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Gandhi A, Shah A, Jones MP, et al. Methane positive small intestinal bacterial overgrowth in inflammatory bowel disease and irritable bowel syndrome: a systematic review and meta‐analysis. Gut Microbes. 2021;13(1):1933313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Pimentel M, Saad RJ, Long MD, Rao SSC. ACG clinical guideline: small intestinal bacterial overgrowth. Am College Gastroenterol. 2020;115(2):165–178. [DOI] [PubMed] [Google Scholar]
- 66. Bures J, Cyrany J, Kohoutova D, et al. Small intestinal bacterial overgrowth syndrome. World J Gastroenterol. 2010;16(24):2978–2990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Rezaie A, Buresi M, Lembo A, et al. Hydrogen and methane‐based breath testing in gastrointestinal disorders: the North American consensus. Am J Gastroenterol. 2017;112(5):775–784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Tang Q, Jin GE, Wang G, et al. Current sampling methods for gut microbiota: a call for more precise devices. Front Cell Infect Microbiol. 2020;10:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Shin AS, Gao X, Bohm M, et al. Characterization of proximal small intestinal microbiota in patients with suspected small intestinal bacterial overgrowth: a cross‐sectional study. Clin Transl Gastroenterol. 2019;10(8):e00073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Talley NJ. What causes functional gastrointestinal disorders? A proposed disease model. Am J Gastroenterol. 2020;115(1):41–48. [DOI] [PubMed] [Google Scholar]
- 71. Khoshini R, Dai SC, Lezcano S, Pimentel M. A systematic review of diagnostic tests for small intestinal bacterial overgrowth. Dig Dis Sci. 2008;53(6):1443–1454. [DOI] [PubMed] [Google Scholar]
- 72. Leite G, Morales W, Weitsman S, et al. The duodenal microbiome is altered in small intestinal bacterial overgrowth. PLoS One. 2020;15(7):e0234906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Quigley EMM. Symptoms and the small intestinal microbiome—the unknown explored. Nat Rev Gastroenterol Hepatol. 2019;16(8):457–458. [DOI] [PubMed] [Google Scholar]
- 74. Kastl AJ, Terry NA, Wu GD, Albenberg LG. The structure and function of the human small intestinal microbiota: current understanding and future directions. Cell Mol Gastroenterol Hepatol. 2020;9(1):33–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Shah A, Morrison M, Burger D, et al. Systematic review with meta‐analysis: the prevalence of small intestinal bacterial overgrowth in inflammatory bowel disease. Aliment Pharmacol Ther. 2019;49(6):624–635. [DOI] [PubMed] [Google Scholar]
- 76. Reddymasu SC, Sostarich S, McCallum RW. Small intestinal bacterial overgrowth in irritable bowel syndrome: are there any predictors? BMC Gastroenterol. 2010;10:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Tziatzios G, Gkolfakis P, Papanikolaou IS, et al. High prevalence of small intestinal bacterial overgrowth among functional dyspepsia patients. Dig Dis. 2021;39(4):382–390. [DOI] [PubMed] [Google Scholar]
- 78. Gurusamy SR, Shah A, Talley NJ, et al. Small intestinal bacterial overgrowth in functional dyspepsia: a systematic review and meta‐analysis. Am J Gastroenterol. 2021;116(5):935–942. [DOI] [PubMed] [Google Scholar]
- 79. Shah A, Talley NJ, Koloski N, et al. Duodenal bacterial load as determined by quantitative polymerase chain reaction in asymptomatic controls, functional gastrointestinal disorders and inflammatory bowel disease. Aliment Pharmacol Ther. 2020;52(1):155–167. [DOI] [PubMed] [Google Scholar]
- 80. Gatta L, Scarpignato C. Systematic review with meta‐analysis: rifaximin is effective and safe for the treatment of small intestine bacterial overgrowth. Aliment Pharmacol Ther. 2017;45(5):604–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Tan VP, Liu KS, Lam FY, Hung IF, Yuen MF, Leung WK. Randomised clinical trial: rifaximin versus placebo for the treatment of functional dyspepsia. Aliment Pharmacol Ther. 2017;1(6):767–776. [DOI] [PubMed] [Google Scholar]
- 82. D'incà R, Pomerri F, Vettorato MG, et al. Interaction between rifaximin and dietary fibre in patients with diverticular disease. Aliment Pharmacol Ther. 2007;25(7):771–779. [DOI] [PubMed] [Google Scholar]
- 83. Furnari M, Parodi A, Gemignani L, et al. Clinical trial: the combination of rifaximin with partially hydrolysed guar gum is more effective than rifaximin alone in eradicating small intestinal bacterial overgrowth. Aliment Pharmacol Ther. 2010;32(8):1000–1006. [DOI] [PubMed] [Google Scholar]
- 84. Cuoco L, Salvagnini M. Small intestine bacterial overgrowth in irritable bowel syndrome: a retrospective study with rifaximin. Minerva Gastroenterol Dietol. 2006;52(1):89–95. [PubMed] [Google Scholar]
- 85. Tursi A, Brandimarte G, Giorgetti GM, Elisei W. Assessment of small intestinal bacterial overgrowth in uncomplicated acute diverticulitis of the colon. World J Gastroenterol. 2005;11(18):2773–2776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Biancone L, Vernia P, Agostini D, Ferrieri A, Pallone F. Effect of rifaximin on intestinal bacterial overgrowth in Crohn's disease as assessed by the H2‐Glucose Breath Test. Curr Med Res Opin. 2000;16(1):14–20. [DOI] [PubMed] [Google Scholar]
- 87. Saffouri GB, Shields‐Cutler RR, Chen J, et al. Small intestinal microbial dysbiosis underlies symptoms associated with functional gastrointestinal disorders. Nat Commun. 2019;10(1):2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Zhong L, Shanahan ER, Raj A, et al. Dyspepsia and the microbiome: time to focus on the small intestine. Gut. 2017;1(6):1168–1169. [DOI] [PubMed] [Google Scholar]
- 89. Fukui A, Takagi T, Naito Y, et al. Higher levels of streptococcus in upper gastrointestinal mucosa associated with symptoms in patients with functional dyspepsia. Digestion. 2020;101(1):38–45. [DOI] [PubMed] [Google Scholar]
- 90. Cervantes J, Michael M, Hong B‐Y, et al. Investigation of oral, gastric, and duodenal microbiota in patients with upper gastrointestinal symptoms. Journal of Investigative Medicine. 2021;69(4):870–877. doi: 10.1136/jim-2020-001642 [DOI] [PubMed] [Google Scholar]
- 91. Wauters L, Tito RY, Ceulemans M, et al. Duodenal dysbiosis and relation to the efficacy of proton pump inhibitors in functional dyspepsia. Int J Mol Sci. 2021;22(24):13609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Hoedt EC, Shanahan ER, Keely S, et al. Draft genome sequence of streptococcus salivarius AGIRA0003, isolated from functional gastrointestinal disorder duodenal tissue. Microbiol Resour Announc. 2021;10(39):e0075821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Calanni F, Renzulli C, Barbanti M, Viscomi GC. Rifaximin: beyond the traditional antibiotic activity. J Antibiot (Tokyo). 2014;67(9):667–670. [DOI] [PubMed] [Google Scholar]
- 94. Pimentel M, Lembo A, Chey WD, et al. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med. 2011;364(1):22–32. [DOI] [PubMed] [Google Scholar]
- 95. Maccaferri S, Vitali B, Klinder A, et al. Rifaximin modulates the colonic microbiota of patients with Crohn's disease: an in vitro approach using a continuous culture colonic model system. J Antimicrob Chemother. 2010;65(12):2556–2565. [DOI] [PubMed] [Google Scholar]
- 96. Ponziani FR, Zocco MA, D'Aversa F, Pompili M, Gasbarrini A. Eubiotic properties of rifaximin: disruption of the traditional concepts in gut microbiota modulation. World J Gastroenterol. 2017;23(25):4491–4499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Paula H, Grover M, Halder SL, et al. Non‐enteric infections, antibiotic use, and risk of development of functional gastrointestinal disorders. Neurogastroenterol Motil. 2015;27(11):1580–1586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Villarreal AA, Aberger FJ, Benrud R, Gundrum JD. Use of broad‐spectrum antibiotics and the development of irritable bowel syndrome. WMJ. 2012;111(1):17–20. [PubMed] [Google Scholar]
- 99. Takagi A, Yanagi H, Ozawa H, et al. Effects of lactobacillus gasseri OLL2716 on helicobacter pylori‐associated dyspepsia: a multicenter randomized double‐blind controlled trial. Gastroenterol Res Pract. 2016;2016:7490452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Ohtsu T, Takagi A, Uemura N, et al. The ameliorating effect of lactobacillus gasseri OLL2716 on functional dyspepsia in helicobacter pylori‐uninfected individuals: a randomized controlled study. Digestion. 2017;96(2):92–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Nakae H, Tsuda A, Matsuoka T, Mine T, Koga Y. Gastric microbiota in the functional dyspepsia patients treated with probiotic yogurt. BMJ Open Gastroenterol. 2016;1(1):e000109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Igarashi M, Nakae H, Matsuoka T, et al. Alteration in the gastric microbiota and its restoration by probiotics in patients with functional dyspepsia. BMJ Open Gastroenterol. 2017;4(1):e000144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Wauters L, Slaets H, De Paepe K, et al. Efficacy and safety of spore‐forming probiotics in the treatment of functional dyspepsia: a pilot randomised, double‐blind, placebo‐controlled trial. Lancet Gastroenterol Hepatol. 2021;6(10):784–792. [DOI] [PubMed] [Google Scholar]
- 104. Lo WK, Chan WW. Proton pump inhibitor use and the risk of small intestinal bacterial overgrowth: a meta‐analysis. Clin Gastroenterol Hepatol. 2013;11(5):483–490. [DOI] [PubMed] [Google Scholar]
- 105. Imhann F, Bonder MJ, Vich Vila A, et al. Proton pump inhibitors affect the gut microbiome. Gut. 2016;65(5):740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Jackson MA, Goodrich JK, Maxan M‐E, et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut. 2016;65(5):749–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Takagi T, Naito Y, Inoue R, et al. The influence of long‐term use of proton pump inhibitors on the gut microbiota: an age‐sex‐matched case‐control study. J Clin Biochem Nutr. 2018;62(1):100–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Weitsman S, Celly S, Leite G, et al. Effects of proton pump inhibitors on the small bowel and stool microbiomes. Dig Dis Sci. 2022;67(1):224–232. [DOI] [PubMed] [Google Scholar]
- 109. Pinto‐Sanchez MI, Yuan Y, Bercik P, Moayyedi P. Proton pump inhibitors for functional dyspepsia. Cochrane Database Syst Rev. 2017;3(3):Cd011194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Olsen I, Yamazaki K. Can oral bacteria affect the microbiome of the gut? J Oral Microbiol. 2019;11(1):1586422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Atarashi K, Suda W, Luo C, et al. Ectopic colonization of oral bacteria in the intestine drives T(H)1 cell induction and inflammation. Science (New York, NY). 2017;358(6361):359–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Futagami S, Itoh T, Sakamoto C. Systematic review with meta‐analysis: post‐infectious functional dyspepsia. Aliment Pharmacol Ther. 2015;41(2):177–188. [DOI] [PubMed] [Google Scholar]
- 113. Kindt S, Tertychnyy A, de Hertogh G, Geboes K, Tack J. Intestinal immune activation in presumed post‐infectious functional dyspepsia. Neurogastroenterol Motil. 2009;21(8):832–e856. [DOI] [PubMed] [Google Scholar]
- 114. Futagami S, Shindo T, Kawagoe T, et al. Migration of eosinophils and CCR2‐/CD68‐double positive cells into the duodenal mucosa of patients with postinfectious functional dyspepsia. Am J Gastroenterol. 2010;105(8):1835–1842. [DOI] [PubMed] [Google Scholar]
- 115. Li X, Chen H, Lu H, et al. The study on the role of inflammatory cells and mediators in post‐infectious functional dyspepsia. Scand J Gastroenterol. 2010;45(5):573–581. [DOI] [PubMed] [Google Scholar]
- 116. Schwille‐Kiuntke J, Enck P, Zendler C, et al. Postinfectious irritable bowel syndrome: follow‐up of a patient cohort of confirmed cases of bacterial infection with Salmonella or Campylobacter. Neurogastroenterol Motil. 2011;23(11):e479–488. [DOI] [PubMed] [Google Scholar]
- 117. Ford AC, Thabane M, Collins SM, et al. Prevalence of uninvestigated dyspepsia 8 years after a large waterborne outbreak of bacterial dysentery: a cohort study. Gastroenterology. 2010;138(5):1727–1736. quiz e1712. [DOI] [PubMed] [Google Scholar]
- 118. Mearin F, Pérez‐Oliveras M, Perelló A, et al. Dyspepsia and irritable bowel syndrome after a Salmonella gastroenteritis outbreak: one‐year follow‐up cohort study. Gastroenterology. 2005;129(1):98–104. [DOI] [PubMed] [Google Scholar]
- 119. Jones MP, Talley NJ, Eslick GD, Dubois D, Tack J. Community subgroups in dyspepsia and their association with weight loss. Am J Gastroenterol. 2008;103(8):2051–2060. [DOI] [PubMed] [Google Scholar]
- 120. Cheung CKY, Lan LL, Kyaw M, et al. Up‐regulation of transient receptor potential vanilloid (TRPV) and down‐regulation of brain‐derived neurotrophic factor (BDNF) expression in patients with functional dyspepsia (FD). Neurogastroenterol Motil. 2018;30(2):e13176. [DOI] [PubMed] [Google Scholar]
- 121. Wauters L, Nightingale S, Talley NJ, Sulaiman B, Walker MM. Functional dyspepsia is associated with duodenal eosinophilia in an Australian paediatric cohort. Aliment Pharmacol Ther. 2017;45(10):1358–1364. [DOI] [PubMed] [Google Scholar]
- 122. Collins JS, Hamilton PW, Watt PC, Sloan JM, Love AH. Quantitative histological study of mucosal inflammatory cell densities in endoscopic duodenal biopsy specimens from dyspeptic patients using computer linked image analysis. Gut. 1990;31(8):858–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Gantuya B, El‐Serag HB, Matsumoto T, et al. Gastric microbiota in helicobacter pylori‐negative and ‐positive gastritis among high incidence of gastric cancer area. Cancers (Basel). 2019;11(4):504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Maldonado‐Contreras A, Goldfarb KC, Godoy‐Vitorino F, et al. Structure of the human gastric bacterial community in relation to Helicobacter pylori status. ISME J. 2011;5(4):574–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Moayyedi P, Lacy BE, Andrews CN, Enns RA, Howden CW, Vakil N. ACG and CAG clinical guideline: management of dyspepsia. Am J Gastroenterol. 2017;112(7):988–1013. [DOI] [PubMed] [Google Scholar]
- 126. Lee SP, Lee SY, Kim JH, Sung IK, Park HS, Shim CS. Factors related to upper gastrointestinal symptom generation in 2275 helicobacter pylori seroprevalent adults. Dig Dis Sci. 2017;62(6):1561–1570. [DOI] [PubMed] [Google Scholar]
- 127. Moayyedi P, Soo S, Deeks J, et al. Eradication of helicobacter pylori for non‐ulcer dyspepsia. Cochrane Database Syst Rev. 2006(2):Cd002096. [DOI] [PubMed] [Google Scholar]
- 128. Wu HJ, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2012;3(1):4–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Tack J, Schol J, Van den Houte K, Huang IH, Carbone F. Paradigm shift: functional dyspepsia—a “leaky gut” disorder? Am College Gastroenterol. 2021;116(2):274–275. [DOI] [PubMed] [Google Scholar]
- 130. Taki M, Oshima T, Li M, et al. Duodenal low‐grade inflammation and expression of tight junction proteins in functional dyspepsia. Neurogastroenterol Motil. 2019;31(10):e13576. [DOI] [PubMed] [Google Scholar]
- 131. Al‐Sadi RM, Ma TY. IL‐1β causes an increase in intestinal epithelial tight junction permeability. J Immunol. 2007;178(7):4641–4649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Wlodarska M, Thaiss C, Nowarski R, et al. NLRP6 inflammasome orchestrates the colonic host‐microbial interface by regulating goblet cell mucus secretion. Cell. 2014;156(5):1045–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Gagliani N, Palm NW, de Zoete MR, Flavell RA. Inflammasomes and intestinal homeostasis: regulating and connecting infection, inflammation and the microbiota. Int Immunol. 2014;26(9):495–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Henao‐Mejia J, Elinav E, Jin C, et al. Inflammasome‐mediated dysbiosis regulates progression of NAFLD and obesity. Nature. 2012;482(7384):179–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Elinav E, Strowig T, Kau A, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145(5):745–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Bruce JK, Burns GL, Sinn Soh W, et al. Defects in NLRP6, autophagy and goblet cell homeostasis are associated with reduced duodenal CRH receptor 2 expression in patients with functional dyspepsia. Brain Behav Immun. 2022;101:335–345. [DOI] [PubMed] [Google Scholar]
- 137. Du L, Chen B, Kim JJ, Chen X, Dai N. Micro‐inflammation in functional dyspepsia: a systematic review and meta‐analysis. Neurogastroenterol Motil. 2018;30(4):e13304. [DOI] [PubMed] [Google Scholar]
- 138. Schäppi MG, Borrelli O, Knafelz D, et al. Mast cell‐nerve interactions in children with functional dyspepsia. J Pediatr Gastroenterol Nutr. 2008;47(4):472–480. [DOI] [PubMed] [Google Scholar]
- 139. Sarnelli G, Pesce M, Seguella L, et al. Impaired duodenal palmitoylethanolamide release underlies acid‐induced mast cell activation in functional dyspepsia. Cell Mol Gastroenterol Hepatol. 2021;11(3):841–855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Friesen CA, Neilan NA, Schurman JV, Taylor DL, Kearns GL, Abdel‐Rahman SM. Montelukast in the treatment of duodenal eosinophilia in children with dyspepsia: effect on eosinophil density and activation in relation to pharmacokinetics. BMC Gastroenterol. 2009;9:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Friesen CA, Kearns GL, Andre L, Neustrom M, Roberts CC, Abdel‐Rahman SM. Clinical efficacy and pharmacokinetics of montelukast in dyspeptic children with duodenal eosinophilia. J Pediatr Gastroenterol Nutr. 2004;38(3):343–351. [DOI] [PubMed] [Google Scholar]
- 142. Friesen CA, Sandridge L, Andre L, Roberts CC, Abdel‐Rahman SM. Mucosal eosinophilia and response to H1/H2 antagonist and cromolyn therapy in pediatric dyspepsia. Clin Pediatr (Phila). 2006;45(2):143–147. [DOI] [PubMed] [Google Scholar]
- 143. Talley NJ, Walker MM, Jones M, et al. Letter: budesonide for functional dyspepsia with duodenal eosinophilia—randomised, double‐blind, placebo‐controlled parallel‐group trial. Aliment Pharmacol Ther. 2021;53(12):1332–1333. [DOI] [PubMed] [Google Scholar]
- 144. Zhang XI, Cheng E, Huo X, et al. Omeprazole blocks STAT6 binding to the eotaxin‐3 promoter in eosinophilic esophagitis cells. PLoS One. 2012;7(11):e50037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Min J‐Y, Ocampo CJ, Stevens WW, et al. Proton pump inhibitors decrease eotaxin‐3/CCL26 expression in patients with chronic rhinosinusitis with nasal polyps: possible role of the nongastric H,K‐ATPase. J Allergy Clin Immunol. 2017;139(1):130–141.e111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Wauters L, Ceulemans M, Frings D, et al. Proton pump inhibitors reduce duodenal eosinophilia, mast cells, and permeability in patients with functional dyspepsia. Gastroenterology. 2021;160(5):1521–1531.e1529. [DOI] [PubMed] [Google Scholar]
- 147. Potter MDE, Wood NK, Walker MM, Jones MP, Talley NJ. Proton pump inhibitors and suppression of duodenal eosinophilia in functional dyspepsia. Gut. 2019;68(7):1339. [DOI] [PubMed] [Google Scholar]
- 148. Burns G, Carroll G, Mathe A, et al. Evidence for local and systemic immune activation in functional dyspepsia and the irritable bowel syndrome: a systematic review. Am J Gastroenterol. 2019;114(3):429–436. [DOI] [PubMed] [Google Scholar]
- 149. Liebregts T, Adam B, Bredack C, et al. Small bowel homing T cells are associated with symptoms and delayed gastric emptying in functional dyspepsia. Am J Gastroenterol. 2011;106(6):1089–1098. [DOI] [PubMed] [Google Scholar]
- 150. Dlugosz A, Zakikhany K, Acevedo N, D'Amato M, Lindberg G. Increased expression of toll‐like receptors 4, 5, and 9 in small bowel mucosa from patients with irritable bowel syndrome. Biomed Res Int. 2017;2017:9624702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Testro AG, Visvanathan K. Toll‐like receptors and their role in gastrointestinal disease. J Gastroenterol Hepatol. 2009;24(6):943–954. [DOI] [PubMed] [Google Scholar]
- 152. Langhorst J, Wieder A, Michalsen A, Musial F, Dobos GJ, Rueffer A. Activated innate immune system in irritable bowel syndrome? Gut. 2007;56(9):1325–1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Atarashi K, Tanoue T, Shima T, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331(6015):337–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T‐cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A. 2010;107(27):12204–12209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122(1):107–118. [DOI] [PubMed] [Google Scholar]
- 156. Duncanson K, Talley N, Walker M, Burrows T. Food and functional dyspepsia: a systematic review. J Hum Nutr Diet. 2018;31(3):390–407. [DOI] [PubMed] [Google Scholar]
- 157. Duncanson K, Burns G, Pryor J, Keely S, Talley NJ. Mechanisms of food‐induced symptom induction and dietary management in functional dyspepsia. Nutrients. 2021;13(4):1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Chey WD. The role of food in the functional gastrointestinal disorders: introduction to a manuscript series. Am College Gastroenterol. 2013;108(5):694–697. [DOI] [PubMed] [Google Scholar]
- 159. Puthanmadhom Narayanan S, Linden DR, Peters SA, et al. Duodenal mucosal secretory disturbances in functional dyspepsia. Neurogastroenterol Motil. 2021;33(1):e13955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Feinle C, Meier O, Otto B, D'Amato M, Fried M. Role of duodenal lipid and cholecystokinin A receptors in the pathophysiology of functional dyspepsia. Gut. 2001;48(3):347–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Hammer J, Führer M. Clinical characteristics of functional dyspepsia depending on chemosensitivity to capsaicin. Neurogastroenterol Motil. 2017;29(10):1–12. [DOI] [PubMed] [Google Scholar]
- 162. Keshteli AH, Haghighatdoost F, Azadbakht L, et al. Dietary glycaemic index and glycaemic load and upper gastrointestinal disorders: results from the SEPAHAN study. J Hum Nutr Diet. 2017;30(6):714–723. [DOI] [PubMed] [Google Scholar]
- 163. Keshteli AH, Madsen K, Nickurak C, et al. Adherence to an Anti‐Inflammatory Diet Prevents Increases in Colonic Inflammation in Ulcerative Colitis Patients in Remission. Gastroenterology. 2017;152(5):S599. doi: 10.1016/s0016-5085(17)32142-x [DOI] [Google Scholar]
- 164. Salari‐Moghaddam A, Keshteli AH, Esmaillzadeh A, Adibi P. Adherence to the pro‐inflammatory diet in relation to prevalence of irritable bowel syndrome. Nutr J. 2019;18(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Marlow G, Ellett S, Ferguson IR, et al. Transcriptomics to study the effect of a Mediterranean‐inspired diet on inflammation in Crohn's disease patients. Human Genomics. 2013;7(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Green J, Davis J, Berk M, et al. Efficacy and safety of fecal microbiota transplantation for the treatment of diseases other than Clostridium difficile infection: a systematic review and meta‐analysis. Gut Microbes. 2020;12:1–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. El‐Salhy M, Valeur J, Hausken T, Gunnar HJ. Changes in fecal short‐chain fatty acids following fecal microbiota transplantation in patients with irritable bowel syndrome. Neurogastroenterol Motil. 2021;33(2):e13983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Chassard C, Dapoigny M, Scott KP, et al. Functional dysbiosis within the gut microbiota of patients with constipated‐irritable bowel syndrome. Aliment Pharmacol Ther. 2012;35(7):828–838. [DOI] [PubMed] [Google Scholar]
- 169. 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(5):e63893. doi: 10.1371/journal.pone.0063893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Matsunami M, Tarui T, Mitani K, et al. Luminal hydrogen sulfide plays a pronociceptive role in mouse colon. Gut. 2009;58(6):751–761. [DOI] [PubMed] [Google Scholar]
- 171. Rumessen JJ, Gudmand‐Høyer E. Fructans of chicory: intestinal transport and fermentation of different chain lengths and relation to fructose and sorbitol malabsorption. Am J Clin Nutr. 1998;68(2):357–364. [DOI] [PubMed] [Google Scholar]
- 172. Tana C, Umesaki Y, Imaoka A, Handa T, Kanazawa M, Fukudo S. Altered profiles of intestinal microbiota and organic acids may be the origin of symptoms in irritable bowel syndrome. Neurogastroenterol Motil. 2010;22(5):512–519, e114–515. [DOI] [PubMed] [Google Scholar]
- 173. De Giorgio R, Volta U, Gibson PR. Sensitivity to wheat, gluten and FODMAPs in IBS: facts or fiction? Gut. 2016;65(1):169–178. [DOI] [PubMed] [Google Scholar]
- 174. Gibson PR, Varney J, Malakar S, Muir JG. Food components and irritable bowel syndrome. Gastroenterology. 2015;148(6):1158–1174.e1154. [DOI] [PubMed] [Google Scholar]
- 175. Biesiekierski JR, Peters SL, Newnham ED, Rosella O, Muir JG, Gibson PR. No effects of gluten in patients with self‐reported non‐celiac gluten sensitivity after dietary reduction of fermentable, poorly absorbed, short‐chain carbohydrates. Gastroenterology. 2013;145(2):320–328.e1‐3. [DOI] [PubMed] [Google Scholar]
- 176. Wilder‐Smith C, Olesen SS, Materna A, Drewes A. Predictors of response to a low‐FODMAP diet in patients with functional gastrointestinal disorders and lactose or fructose intolerance. Aliment Pharmacol Ther. 2017;45(8):1094–1106. [DOI] [PubMed] [Google Scholar]
- 177. Masuy I, Van Oudenhove L, Tack J, Biesiekierski J. Effect of intragastric FODMAP infusion on upper gastrointestinal motility, gastrointestinal, and psychological symptoms in irritable bowel syndrome vs healthy controls. Neurogastroenterol Motil. 2018;30(1):e13167. [DOI] [PubMed] [Google Scholar]
- 178. Goyal O, Nohria S, Batta S, Dhaliwal A, Goyal P, Sood A. Low fermentable oligosaccharides, disaccharides, monosaccharides, and polyols diet versus traditional dietary advice for functional dyspepsia: a randomized controlled trial. J Gastroenterol Hepatol. 2022;37(2):301‐309. [DOI] [PubMed] [Google Scholar]
- 179. Leech B, McIntyre E, Steel A, Sibbritt D. Risk factors associated with intestinal permeability in an adult population: a systematic review. Int J Clin Pract. 2019;73(10):e13385. [DOI] [PubMed] [Google Scholar]
- 180. Holtmann G, Kelly DG, Sternby B, DiMagno EP. Survival of human pancreatic enzymes during small bowel transit: effect of nutrients, bile acids, and enzymes. Am J Physiol. 1997;273(2 Pt 1):G553–558. [DOI] [PubMed] [Google Scholar]
- 181. Tome D, Schwarz J, Darcel N, Fromentin G. Protein, amino acids, vagus nerve signaling, and the brain. Am J Clin Nutr. 2009;90(3):838S–843S. [DOI] [PubMed] [Google Scholar]
- 182. Sandhu KV, Sherwin E, Schellekens H, Stanton C, Dinan TG, Cryan JF. Feeding the microbiota‐gut‐brain axis: diet, microbiome, and neuropsychiatry. Transl Res. 2017;179:223–244. [DOI] [PubMed] [Google Scholar]
- 183. Tuck C, Vanner S. Dietary therapies for functional bowel symptoms: recent advances, challenges, and future directions. Neurogastroenterol Motil. 2018;30(1):e13238. [DOI] [PubMed] [Google Scholar]
- 184. Rohr MW, Narasimhulu CA, Rudeski‐Rohr TA, Parthasarathy S. Negative effects of a high‐fat diet on intestinal permeability: a review. Advances in Nutrition. 2020;11(1):77–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Magne F, Gotteland M, Gauthier L, et al. The firmicutes/bacteroidetes ratio: a relevant marker of gut dysbiosis in obese patients? Nutrients. 2020;12(5):1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Ridlon JM, Harris SC, Bhowmik S, Kang DJ, Hylemon PB. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes. 2016;7(1):22–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Wauters L, Ceulemans M, Lambaerts M, et al. Association between duodenal bile salts and gastric emptying in patients with functional dyspepsia. Gut. 2021;70(11):2208‐2210. [DOI] [PubMed] [Google Scholar]
- 188. Cowan CSM, Dinan TG, Cryan JF. Annual Research Review: Critical windows – the microbiota–gut–brain axis in neurocognitive development. Journal of Child Psychology and Psychiatry. 2020;61(3):353–371. doi: 10.1111/jcpp.13156 [DOI] [PubMed] [Google Scholar]
- 189. Einhorn M. Diseases of the Stomach: A Text‐Book for Practitioners and Students. W. Wood; 2010. [Google Scholar]
- 190. Vu J, Kushnir V, Cassell B, Gyawali CP, Sayuk GS. The impact of psychiatric and extraintestinal comorbidity on quality of life and bowel symptom burden in functional GI disorders. Neurogastroenterol Motil. 2014;26(9):1323–1332. [DOI] [PubMed] [Google Scholar]
- 191. Brook RA, Kleinman NL, Choung RS, Smeeding JE, Talley NJ. Excess comorbidity prevalence and cost associated with functional dyspepsia in an employed population. Dig Dis Sci. 2012;57(1):109–118. [DOI] [PubMed] [Google Scholar]
- 192. Matsuzaki J, Suzuki H, Fukushima Y, et al. High frequency of overlap between functional dyspepsia and overactive bladder. Neurogastroenterol Motil. 2012;24(9):821–827. [DOI] [PubMed] [Google Scholar]
- 193. Jones MP, Walker MM, Ford AC, Talley NJ. The overlap of atopy and functional gastrointestinal disorders among 23,471 patients in primary care. Aliment Pharmacol Ther. 2014;40(4):382–391. [DOI] [PubMed] [Google Scholar]
- 194. Persson R, Wensaas KA, Hanevik K, Eide GE, Langeland N, Rortveit G. The relationship between irritable bowel syndrome, functional dyspepsia, chronic fatigue and overactive bladder syndrome: a controlled study 6 years after acute gastrointestinal infection. BMC Gastroenterol. 2015;15:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Haag S, Senf W, Tagay S, et al. Is there a benefit from intensified medical and psychological interventions in patients with functional dyspepsia not responding to conventional therapy? Aliment Pharmacol Ther. 2007;25(8):973–986. [DOI] [PubMed] [Google Scholar]
- 196. Van Oudenhove L, Aziz Q. The role of psychosocial factors and psychiatric disorders in functional dyspepsia. Nat Rev Gastroenterol Hepatol. 2013;10(3):158–167. [DOI] [PubMed] [Google Scholar]
- 197. Barberio B, Pinto‐Sanchez MI, Bercik P, et al. Derivation and validation of a novel method to subgroup patients with functional dyspepsia: beyond upper gastrointestinal symptoms. Aliment Pharmacol Ther. 2021;53(2):253–264. [DOI] [PubMed] [Google Scholar]
- 198. Koloski NA, Talley NJ, Boyce PM. Does psychological distress modulate functional gastrointestinal symptoms and health care seeking? A prospective, community Cohort study. Am J Gastroenterol. 2003;98(4):789–797. [DOI] [PubMed] [Google Scholar]
- 199. 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(9):1284–1290. doi: 10.1136/gutjnl-2011-300474 [DOI] [PubMed] [Google Scholar]
- 200. Koloski NA, Jones M, Talley NJ. Evidence that independent gut‐to‐brain and brain‐to‐gut pathways operate in the irritable bowel syndrome and functional dyspepsia: a 1‐year population‐based prospective study. Aliment Pharmacol Ther. 2016;44(6):592–600. [DOI] [PubMed] [Google Scholar]
- 201. Keely S, Walker MM, Marks E, Talley NJ. Immune dysregulation in the functional gastrointestinal disorders. Eur J Clin Invest. 2015;45(12):1350–1359. [DOI] [PubMed] [Google Scholar]
- 202. Barnes MJ, Powrie F. Regulatory T cells reinforce intestinal homeostasis. Immunity. 2009;31(3):401–411. [DOI] [PubMed] [Google Scholar]
- 203. Liu C‐J, Hu L‐Y, Yeh C‐M, et al. Irritable brain caused by irritable bowel? A nationwide analysis for irritable bowel syndrome and risk of bipolar disorder. PLoS One. 2015;10(3):e0118209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Lee I‐S, Wang H, Chae Y, Preissl H, Enck P. Functional neuroimaging studies in functional dyspepsia patients: a systematic review. Neurogastroenterol Motil. 2016;28(6):793–805. [DOI] [PubMed] [Google Scholar]
- 205. Zhou G, Liu P, Zeng F, et al. Increased interhemispheric resting‐state functional connectivity in functional dyspepsia: a pilot study. NMR Biomed. 2013;26(4):410–415. [DOI] [PubMed] [Google Scholar]
- 206. Zeng F, Qin W, Liang F, et al. Abnormal resting brain activity in patients with functional dyspepsia is related to symptom severity. Gastroenterology. 2011;141(2):499–506. [DOI] [PubMed] [Google Scholar]
- 207. Van Oudenhove L, Vandenberghe J, Dupont P, et al. Abnormal Regional Brain Activity During Rest and (Anticipated) Gastric Distension in Functional Dyspepsia and the Role of Anxiety: A H2 15O‐PET Study. American Journal of Gastroenterology. 2010;105(4):913–924. doi: 10.1038/ajg.2010.39 [DOI] [PubMed] [Google Scholar]
- 208. Vandenberghe J, Dupont P, Van Oudenhove L, et al. Regional cerebral blood flow during gastric balloon distention in functional dyspepsia. Gastroenterology. 2007;132(5):1684–1693. [DOI] [PubMed] [Google Scholar]
- 209. Zhou G, Qin W, Zeng F, et al. White‐matter microstructural changes in functional dyspepsia: a diffusion tensor imaging study. Am College Gastroenterol. 2013;108(2):260–269. [DOI] [PubMed] [Google Scholar]
- 210. Kiank C, Taché Y, Larauche M. Stress‐related modulation of inflammation in experimental models of bowel disease and post‐infectious irritable bowel syndrome: role of corticotropin‐releasing factor receptors. Brain Behav Immun. 2010;24(1):41–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Söderholm JD, Perdue MH. Stress and gastrointestinal tract. II. Stress and intestinal barrier function. Am J Physiol Gastrointest Liver Physiol. 2001;280(1):G7–g13. [DOI] [PubMed] [Google Scholar]
- 212. Qin H‐Y, Cheng C‐W, Tang X‐D, Bian Z‐X. Impact of psychological stress on irritable bowel syndrome. World J Gastroenterol. 2014;20(39):14126–14131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Bonaz B. Visceral sensitivity perturbation integration in the brain‐gut axis in functional digestive disorders. J Physiol Pharmacol. 2003;54(Suppl 4):27–42. [PubMed] [Google Scholar]
- 214. Farhadi A, Fields JZ, Keshavarzian A. Mucosal mast cells are pivotal elements in inflammatory bowel disease that connect the dots: stress, intestinal hyperpermeability and inflammation. World J Gastroenterol. 2007;13(22):3027–3030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Jones MP, Tack J, Van Oudenhove L, et al. Mood and anxiety disorders precede development of functional gastrointestinal disorders in patients but not in the population. Clin Gastroenterol Hepatol. 2017;15(7):1014–1020.e1014. [DOI] [PubMed] [Google Scholar]
- 216. Theoharides TC, Cochrane DE. Critical role of mast cells in inflammatory diseases and the effect of acute stress. J Neuroimmunol. 2004;146(1–2):1–12. [DOI] [PubMed] [Google Scholar]
- 217. Jung HK, Talley NJ. Role of the duodenum in the pathogenesis of functional dyspepsia: a paradigm shift. J Neurogastroenterol Motil. 2018;24(3):345–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Liu D‐R, Xu X‐J, Yao S‐K. Increased intestinal mucosal leptin levels in patients with diarrhea‐predominant irritable bowel syndrome. World J Gastroenterol. 2018;24(1):46–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Schurman JV, Singh M, Singh V, Neilan N, Friesen CA. Symptoms and subtypes in pediatric functional dyspepsia: relation to mucosal inflammation and psychological functioning. J Pediatr Gastroenterol Nutr. 2010;51(3):298–303. [DOI] [PubMed] [Google Scholar]
- 220. Collins SM, Bercik P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology. 2009;136(6):2003–2014. [DOI] [PubMed] [Google Scholar]
- 221. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nat Rev Gastroenterol Hepatol. 2009;6(5):306–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Forsythe P, Sudo N, Dinan T, Taylor VH, Bienenstock J. Mood and gut feelings. Brain Behav Immun. 2010;24(1):9–16. [DOI] [PubMed] [Google Scholar]
- 223. Misiak B, Łoniewski I, Marlicz W, et al. The HPA axis dysregulation in severe mental illness: can we shift the blame to gut microbiota? Prog Neuropsychopharmacol Biol Psychiatry. 2020;102:109951. [DOI] [PubMed] [Google Scholar]
- 224. Wilmes L, Collins JM, O'Riordan KJ, O’Mahony SM, Cryan JF, Clarke G. Of bowels, brain and behavior: a role for the gut microbiota in psychiatric comorbidities in irritable bowel syndrome. Neurogastroenterol Motil. 2021;33(3):e14095. [DOI] [PubMed] [Google Scholar]
- 225. Tillisch K, Labus J, Kilpatrick L, et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology. 2013;144(7):1394‐1401, 1401.e1391‐1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Guyonnet D, Schlumberger A, Mhamdi L, Jakob S, Chassany O. Fermented milk containing Bifidobacterium lactis DN‐173 010 improves gastrointestinal well‐being and digestive symptoms in women reporting minor digestive symptoms: a randomised, double‐blind, parallel, controlled study. Br J Nutr. 2009;102(11):1654–1662. [DOI] [PubMed] [Google Scholar]
- 227. Agrawal A, Houghton LA, Morris J, et al. Clinical trial: the effects of a fermented milk product containing Bifidobacterium lactis DN‐173 010 on abdominal distension and gastrointestinal transit in irritable bowel syndrome with constipation. Aliment Pharmacol Ther. 2009;29(1):104–114. [DOI] [PubMed] [Google Scholar]
- 228. Pinto‐Sanchez MI, Hall GB, Ghajar K, et al. Probiotic bifidobacterium longum NCC3001 reduces depression scores and alters brain activity: a pilot study in patients with irritable bowel syndrome. Gastroenterology. 2017;153(2):448–459.e448. [DOI] [PubMed] [Google Scholar]
- 229. Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011;108(38):16050–16055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Messaoudi M, Violle N, Bisson J‐F, 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(4):256–261. [DOI] [PubMed] [Google Scholar]
- 231. Bercik P, Park AJ, Sinclair D, et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut‐brain communication. Neurogastroenterol Mot. 2011;23(12):1132–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety‐like behavior and central neurochemical change in germ‐free mice. Neurogastroenterol Motil. 2011;23(3):pp. 255–264, e119. [DOI] [PubMed] [Google Scholar]
- 233. Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J Psychiatr Res. 2008;43(2):164–174. [DOI] [PubMed] [Google Scholar]
- 234. Kamiya T, Wang L, Forsythe P, et al. Inhibitory effects of Lactobacillus reuteri on visceral pain induced by colorectal distension in Sprague‐Dawley rats. Gut. 2006;55(2):191–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Verdú EF, Bercik P, Verma‐Gandhu M, et al. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut. 2006;55(2):182–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med. 2007;13(1):35–37. [DOI] [PubMed] [Google Scholar]
- 237. Turnbaugh PJ, Quince C, Faith JJ, et al. Organismal, genetic, and transcriptional variation in the deeply sequenced gut microbiomes of identical twins. Proc Natl Acad Sci U S A. 2010;107(16):7503–7508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Martínez I, Muller CE, Walter J. Long‐term temporal analysis of the human fecal microbiota revealed a stable core of dominant bacterial species. PLoS One. 2013;8(7):e69621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Walker MM, Talley NJ. Review article: bacteria and pathogenesis of disease in the upper gastrointestinal tract–beyond the era of Helicobacter pylori. Aliment Pharmacol Ther. 2014;39(8):767–779. [DOI] [PubMed] [Google Scholar]
- 240. Zoetendal EG, Raes J, van den Bogert B, et al. The human small intestinal microbiota is driven by rapid uptake and conversion of simple carbohydrates. ISME J. 2012;6(7):1415–1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. De Hertogh G, Lemmens B, Verhasselt P, et al. Assessment of the microbiota in microdissected tissues of Crohn's disease patients. Int J Inflam. 2012;1:505674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Mizrahi‐Man O, Davenport ER, Gilad Y. Taxonomic classification of bacterial 16S rRNA genes using short sequencing reads: evaluation of effective study designs. PLoS One. 2013;8(1):e53608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Poretsky R, Rodriguez RL, Luo C, Tsementzi D, Konstantinidis KT. Strengths and limitations of 16S rRNA gene amplicon sequencing in revealing temporal microbial community dynamics. PLoS One. 2014;9(4):e93827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Zhang J, Zhu C, Guan R, et al. Microbial profiles of a drinking water resource based on different 16S rRNA V regions during a heavy cyanobacterial bloom in Lake Taihu, China. Environ Sci Pollut Res Int. 2017;24(14):12796–12808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Bharti R, Grimm DG. Current challenges and best‐practice protocols for microbiome analysis. Brief Bioinform. 2021;22(1):178–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Booijink CCGM, El‐Aidy S, Rajilić‐Stojanović M, et al. High temporal and inter‐individual variation detected in the human ileal microbiota. Environ Microbiol. 2010;12(12):3213–3227. [DOI] [PubMed] [Google Scholar]
- 247. Talley NJ, Holtmann G, Walker MM. Therapeutic strategies for functional dyspepsia and irritable bowel syndrome based on pathophysiology. J Gastroenterol. 2015;50(6):601–613. [DOI] [PubMed] [Google Scholar]
- 248. Nava GM, Carbonero F, Croix JA, Greenberg E, Gaskins HR. Abundance and diversity of mucosa‐associated hydrogenotrophic microbes in the healthy human colon. ISME J. 2012;6(1):57–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Hong PY, Croix JA, Greenberg E, Gaskins HR, Mackie RI. Pyrosequencing‐based analysis of the mucosal microbiota in healthy individuals reveals ubiquitous bacterial groups and micro‐heterogeneity. PLoS One. 2011;6(9):e25042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Zhang Z, Geng J, Tang X, et al. Spatial heterogeneity and co‐occurrence patterns of human mucosal‐associated intestinal microbiota. ISME J. 2014;8(4):881–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Vaga S, Lee S, Ji B, et al. Compositional and functional differences of the mucosal microbiota along the intestine of healthy individuals. Sci Rep. 2020;10(1):14977. [DOI] [PMC free article] [PubMed] [Google Scholar]
