Review Open Access
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Mar 26, 2020; 8(6): 1013-1025
Published online Mar 26, 2020. doi: 10.12998/wjcc.v8.i6.1013
Microbiota-gut-brain axis and its affect inflammatory bowel disease: Pathophysiological concepts and insights for clinicians
Emanuele Sinagra, Gastroenterology and Endoscopy Unit, Fondazione Istituto Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, Cefalù 90015, Italy
Emanuele Sinagra, Euro-Mediterranean Institute of Science and Technology, Palermo 90100, Italy
Erika Utzeri, Nuova Casa di Cura di Decimomannu, Cagliari 09100, Italy
Gaetano Cristian Morreale, Section of Gastroenterology, S. Elia-Raimondi Hospital, Caltanissetta 93100, Italy
Carlo Fabbri, Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena, Azienda USL Romagna, Forlì 47121, Italy
Fabio Pace, Unit of Gastroenterology, Bolognini Hospital, Bergamo 24100, Italy
Andrea Anderloni, Digestive Endoscopy Unit, Division of Gastroenterology, Humanitas Research Hospital, Rozzano 20089, Italy
ORCID number: Emanuele Sinagra (0000-0002-8528-0384); Erika Utzeri (0000-0002-5125-118X); Gaetano Cristian Morreale (0000-0001-8954-7819); Carlo Fabbri (0000-0003-2430-7149); Fabio Pace (0000-0002-8727-5757); Andrea Anderloni (0000-0002-1021-0031).
Author contributions: Sinagra E designed the study; Sinagra E, Utzeri E, and Morreale GC wrote the paper; Pace F and Jones R contributed to the revision of the manuscript; Anderloni A supervised the work.
Conflict-of-interest statement: All the authors declare that this research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors, and thus there is no conflict of interest regarding this paper.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Emanuele Sinagra, MD, PhD, Doctor, Gastroenterology and Endoscopy Unit, Fondazione Istituto Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, Cefalù 90015, Italy. emanuelesinagra83@googlemail.com
Received: December 30, 2019
Peer-review started: December 30, 2019
First decision: January 19, 2020
Revised: February 14, 2020
Accepted: March 5, 2020
Article in press: March 5, 2020
Published online: March 26, 2020
Processing time: 86 Days and 10.2 Hours

Abstract

Despite the bi-directional interaction between gut microbiota and the brain not being fully understood, there is increasing evidence arising from animal and human studies that show how this intricate relationship may facilitate inflammatory bowel disease (IBD) pathogenesis, with consequent important implications on the possibility to improve the clinical outcomes of the diseases themselves, by acting on the different components of this system, mainly by modifying the microbiota. With the emergence of precision medicine, strategies in which patients with IBD might be categorized other than for standard gut symptom complexes could offer the opportunity to tailor therapies to individual patients. The aim of this narrative review is to elaborate on the concept of the gut-brain-microbiota axis and its clinical significance regarding IBD on the basis of recent scientific literature, and finally to focus on pharmacological therapies that could allow us to favorably modify the function of this complex system.

Key Words: Irritable bowel syndrome, Inflammatory bowel disease, Gut-brain axis, Therapy

Core tip: The complex interplay between gut microbiota and the brain, and vice versa, has recently become not only the focus of neuroscience, but also the start point for research regarding many diseases such as inflammatory bowel diseases (IBD) and irritable bowel syndrome. The bi-directional interaction between gut microbiota and the brain is not completely understood. Nonetheless, there is increasing evidence arising from animal and human studies that show how this intricate relationship may contribute to the pathogenesis of IBD, with consequent important implications for the possibility to improve the clinical outcomes of the diseases themselves by acting on the different components of this axis. With the emergence of precision medicine, strategies in which patients with IBD might be categorized other than for standard gut symptom complexes could offer the opportunity to tailor therapies to individual patients.



INTRODUCTION

The complex interplay between gut microbiota and the brain, and vice versa, has recently become not only the focus of neuroscience, but also the start point for research regarding many diseases such as inflammatory bowel diseases (IBD) and irritable bowel syndrome (IBS)[1-4].

The bi-directional interaction between gut microbiota and the brain is not completely understood. Nonetheless, there is increasing evidence arising from animal and human studies that show how this intricate relationship may contribute to the pathogenesis of IBD, with consequent important implications for the possibility to improve the clinical outcomes of the diseases themselves by acting on the different components of this axis[5-7].

The aim of this narrative review is to elaborate on the concept of the-gut-brain-microbiota axis and its clinical significance regarding IBD on the basis of recent scientific literature, and finally to focus on pharmacological therapies that could allow us to favorably modify the function of this complex system.

WHAT IS THE GUT-BRAIN-MICROBIOTA AXIS?

While the idea that the brain can alter intestinal functions has long been recognized and accepted, the concept that signals from the gut can have effects on mood, behavior and cognitive function is less widely accepted.

First of all, it should be highlighted that the digestive system is very complex. This is due to the fact that it represents not only a surface of about 300 square meters for absorption, but it also a very complex hormonal system that produces more than thirty hormones, and of a very complex immune system, since it contains 60%-70% of a person’s immune cells. Furthermore, it is an extremely innervated system, with an extensive intrinsic nervous system, namely, the enteric nervous system (ENS), that controls bowel function even though it is completely separate from the central nervous system (CNS)[8,9].

Finally, our digestive system contains various microorganisms [about 10 (14)], such as bacteria, fungi, parasites, and viruses, and more than 100 million bacteria reside in the human gastrointestinal tract, establishing a mutually beneficial symbiotic state with the human organism. The gut microbiota controls the development and homeostasis of the host by acting on human metabolism and immune function, and also by controlling xenobiotic and drug metabolism, maintaining the structural integrity of the gut mucosal barrier, regulating the motility of the gut and promoting protection against pathogens.

The gut-brain-microbiota axis is defined as a two-way communication system that allows intestinal microbes to communicate with the brain and vice versa. This system, which has not been entirely explored, is based on neural, endocrine, immunological and metabolic pathways[10,11]. This network contains several levels of communication and complex interplay, shown in Figure 1, namely[12]: (1) Gut microbiota, acting bi-directionally in this system through several mechanisms; (2) The ENS, which is made up of interneurons, sensory neurons, motor neurons, and neurotransmitters; (3) The autonomic nervous system (ANS), the pivotal modulator of the ENS; (4) The entero-endocrine and immune signaling agents; (5) The neuroendocrine signaling network mediated by the hypothalamic-pituitary-adrenal (HPA) axis, which is activated by the integrative reactions of specific centers in the CNS, and represents a central integrative system mandatory for the successful physiological adaptation of our organism to stress; and (6) The CNS, includes the hypothalamus, amygdala, and hippocampus and their interaction with emotional centers localized within the limbic system, which are mainly involved in the control of body reaction in response to stress.

Figure 1
Figure 1 The main actors in the gut–brain-microbiota axis. The gut-brain-microbiota axis is defined as a two-way communication system that allows intestinal microbes to communicate with the brain and vice versa. The multiple inter-related structural networks of the central nervous system regulates autonomic nervous system input that alter gut microbial composition and function indirectly by modulating the microbial environment in the gut, and by affecting also the immune response. On the other hand, the gut microbiota can interact with the brain indirectly via gut-derived metabolites by acting on afferent vagal and/or spinal nerve endings, or directly via microbe-generated signals which act on enteroendocrine cells. Furthermore, the neuroendocrine signaling network mediated by the hypothalamic-pituitary-adrenal) axis, which is activated by the integrative reactions of specific centers in the central nervous system, represents a central integrative system mandatory for the successful physiological adaptation of our organism to different stressors.

In this system, in presence of a stressor or perturbating agent, inflammatory cytokines as IL1-beta, IL6 and TNF-α, several chemical substances, such as short chain fatty acids (SCFA), and microbial substances, can modify the ANS causing the secretion of cortisol and adrenaline from the adrenal glands through CRF and ACTH-dependent pathways[13].

Gut microbiota and neuroactive products (released from the enteroendocrine and immune system) impair intestinal secretion and compromise the integrity of intestinal mucosa in response to perturbating agents[13]. Therefore, several stressors render the intestinal mucosa more permeable to bacterial cytotoxins and neurotransmitters (norepinephrine), and cause the development of inflammation. In all these situations associated with stress, microbial agents translocate from the intestinal lumen into the systemic circulation, affecting the central and peripheral organs[13].

Efferent communication between the CNS and the gut mucosa include the vagal nerve and pelvic parasympathetic efferents and postganglionic sympathetic neurons[13,14].

Furthermore, the stress-induced activation of the HPA results in an enhanced production of corticosteroids from the adrenal glands[13,15,16]. Furthermore, the sympathetic autonomic system also becomes activated, thus leading to the secretion of catecholamines such as epinephrine and norepinephrine[13,17].

Gut microbiota and its effect on the gut- brain axis

Gut microbiota is one of the densest, and quickly developing bacterial ecosystems and is characterised by great biodiversity. The intestinal microbiome, which is its collective genome, is an adaptive entity that varies in response to food intake, daily habits and surroundings, providing the host with an extra metabolic plasticity as well as functions that humans have not developed[18,19].

Several experimental approaches have been used to study the modulatory effect of gut microbiota on gut-brain interactions, including gut microbial manipulation with antibiotics[20,21], fecal microbial transplantation[21,22], and the use of germ-free animal models[20].

For a recent overview of the complex mechanism of the microbiota-gut-brain axis, the reader is referred to the work by Carabotti et al[23]. In summary, microbiota modulation of the intestinal barrier is an important mechanism, with a net effect of strengthening tight junction integrity[23,24]. Gut microbiota is also involved in hippocampal neurogenesis and expression in hypothalamic genes involved in synaptic plasticity[25]. Furthermore, gut microbiota modulates pain perception and gut motility by acting on enteric sensory afferents[26] through the production of local neurotransmitters, such as gamma-aminobutyric acid, serotonin, nitric oxide, melatonin, catecholamine, histamine, acetylcholine and hydroxide sulfite[27-29], but also neurotrophic factors (brain-derived neurotrophic factor)[23]. Finally, microbiota affects the immune regulation of the intestinal mucosa by means of several mechanisms, such as the increase of substance P in the ENS, and the down-regulation of proteases[30,31], and it produces small molecules, such as SCFA involved in many neural processes, such as the stimulation of sympathetic activity and the release of serotonin[23].

On the other hand, gut microbiota is affected by the brain through the secretion of signaling molecules (i.e., catecholamines and gamma-aminobutyric acid) by neurons, immune and enterocromaffin cells, which may affect the microbiota itself[23]. Furthermore, controlled by the brain, also intestinal motility and permeability, as well as the production of mucus, acid, bicarbonates and fluids, are modulated through direct action (host-enteric microbiota signaling) and indirectly (by the actions of the HPA axis or by variations of the composition, and therefore of the function, of microbiota)[23,32].

WHAT IS THE CLINICAL SIGNIFICANCE OF THE CHANGES TO THE GUT-BRAIN-AXIS MICROBIOTA IN INFLAMMATORY BOWEL DISEASE?

IBD pathogenesis is only partly understood, and some studies highlight a link with intestinal microbiota. In particular, the bidirectional relationship of the gut-brain axis, seems to be central also in the onset and development of IBD.

How is dysbiosis involved in the pathogenesis of IBD?

Similar to IBS, there is an alteration of the balance between microbiota and the gastrointestinal tract, with the onset of dysbiosis[33,34]. The dysbiosis present in patients with IBD is characterized by reduced bacterial diversity, reduction of Bacteroidetes and Firmicutes, and an increase in Proteobacteria[35], in particular Escherichia coli[36]. Other species such as Faecalibacterium prausnitzii and Roseburia hominis are selectively reduced in patients with IBD[37,38]. Patients with active IBD have a lower abundance of intestinal flora than patients in remission[39]. An alteration of intestinal bacterial flora in genetically susceptible individuals can lead to abnormal intestinal immune responses and intestinal imbalance[40]. Interestingly, antibiotics may have some effect on IBD symptoms[41], and it is known that the predisposing genes involved in IBD pathogenesis are those with an important role in recognition of pathogen microbes[42] or in tolerance of commensals.

Furthermore, as discussed below, a confirmation of the influence of the microbiota on IBD pathogenesis is given by the benefits of using probiotics or prebiotics for these patients. The use of selected probiotics is effective in inducing and maintaining remission in ulcerative colitis (UC)[43] and in the treatment of pouchitis[44], whereas the use of prebiotics has been shown to be successful in reducing inflammation and bringing about remission in UC[45]. Data are far less impressive in Crohn's disease (CD)[46,47]. Faecal transplantation also appears to produce a modest increase in remission rates in patients with IBD[48].

How inflammation could modify the function of the gut-brain microbiota axis? Lessons from animal and human studies

IBD results in an inflammatory reaction in the brain, the activation of the hypothalamic-pituitary-adrenal axis and parts of the brain involved in behavioral alteration, an alteration of the blood-brain barrier and an intestinal-microbiota imbalance[49]. Pro-inflammatory cytokines play a crucial role in the IBD pathogenesis and interact with the CNS directly by means of the blood-brain barrier or via the vagus nerve[50]. Such inflammatory pathway via cytokines works by dysregulating HPA by over-activating microglia, altering neuroplasticity, and inducing structural and functional changes in the brain[49]. Several mouse studies showed that cytokines determine the activation of astrocytes, thus affecting neural functions during the processes of inflammation during the period between behavioral effects such as depression[51]. These in fact can affect the HPA axis, activating it and consequently causing an increase in glucocorticoids involved in pathogenesis of depression[52-54].

Other animal models have been used to investigate the relationship between gastrointestinal disorders and psychological manifestations. For example, maternal separation is a stressor induced in the early stages of life[55]. Other examples of chronic stressors are housing problems[56,57] and overcrowding[58], such stressors can induce gastrointestinal dysfunction or increase susceptibility to chemically induced colitis. These studies may explain the link between stress and gastrointestinal disorders and therefore IBS-like symptoms in patients with IBD, although not entirely specific to IBD.

In further animal studies, the mechanism of immune activation in the gut and its interaction with the central nervous system were explained[59,60].

There are several murine models of IBD, which include, for example, the use of dextran sodium sulfate (DSS), the colorectal instillation of dinitrobenzenic sulfonic acid and 2, 4, 6-trinitrobenzenesulfonic acid (TNBS)[61-67], that show an increase in hippocampal, cerebral and hypothalamic pro-inflammatory cytokines. Indeed, the disruption of the blood-brain barrier and the leukocyte infiltration of the brain following colic inflammation leaves the CNS vulnerable to inflammation mediators and to substances of bacterial and viral derivation[67].

What is the role of the stress, considered to be a perturbating agent of brain-gut-microbiota axis, in IBD?

Stress may have a deleterious effect on IBD, through several pathways, as reviewed by Bonaz et al[68], including the activation of mast cells and the CNS and the inhibition of the vagus nerve on inflammatory pathways, decreasing its anti-inflammatory effects and increasing sympathetic tone[69]; this may lead to the inhibition of immune defenses and development/increase of intestinal inflammation. It has been shown that stress induces an imbalance in the ANS in patients with IBD and a vagal dysfunction in patients with UC[68]. For patients with IBD there is a correlation between ANS imbalance, psychological disorders and pro-inflammatory profiles[70,71].

Important factors are also the effect of early childhood stressful events on colitis (the HPA axis is determined by early childhood events, and neonatal inflammatory stimuli exert long-term changes on HPA activity) and the impact of depression on exacerbation of colitis possibly through proinflammatory cytokines. Last, but not least, stress and CRF increase intestinal permeability as observed in mouse models, with passage of intestinal bacteria through the epithelial barrier[72,73]. In patients with IBD, changes in stress-mediated intestinal microbiota may create susceptibility toinfection and alter neural activity in stress-sensitive areas of the brain[74].

Dysbiosis can directly affect mental health in patients with IBD[75]. Behavioral disorders such as stress, anxiety and depression may change the composition of the intestinal flora and may influence the activity and recurrence of CD as demonstrated by numerous studies[76,77]. Recent studies have highlighted that the most involved mechanisms in stress signaling, on the cellular level, are endoplasmic reticulum stress, oxidative stress and hypoxia[78]. The subsequent host cellular response to these mechanisms interact with gut microbiota, thus modifying the microbiological microenvironment of the gastrointestinal tract[78].

Indeed, exposing mice to stressful stimuli resulted in the alteration of gut microbiota by reducing anti-inflammatory bacteria, in particular Lactobacillus[79-81] and Lachnospiraceae[79,82]. Furthermore, psychological stress reduces the biosynthesis and metabolism of short chain fatty acids, which may increase susceptibility toward intestinal inflammation and further IBD[79,83].

It is now known that IBD is associated with psychological symptoms such as anxiety and depression, prevalent during active disease states and, as also observed in animal models of IBD, there are no differences in occurrence of CD and UC. Mood disorders may also influence the course of IBD because it is hypothesized that stress may be a risk factor for recurrence for patients of this type[84]. Probably, the depressive behavior observed in patients with IBD constitutes a comorbidity[49], which worsens the state of intestinal disease[85].

In the murine models of DSS colitis, induction of depression with olfactory bulbectomy or intracerebroventricular injection of reserpine was associated with the reactivation of inflammation in mice with quiescent colitis, with effects mediated by the increase of pro-inflammatory cytokines[86]. In contrast, the administration of tricyclic antidepressants prevented the reactivation of colitis for depressed mice but not in mice without depression[87]. As previously mentioned, in many studies conducted on adults and children it is clear that both UC and CD are associated with a higher incidence of psychological symptoms[88], with an association between disease and mood. These data are confirmed by a recent systematic review, with equal rates in both sexes, but slightly higher for CD than for UC[89]. Depression and anxiety are 2 to 3 times higher in patients with IBD than in the general population, affecting respectively 25% and 30% of people with IBD[90]. Patients with IBD who suffer from psychiatric illness have a reduced chance of remission, and the condition worsens over a longer period of time[91]. An increased risk of psychiatric disorders is also observed in adolescents and children with IBD[92]. Adolescent patients with IBD are more likely to have mild behavioral and cognitive disturbances, particularly verbal memory loss[93]. Active IBD correlates significantly with increased psychological disorders[94,95] and the highest pain scores are strong predictors of depression in UC and CD[96]. In patients with UC, depression is usually diagnosed in the year before the onset of disease symptoms, while for patients with CD depression follows the diagnosis of the disease[97]. Being female is also a predictor of anxiety and depression with IBD[98,99], with IBD having a bigger effect on health-related quality of life[100] and greater concomitance of symptoms similar to IBS[101]. The manifestation of mood disorders by IBD patients correlates with a greater risk of requiring surgery and of incurring secondary FGID development[102].

In the Manitoba IBD study, a cohort of patients with IBD monitored every 6 mo, and with annual interviews over a period of 12 years, psychological disorders were highlighted as a major factor in health perception for the IBD cohort[103]. In another study involving 600 subjects with IBD, using health problem surveys conducted quarterly for 1 year, about 50% of patients showed a certain type of stress, most frequently family stress, followed by work or school and finance related stress[104]. It was observed that psychological factors, important life events, high anxiety and highly negative feelings during the previous 3 mo were closely associated with the occurrence of a flare up. Targownik et al[105] observed that perceived stress correlated closely with the symptoms of active disease, but without any correlation between the symptom scores and the degree of inflammation associated with CD, and only a weak correlation associated with UC, concluding that there may be an association between the perceived stress and the symptoms of IBD regardless of inflammatory activity.

Furthermore, IBD also influence the volume of gray matter and the size of the brain according to an MRI study, which observed that patients with CD displayed a decrease in the volume of gray matter in the frontal cortex and in the cortex of the anterior cingulate[106].

Interestingly, Gracie et al[107] recently found evidence in a 24 mo study of CD or UC patients of a reciprocal relationship between IBD occurence or severity and psychological illness, thus concluding that IBD patients’ psychological health should be monitored.

WHAT THERAPEUTIC INTERVENTIONS ARE THERE THAT TARGET THE GUT-BRAIN-MICROBIOTA AXIS WHEN IBD OCCURS?
Manipulation of gut microbiota

Since gut microflora may trigger changes leading to IBD, the manipulation of the microbiota through administration of probiotics, prebiotics, synbiotics, dietary modifications and faecal transplantation are potentially promising approaches to gastrointestinal diseases, including IBD[108-111].

The literature concerning the use of probiotics for the bringing about and preservation of CD remission is diverse in content and hard to interpret. The reasons for such heterogeneity are several: The different probiotics (strain and doses) used, the differences in study duration, the features of the included patients, and the measured endpoints[112]. Furthermore, two meta-analyses[112,113] on the effects of probiotics as a group indicated that their impact was no different from placebo.

With regard to UC, it was shown that the use of Escherichia coli Nissle 1917 strain for patients with UC was effective in maintaining remission as mesalazine therapy alone[113,114].

Successively, Shadnoush et al[115] showed that consuming yogurt containing Bifidobacterium and Lactobacillus may contribute to the maintenance of the homeostasis in gastrointestinal tract and regulate pro- and anti-inflammatory responses by the intestinal immune cells[113] and may therefore be advised for patients with active IBD[113,115].

Similarly, the consumption of Bifidobacteria-fermented milk was observed to exert a possible preventive effect on the recurrence of UC and helped to maintain its remission[113,116], while the use of combined treatment with Lactobacillus GG and mesalazine was found to be more effective in prolonging the relapse-free period than treatment with Lactobacillus GG and mesalazine alone for UC patients[113,116].

With regard to pouchitis, VSL3, a probiotic preparation containing 8 strains, when used for patients with pouchitis in small controlled trials and it was found to be beneficial for the primary and secondary prevention of pouchitis[113,117-120].

In contrast to the many studies on probiotics, there have been few studies (with conflicting results) that have addressed the role of prebiotics in encouraging the increase in number and/or activity of one or a limited class of bacteria in the gastrointestinal tract with a resulting improvement in the host’s IBD[113,119-121].

In these studies it was reported that increasing the SCFA concentration in the gut (as a result of the consumption of prebiotics) enhances growth of protective bacteria (symbionts), while limiting the growth of pathobionts[113,122-125].

With regards to synbiotics, they influence the development of beneficial intestinal microflora through the use of probiotics, whereas prebiotics inhibit the growth of pathogenic bacteria[113,126,127].

Finally, we move on to psychobiotics, which can be defined as live bacteria (probiotics) that “confer mental health benefits through interactions with commensal gut bacteria”[128]. Such probiotics have an effect on emotional, cognitive, systemic, and neural variables that determine psychological well-being[128]. Both rodents and human studies[129], as reviewed by Sarkar et al[129], showed the complex interactions between the modulation of gut microbiota with the gut-brain axis.

For IBD, data about the use of psychobiotics are limited to animal models, where they affected the gut-brain axis by modifying the immune system[129].

Other therapeutic interventions targeting the gut-brain axis in cases of inflammatory bowel diseases

Several factors interplay including triggering of the brain’s inflammation response system, the hypothalamic-pituitary-adrenal axis, and areas of the brain are associated with altered behaviour, changes in the integrity of the blood brain barrier, and an emerging role of gut microbiota and the response to probiotics in IBD cases. It is advised that IBD patients be monitored for psychological problems and treated appropriately as intervening can better quality of life and could diminish rates of relapse[130]. Several evidence-based treatments are available for most causes of psychological distress in IBD patients, the most widely accepted being rooted in cognitive behavioral theory[131].

Mindfulness interventions do not seem to impact disease activity and other disease-related factors in patients with UC or CD[132]. These findings are consistent with data from other psychosocial interventions[132-134] which did not demonstrate a positive impact on IBD clinical course. There is limited preliminary evidence that mindfulness interventions may have a positive impact on some inflammatory markers.

Patients want their gastroenterologist to discuss psychological issues during routine visits, and many are open to or desire referral to qualified mental health providers for concurrent treatment[131].

An alternative therapy to conventional anti-TNF-α treatment, based on gut-brain interactions, is stimulation of the cholinergic anti-inflammatory pathway, either pharmacologically or through vagal nerve stimulation or nutrition, as reviewed by Bonaz et al[70]. For example, galantamine is a centrally acting acetylcholinesterase inhibitor and a positive allosteric modulator of nicotinic receptors, including alpha7nAChR, which stimulates efferent VN activity, suppresses serum TNF-α and IL-6 levels[135]. In contrast, other drugs evaluated in this setting. i.e., CNI-1493, which is a tetravalent guanylhydrazone that inhibits production of proinflammatory cytokines in macrophages[136], and GTS-21, an alpha7nAChR agonist, were both associated with markedly reduced TNF-α, IL-6, and IL-1ra plasma concentrations[137]. Finally, another alpha7nAChR agonist, AR-R17779, has been used with success in a mouse model of postoperative ileus[138].

CONCLUSION

In conclusion, lines of communication between the brain and gut have a crucial role in the biology, clinical manifestations and clinical outcomes of gut diseases such as IBD. The central pathways through which this interplay is mediated are neural and immune networks, including interplay between these systems. The bidirectional trafficking of these signals opens the possibility of some gut diseases being instigated by aberrant brain function, and conversely, gut homeostasis disorder being responsible for instigating brain pathology and, particularly, mood disorders in other patients. Overall, the mucosal immune system is potentially the key gatekeeper of these pathways as it specifically interacts with nerves, the luminal microbiota and it is an important target of the stress response. What effect these insights will have on clinical practice remains to be clarified, although it is conceptually appealing to envisage the emergence of new diagnostic criteria, biomarkers and psychological scoring tools in patients with gut diseases. With the emergence of precision medicine, strategies in which patients with IBD might be classified beyond standard gut symptom complexes might offer the opportunity to tailor therapies to individual patients. In particular, anti-cytokine therapy or efforts to manipulate the intestinal microbiota could hold the key to effectively uncoupling pathological gut-brain interplay, with the potential to disrupt the proximal drivers of these important diseases. It seems to be with good reason that experts have said “as we enter a new era of patient-centered health care, treating the “brain” is as important as the “gut” for comprehensive, whole-person IBD management”[139].

Footnotes

Manuscript source: Invited manuscript

Specialty type: Medicine, research and experimental

Country of origin: Italy

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Tsujikawa T, Sirin G, Yuan JY S-Editor: Zhang L L-Editor: A E-Editor: Liu JH

References
1.  Schmidt C. Mental health: thinking from the gut. Nature. 2015;518:S12-S15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 106]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
2.  Smith PA. The tantalizing links between gut microbes and the brain. Nature. 2015;526:312-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 128]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
3.  Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K. Gut microbes and the brain: paradigm shift in neuroscience. J Neurosci. 2014;34:15490-15496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
4.  Wang HX, Wang YP. Gut Microbiota-brain Axis. Chin Med J (Engl). 2016;129:2373-2380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 229]  [Article Influence: 32.7]  [Reference Citation Analysis (0)]
5.  Simrén M, Barbara G, Flint HJ, Spiegel BM, Spiller RC, Vanner S, Verdu EF, Whorwell PJ, Zoetendal EG; Rome Foundation Committee. Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut. 2013;62:159-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 655]  [Cited by in F6Publishing: 608]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
6.  Mayer EA, Savidge T, Shulman RJ. Brain-gut microbiome interactions and functional bowel disorders. Gastroenterology. 2014;146:1500-1512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 289]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
7.  Ford AC, Quigley EM, Lacy BE, Lembo AJ, Saito YA, Schiller LR, Soffer EE, Spiegel BM, Moayyedi P. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol. 2014;109:1547-61; quiz 1546, 1562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 487]  [Cited by in F6Publishing: 447]  [Article Influence: 44.7]  [Reference Citation Analysis (0)]
8.  Bayliss WM, Starling EH. The movements and innervation of the small intestine. J Physiol. 1899;24:99-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 636]  [Cited by in F6Publishing: 583]  [Article Influence: 34.3]  [Reference Citation Analysis (0)]
9.  Furness JB. The enteric nervous system and neurogastroenterology. Nat Rev Gastroenterol Hepatol. 2012;9:286-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 856]  [Cited by in F6Publishing: 941]  [Article Influence: 78.4]  [Reference Citation Analysis (0)]
10.  Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol. 2009;6:306-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 760]  [Cited by in F6Publishing: 829]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
11.  El Aidy S, Dinan TG, Cryan JF. Gut Microbiota: The Conductor in the Orchestra of Immune-Neuroendocrine Communication. Clin Ther. 2015;37:954-967.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Pellissier S, Bonaz B. The Place of Stress and Emotions in the Irritable Bowel Syndrome. Vitam Horm. 2017;103:327-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
13.  Brzozowski B, Mazur-Bialy A, Pajdo R, Kwiecien S, Bilski J, Zwolinska-Wcislo M, Mach T, Brzozowski T. Mechanisms by which Stress Affects the Experimental and Clinical Inflammatory Bowel Disease (IBD): Role of Brain-Gut Axis. Curr Neuropharmacol. 2016;14:892-900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 92]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
14.  Keightley PC, Koloski NA, Talley NJ. Pathways in gut-brain communication: evidence for distinct gut-to-brain and brain-to-gut syndromes. Aust N Z J Psychiatry. 2015;49:207-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
15.  Israeli E, Hershcovici T, Berenshtein E, Zannineli G, Wengrower D, Weiss O, Chevion M, Goldin E. The effect of restraint stress on the normal colon and on intestinal inflammation in a model of experimental colitis. Dig Dis Sci. 2008;53:88-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
16.  Chowdrey HS, Jessop DS, Lightman SL. Substance P stimulates arginine vasopressin and inhibits adrenocorticotropin release in vivo in the rat. Neuroendocrinology. 1990;52:90-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 53]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
17.  Söderholm JD. Effect of stress on intestinal mucosal functions. In: Johnson LR, Ghishan FK, Kaunitz JD, Merchant JL, Said HM, Wood JD. Physiology of the Gastrointestinal Tract. 5th ed. In: Elsevier Inc 2012; 1979-2000.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Sinagra E, Morreale GC, Mohammadian G, Fusco G, Guarnotta V, Tomasello G, Cappello F, Rossi F, Amvrosiadis G, Raimondo D. New therapeutic perspectives in irritable bowel syndrome: Targeting low-grade inflammation, immuno-neuroendocrine axis, motility, secretion and beyond. World J Gastroenterol. 2017;23:6593-6627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 32]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (2)]
19.  Ash C, Mueller K. Manipulating the Microbiota. Science. 2016;352:530-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
20.  Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest. 2015;125:926-938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 774]  [Cited by in F6Publishing: 867]  [Article Influence: 96.3]  [Reference Citation Analysis (0)]
21.  Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y, Blennerhassett P, Macri J, McCoy KD, Verdu EF, Collins SM. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141:599-609, 609.e1-609.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1135]  [Cited by in F6Publishing: 1113]  [Article Influence: 85.6]  [Reference Citation Analysis (0)]
22.  Collins SM, Kassam Z, Bercik P. The adoptive transfer of behavioral phenotype via the intestinal microbiota: experimental evidence and clinical implications. Curr Opin Microbiol. 2013;16:240-245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 140]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
23.  Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28:203-209.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Ait-Belgnaoui A, Colom A, Braniste V, Ramalho L, Marrot A, Cartier C, Houdeau E, Theodorou V, Tompkins T. Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterol Motil. 2014;26:510-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 278]  [Article Influence: 27.8]  [Reference Citation Analysis (0)]
25.  Kunze WA, Mao YK, Wang B, Huizinga JD, Ma X, Forsythe P, Bienenstock J. Lactobacillus reuteri enhances excitability of colonic AH neurons by inhibiting calcium-dependent potassium channel opening. J Cell Mol Med. 2009;13:2261-2270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 242]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
26.  Iyer LM, Aravind L, Coon SL, Klein DC, Koonin EV. Evolution of cell-cell signaling in animals: did late horizontal gene transfer from bacteria have a role? Trends Genet. 2004;20:292-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 145]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
27.  Asano Y, Hiramoto T, Nishino R, Aiba Y, Kimura T, Yoshihara K, Koga Y, Sudo N. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am J Physiol Gastrointest Liver Physiol. 2012;303:G1288-G1295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 389]  [Cited by in F6Publishing: 393]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
28.  Sobko T, Huang L, Midtvedt T, Norin E, Gustafsson LE, Norman M, Jansson EA, Lundberg JO. Generation of NO by probiotic bacteria in the gastrointestinal tract. Free Radic Biol Med. 2006;41:985-991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 83]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
29.  Verdú EF, Bercik P, Verma-Gandhu M, Huang XX, Blennerhassett P, Jackson W, Mao Y, Wang L, Rochat F, Collins SM. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut. 2006;55:182-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 319]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
30.  Saito T, Bunnett NW. Protease-activated receptors: regulation of neuronal function. Neuromolecular Med. 2005;7:79-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
31.  Biancheri P, Di Sabatino A, Corazza GR, MacDonald TT. Proteases and the gut barrier. Cell Tissue Res. 2013;351:269-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
32.  González-Arancibia C, Escobar-Luna J, Barrera-Bugueño C, Díaz-Zepeda C, González-Toro MP, Olavarría-Ramírez L, Zanelli-Massai F, Gotteland M, Bravo JA, Julio-Pieper M. What goes around comes around: novel pharmacological targets in the gut-brain axis. Therap Adv Gastroenterol. 2016;9:339-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
33.  Collins SM, Bercik P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology. 2009;136:2003-2014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 386]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
34.  Lepage P, Colombet J, Marteau P, Sime-Ngando T, Doré J, Leclerc M. Dysbiosis in inflammatory bowel disease: a role for bacteriophages? Gut. 2008;57:424-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 156]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
35.  Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci USA. 2007;104:13780-13785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3075]  [Cited by in F6Publishing: 3234]  [Article Influence: 190.2]  [Reference Citation Analysis (1)]
36.  Chassaing B, Rolhion N, de Vallée A, Salim SY, Prorok-Hamon M, Neut C, Campbell BJ, Söderholm JD, Hugot JP, Colombel JF, Darfeuille-Michaud A. Crohn disease--associated adherent-invasive E. coli bacteria target mouse and human Peyer's patches via long polar fimbriae. J Clin Invest. 2011;121:966-975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 191]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
37.  Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán LG, Gratadoux JJ, Blugeon S, Bridonneau C, Furet JP, Corthier G, Grangette C, Vasquez N, Pochart P, Trugnan G, Thomas G, Blottière HM, Doré J, Marteau P, Seksik P, Langella P. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci USA. 2008;105:16731-16736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2747]  [Cited by in F6Publishing: 2946]  [Article Influence: 184.1]  [Reference Citation Analysis (0)]
38.  Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, Ballet V, Claes K, Van Immerseel F, Verbeke K, Ferrante M, Verhaegen J, Rutgeerts P, Vermeire S. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63:1275-1283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1036]  [Cited by in F6Publishing: 1170]  [Article Influence: 117.0]  [Reference Citation Analysis (2)]
39.  Prosberg M, Bendtsen F, Vind I, Petersen AM, Gluud LL. The association between the gut microbiota and the inflammatory bowel disease activity: a systematic review and meta-analysis. Scand J Gastroenterol. 2016;51:1407-1415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 136]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
40.  Boyapati R, Satsangi J, Ho GT. Pathogenesis of Crohn's disease. F1000Prime Rep. 2015;7:44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 58]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
41.  Nitzan O, Elias M, Peretz A, Saliba W. Role of antibiotics for treatment of inflammatory bowel disease. World J Gastroenterol. 2016;22:1078-1087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 161]  [Cited by in F6Publishing: 154]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
42.  Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, Lee JC, Schumm LP, Sharma Y, Anderson CA, Essers J, Mitrovic M, Ning K, Cleynen I, Theatre E, Spain SL, Raychaudhuri S, Goyette P, Wei Z, Abraham C, Achkar JP, Ahmad T, Amininejad L, Ananthakrishnan AN, Andersen V, Andrews JM, Baidoo L, Balschun T, Bampton PA, Bitton A, Boucher G, Brand S, Büning C, Cohain A, Cichon S, D'Amato M, De Jong D, Devaney KL, Dubinsky M, Edwards C, Ellinghaus D, Ferguson LR, Franchimont D, Fransen K, Gearry R, Georges M, Gieger C, Glas J, Haritunians T, Hart A, Hawkey C, Hedl M, Hu X, Karlsen TH, Kupcinskas L, Kugathasan S, Latiano A, Laukens D, Lawrance IC, Lees CW, Louis E, Mahy G, Mansfield J, Morgan AR, Mowat C, Newman W, Palmieri O, Ponsioen CY, Potocnik U, Prescott NJ, Regueiro M, Rotter JI, Russell RK, Sanderson JD, Sans M, Satsangi J, Schreiber S, Simms LA, Sventoraityte J, Targan SR, Taylor KD, Tremelling M, Verspaget HW, De Vos M, Wijmenga C, Wilson DC, Winkelmann J, Xavier RJ, Zeissig S, Zhang B, Zhang CK, Zhao H; International IBD Genetics Consortium (IIBDGC), Silverberg MS, Annese V, Hakonarson H, Brant SR, Radford-Smith G, Mathew CG, Rioux JD, Schadt EE, Daly MJ, Franke A, Parkes M, Vermeire S, Barrett JC, Cho JH. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3733]  [Cited by in F6Publishing: 3357]  [Article Influence: 279.8]  [Reference Citation Analysis (0)]
43.  Mallon P, McKay D, Kirk S, Gardiner K. Probiotics for induction of remission in ulcerative colitis. Cochrane Database Syst Rev. 2007;CD005573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 80]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
44.  Lichtenstein L, Avni-Biron I, Ben-Bassat O. The current place of probiotics and prebiotics in the treatment of pouchitis. Best Pract Res Clin Gastroenterol. 2016;30:73-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
45.  Casellas F, Borruel N, Torrejón A, Varela E, Antolin M, Guarner F, Malagelada JR. Oral oligofructose-enriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin. Aliment Pharmacol Ther. 2007;25:1061-1067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 153]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
46.  Benjamin JL, Hedin CR, Koutsoumpas A, Ng SC, McCarthy NE, Hart AL, Kamm MA, Sanderson JD, Knight SC, Forbes A, Stagg AJ, Whelan K, Lindsay JO. Randomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn's disease. Gut. 2011;60:923-929.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 231]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
47.  Lindsay JO, Whelan K, Stagg AJ, Gobin P, Al-Hassi HO, Rayment N, Kamm MA, Knight SC, Forbes A. Clinical, microbiological, and immunological effects of fructo-oligosaccharide in patients with Crohn's disease. Gut. 2006;55:348-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 263]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
48.  Moayyedi P, Surette MG, Kim PT, Libertucci J, Wolfe M, Onischi C, Armstrong D, Marshall JK, Kassam Z, Reinisch W, Lee CH. Fecal Microbiota Transplantation Induces Remission in Patients With Active Ulcerative Colitis in a Randomized Controlled Trial. Gastroenterology. 2015;149:102-109.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 930]  [Cited by in F6Publishing: 965]  [Article Influence: 107.2]  [Reference Citation Analysis (0)]
49.  Maes M, Berk M, Goehler L, Song C, Anderson G, Gałecki P, Leonard B. Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Med. 2012;10:66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 398]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
50.  Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol. 2014;14:329-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1545]  [Cited by in F6Publishing: 1726]  [Article Influence: 172.6]  [Reference Citation Analysis (1)]
51.  Sofroniew MV. Multiple roles for astrocytes as effectors of cytokines and inflammatory mediators. Neuroscientist. 2014;20:160-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 238]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
52.  Dunn AJ. Cytokine activation of the HPA axis. Ann N Y Acad Sci. 2000;917:608-617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 223]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
53.  Miller AH, Pariante CM, Pearce BD. Effects of cytokines on glucocorticoid receptor expression and function. Glucocorticoid resistance and relevance to depression. Adv Exp Med Biol. 1999;461:107-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 142]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
54.  Pariante CM, Lightman SL. The HPA axis in major depression: classical theories and new developments. Trends Neurosci. 2008;31:464-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1187]  [Cited by in F6Publishing: 1254]  [Article Influence: 78.4]  [Reference Citation Analysis (0)]
55.  O'Mahony SM, Clarke G, Dinan TG, Cryan JF. Early-life adversity and brain development: Is the microbiome a missing piece of the puzzle? Neuroscience. 2017;342:37-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 124]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
56.  Reber SO, Birkeneder L, Veenema AH, Obermeier F, Falk W, Straub RH, Neumann ID. Adrenal insufficiency and colonic inflammation after a novel chronic psycho-social stress paradigm in mice: implications and mechanisms. Endocrinology. 2007;148:670-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 158]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
57.  Reber SO, Obermeier F, Straub RH, Veenema AH, Neumann ID. Aggravation of DSS-induced colitis after chronic subordinate colony (CSC) housing is partially mediated by adrenal mechanisms. Stress. 2008;11:225-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
58.  Vicario M, Guilarte M, Alonso C, Yang P, Martínez C, Ramos L, Lobo B, González A, Guilà M, Pigrau M, Saperas E, Azpiroz F, Santos J. Chronological assessment of mast cell-mediated gut dysfunction and mucosal inflammation in a rat model of chronic psychosocial stress. Brain Behav Immun. 2010;24:1166-1175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 82]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
59.  Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol. 2003;3:521-533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1309]  [Cited by in F6Publishing: 1314]  [Article Influence: 62.6]  [Reference Citation Analysis (0)]
60.  Hathaway CA, Appleyard CB, Percy WH, Williams JL. Experimental colitis increases blood-brain barrier permeability in rabbits. Am J Physiol. 1999;276:G1174-G1180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 22]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
61.  Natah SS, Mouihate A, Pittman QJ, Sharkey KA. Disruption of the blood-brain barrier during TNBS colitis. Neurogastroenterol Motil. 2005;17:433-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 60]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
62.  Riazi K, Galic MA, Kuzmiski JB, Ho W, Sharkey KA, Pittman QJ. Microglial activation and TNFalpha production mediate altered CNS excitability following peripheral inflammation. Proc Natl Acad Sci USA. 2008;105:17151-17156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 309]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
63.  Medhi B, Prakash A, Avti PK, Chakrabarti A, Khanduja KL. Intestinal inflammation and seizure susceptibility: understanding the role of tumour necrosis factor-alpha in a rat model. J Pharm Pharmacol. 2009;61:1359-1364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
64.  Wang K, Yuan CP, Wang W, Yang ZQ, Cui W, Mu LZ, Yue ZP, Yin XL, Hu ZM, Liu JX. Expression of interleukin 6 in brain and colon of rats with TNBS-induced colitis. World J Gastroenterol. 2010;16:2252-2259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
65.  Baticic L, Detel D, Kucic N, Buljevic S, Pugel EP, Varljen J. Neuroimmunomodulative properties of dipeptidyl peptidase IV/CD26 in a TNBS-induced model of colitis in mice. J Cell Biochem. 2011;112:3322-3333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
66.  Alhouayek M, Lambert DM, Delzenne NM, Cani PD, Muccioli GG. Increasing endogenous 2-arachidonoylglycerol levels counteracts colitis and related systemic inflammation. FASEB J. 2011;25:2711-2721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 160]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
67.  Sans M, Kawachi S, Soriano A, Palacín A, Morise Z, Granger DN, Piqué JM, Grisham MB, Panés J. Brain endothelial adhesion molecule expression in experimental colitis. Microcirculation. 2001;8:105-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
68.  Bonaz BL, Bernstein CN. Brain-gut interactions in inflammatory bowel disease. Gastroenterology. 2013;144:36-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 423]  [Cited by in F6Publishing: 440]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
69.  Taché Y, Bonaz B. Corticotropin-releasing factor receptors and stress-related alterations of gut motor function. J Clin Invest. 2007;117:33-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 249]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
70.  Bonaz B, Bazin T, Pellissier S. The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis. Front Neurosci. 2018;12:49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 425]  [Cited by in F6Publishing: 621]  [Article Influence: 103.5]  [Reference Citation Analysis (0)]
71.  Santos J, Saunders PR, Hanssen NP, Yang PC, Yates D, Groot JA, Perdue MH. Corticotropin-releasing hormone mimics stress-induced colonic epithelial pathophysiology in the rat. Am J Physiol. 1999;277:G391-G399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 86]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
72.  Bailey MT, Engler H, Sheridan JF. Stress induces the translocation of cutaneous and gastrointestinal microflora to secondary lymphoid organs of C57BL/6 mice. J Neuroimmunol. 2006;171:29-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 90]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
73.  Lyte M, Li W, Opitz N, Gaykema RP, Goehler LE. Induction of anxiety-like behavior in mice during the initial stages of infection with the agent of murine colonic hyperplasia Citrobacter rodentium. Physiol Behav. 2006;89:350-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 231]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
74.  Vidal A, Gómez-Gil E, Sans M, Portella MJ, Salamero M, Piqué JM, Panés J. Health-related quality of life in inflammatory bowel disease patients: the role of psychopathology and personality. Inflamm Bowel Dis. 2008;14:977-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 67]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
75.  Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, Hibberd ML, Forssberg H, Pettersson S. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci USA. 2011;108:3047-3052.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1983]  [Cited by in F6Publishing: 2092]  [Article Influence: 160.9]  [Reference Citation Analysis (0)]
76.  Ananthakrishnan AN, Khalili H, Pan A, Higuchi LM, de Silva P, Richter JM, Fuchs CS, Chan AT. Association between depressive symptoms and incidence of Crohn's disease and ulcerative colitis: results from the Nurses' Health Study. Clin Gastroenterol Hepatol. 2013;11:57-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 98]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
77.  Million M, Taché Y, Anton P. Susceptibility of Lewis and Fischer rats to stress-induced worsening of TNB-colitis: protective role of brain CRF. Am J Physiol. 1999;276:G1027-G1036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 45]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
78.  Cao SS. Cellular Stress Responses and Gut Microbiota in Inflammatory Bowel Disease. Gastroenterol Res Pract. 2018;2018:7192646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
79.  Lobionda S, Sittipo P, Kwon HY, Lee YK. The Role of Gut Microbiota in Intestinal Inflammation with Respect to Diet and Extrinsic Stressors. Microorganisms. 2019;7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 163]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
80.  Galley JD, Mackos AR, Varaljay VA, Bailey MT. Stressor exposure has prolonged effects on colonic microbial community structure in Citrobacter rodentium-challenged mice. Sci Rep. 2017;7:45012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
81.  Mackos AR, Eubank TD, Parry NM, Bailey MT. Probiotic Lactobacillus reuteri attenuates the stressor-enhanced severity of Citrobacter rodentium infection. Infect Immun. 2013;81:3253-3263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
82.  Bharwani A, Mian MF, Foster JA, Surette MG, Bienenstock J, Forsythe P. Structural & functional consequences of chronic psychosocial stress on the microbiome & host. Psychoneuroendocrinology. 2016;63:217-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 199]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
83.  Gao X, Cao Q, Cheng Y, Zhao D, Wang Z, Yang H, Wu Q, You L, Wang Y, Lin Y, Li X, Wang Y, Bian JS, Sun D, Kong L, Birnbaumer L, Yang Y. Chronic stress promotes colitis by disturbing the gut microbiota and triggering immune system response. Proc Natl Acad Sci USA. 2018;115:E2960-E2969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 232]  [Article Influence: 38.7]  [Reference Citation Analysis (0)]
84.  Duffy LC, Zielezny MA, Marshall JR, Byers TE, Weiser MM, Phillips JF, Calkins BM, Ogra PL, Graham S. Relevance of major stress events as an indicator of disease activity prevalence in inflammatory bowel disease. Behav Med. 1991;17:101-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 123]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
85.  Szigethy EM, Youk AO, Benhayon D, Fairclough DL, Newara MC, Kirshner MA, Bujoreanu SI, Mrakotsky C, Bousvaros A, Srinath AI, Keljo DJ, Kupfer DJ, DeMaso DR. Depression subtypes in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2014;58:574-581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
86.  Ghia JE, Blennerhassett P, Deng Y, Verdu EF, Khan WI, Collins SM. Reactivation of inflammatory bowel disease in a mouse model of depression. Gastroenterology. 2009;136:2280-2288.e1-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 116]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
87.  Piechota-Polanczyk A, Fichna J. Review article: the role of oxidative stress in pathogenesis and treatment of inflammatory bowel diseases. Naunyn Schmiedebergs Arch Pharmacol. 2014;387:605-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 255]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
88.  Farrokhyar F, Marshall JK, Easterbrook B, Irvine EJ. Functional gastrointestinal disorders and mood disorders in patients with inactive inflammatory bowel disease: prevalence and impact on health. Inflamm Bowel Dis. 2006;12:38-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 176]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
89.  Mikocka-Walus A, Knowles SR, Keefer L, Graff L. Controversies Revisited: A Systematic Review of the Comorbidity of Depression and Anxiety with Inflammatory Bowel Diseases. Inflamm Bowel Dis. 2016;22:752-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 318]  [Cited by in F6Publishing: 359]  [Article Influence: 44.9]  [Reference Citation Analysis (0)]
90.  Walker JR, Ediger JP, Graff LA, Greenfeld JM, Clara I, Lix L, Rawsthorne P, Miller N, Rogala L, McPhail CM, Bernstein CN. The Manitoba IBD cohort study: a population-based study of the prevalence of lifetime and 12-month anxiety and mood disorders. Am J Gastroenterol. 2008;103:1989-1997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 315]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
91.  Stasi C, Orlandelli E. Role of the brain-gut axis in the pathophysiology of Crohn's disease. Dig Dis. 2008;26:156-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
92.  Loftus EV, Guérin A, Yu AP, Wu EQ, Yang M, Chao J, Mulani PM. Increased risks of developing anxiety and depression in young patients with Crohn's disease. Am J Gastroenterol. 2011;106:1670-1677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 93]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
93.  Filipovic BR, Filipovic BF. Psychiatric comorbidity in the treatment of patients with inflammatory bowel disease. World J Gastroenterol. 2014;20:3552-3563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 65]  [Cited by in F6Publishing: 63]  [Article Influence: 6.3]  [Reference Citation Analysis (1)]
94.  Long MD, Kappelman MD, Martin CF, Chen W, Anton K, Sandler RS. Risk factors for depression in the elderly inflammatory bowel disease population. J Crohns Colitis. 2014;8:113-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 36]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
95.  Clark JG, Srinath AI, Youk AO, Kirshner MA, McCarthy FN, Keljo DJ, Bousvaros A, DeMaso DR, Szigethy EM. Predictors of depression in youth with Crohn disease. J Pediatr Gastroenterol Nutr. 2014;58:569-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
96.  Deberry JJ, Bielefeldt K, Davis BM, Szigethy EM, Hartman DJ, Coates MD. Abdominal pain and the neurotrophic system in ulcerative colitis. Inflamm Bowel Dis. 2014;20:2330-2339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
97.  Kurina LM, Goldacre MJ, Yeates D, Gill LE. Depression and anxiety in people with inflammatory bowel disease. J Epidemiol Community Health. 2001;55:716-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 229]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
98.  Ennaifer R, Elleuch N, Cheikh M, Hefaiedh R, Romdhane H, Ben Nejma H, Belhadj N. Risk factors of psychological disorders in inflammatory bowel disease in a tunisian survey. Results of a cross-sectional study. Tunis Med. 2014;92:723-726.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Panara AJ, Yarur AJ, Rieders B, Proksell S, Deshpande AR, Abreu MT, Sussman DA. The incidence and risk factors for developing depression after being diagnosed with inflammatory bowel disease: a cohort study. Aliment Pharmacol Ther. 2014;39:802-810.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 97]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
100.  Casellas F, López-Vivancos J, Casado A, Malagelada JR. Factors affecting health related quality of life of patients with inflammatory bowel disease. Qual Life Res. 2002;11:775-781.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Berrill JW, Green JT, Hood K, Campbell AK. Symptoms of irritable bowel syndrome in patients with inflammatory bowel disease: examining the role of sub-clinical inflammation and the impact on clinical assessment of disease activity. Aliment Pharmacol Ther. 2013;38:44-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
102.  Cámara RJ, Ziegler R, Begré S, Schoepfer AM, von Känel R; Swiss Inflammatory Bowel Disease Cohort Study (SIBDCS) group. The role of psychological stress in inflammatory bowel disease: quality assessment of methods of 18 prospective studies and suggestions for future research. Digestion. 2009;80:129-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 72]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
103.  Graff LA, Walker JR, Clara I, Lix L, Miller N, Rogala L, Rawsthorne P, Bernstein CN. Stress coping, distress, and health perceptions in inflammatory bowel disease and community controls. Am J Gastroenterol. 2009;104:2959-2969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 77]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
104.  Bernstein CN, Singh S, Graff LA, Walker JR, Miller N, Cheang M. A prospective population-based study of triggers of symptomatic flares in IBD. Am J Gastroenterol. 2010;105:1994-2002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 277]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
105.  Targownik LE, Sexton KA, Bernstein MT, Beatie B, Sargent M, Walker JR, Graff LA. The Relationship Among Perceived Stress, Symptoms, and Inflammation in Persons With Inflammatory Bowel Disease. Am J Gastroenterol. 2015;110:1001-12; quiz 1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 112]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
106.  Agostini A, Filippini N, Benuzzi F, Bertani A, Scarcelli A, Leoni C, Farinelli V, Riso D, Tambasco R, Calabrese C, Rizzello F, Gionchetti P, Ercolani M, Nichelli P, Campieri M. Functional magnetic resonance imaging study reveals differences in the habituation to psychological stress in patients with Crohn's disease versus healthy controls. J Behav Med. 2013;36:477-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
107.  Gracie DJ, Guthrie EA, Hamlin PJ, Ford AC. Bi-directionality of Brain-Gut Interactions in Patients With Inflammatory Bowel Disease. Gastroenterology. 2018;154:1635-1646.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 240]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
108.  Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G, Quraishi MN, Kinross J, Smidt H, Tuohy KM, Thomas LV, Zoetendal EG, Hart A. The gut microbiota and host health: a new clinical frontier. Gut. 2016;65:330-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1341]  [Cited by in F6Publishing: 1385]  [Article Influence: 173.1]  [Reference Citation Analysis (0)]
109.  Moran C, Shanahan F. Editorial: probiotics and IBS - where are we now? Aliment Pharmacol Ther. 2014;40:318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
110.  Pinn DM, Aroniadis OC, Brandt LJ. Is fecal microbiota transplantation (FMT) an effective treatment for patients with functional gastrointestinal disorders (FGID)? Neurogastroenterol Motil. 2015;27:19-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
111.  Sinagra E, Tomasello G, Cappello F, Leone A, Cottone M, Bellavia M, Rossi F, Facella T, Damiani P, Zeenny MN, Damiani F, Abruzzo A, Damiano G, Palumbo VD, Cocchi M, Jurjus A, Spinelli G, Lo Monte AI, Raimondo D. Probiotics, prebiotics and symbiotics in inflammatory bowel diseases: state-of-the-art and new insights. J Biol Regul Homeost Agents. 2013;27:919-933.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Doherty G, Bennett G, Patil S, Cheifetz A, Moss AC. Interventions for prevention of post-operative recurrence of Crohn's disease. Cochrane Database Syst Rev. 2009;CD006873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 95]  [Article Influence: 6.3]  [Reference Citation Analysis (1)]
113.  Wasilewski A, Zielińska M, Storr M, Fichna J. Beneficial Effects of Probiotics, Prebiotics, Synbiotics, and Psychobiotics in Inflammatory Bowel Disease. Inflamm Bowel Dis. 2015;21:1674-1682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 115]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
114.  Kruis W, Fric P, Pokrotnieks J, Lukás M, Fixa B, Kascák M, Kamm MA, Weismueller J, Beglinger C, Stolte M, Wolff C, Schulze J. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut. 2004;53:1617-1623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 851]  [Cited by in F6Publishing: 771]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
115.  Shadnoush M, Shaker Hosseini R, Mehrabi Y, Delpisheh A, Alipoor E, Faghfoori Z, Mohammadpour N, Zaringhalam Moghadam J. Probiotic yogurt Affects Pro- and Anti-inflammatory Factors in Patients with Inflammatory Bowel Disease. Iran J Pharm Res. 2013;12:929-936.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Ishikawa H, Akedo I, Umesaki Y, Tanaka R, Imaoka A, Otani T. Randomized controlled trial of the effect of bifidobacteria-fermented milk on ulcerative colitis. J Am Coll Nutr. 2003;22:56-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 207]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
117.  Zocco MA, dal Verme LZ, Cremonini F, Piscaglia AC, Nista EC, Candelli M, Novi M, Rigante D, Cazzato IA, Ojetti V, Armuzzi A, Gasbarrini G, Gasbarrini A. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther. 2006;23:1567-1574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 283]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
118.  Mimura T, Rizzello F, Helwig U, Poggioli G, Schreiber S, Talbot IC, Nicholls RJ, Gionchetti P, Campieri M, Kamm MA. Once daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis. Gut. 2004;53:108-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 660]  [Cited by in F6Publishing: 578]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
119.  Kuisma J, Mentula S, Jarvinen H, Kahri A, Saxelin M, Farkkila M. Effect of Lactobacillus rhamnosus GG on ileal pouch inflammation and microbial flora. Aliment Pharmacol Ther. 2003;17:509-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 166]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
120.  Gionchetti P, Rizzello F, Venturi A, Brigidi P, Matteuzzi D, Bazzocchi G, Poggioli G, Miglioli M, Campieri M. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology. 2000;119:305-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1077]  [Cited by in F6Publishing: 919]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
121.  Gionchetti P, Rizzello F, Helwig U, Venturi A, Lammers KM, Brigidi P, Vitali B, Poggioli G, Miglioli M, Campieri M. Prophylaxis of pouchitis onset with probiotic therapy: a double-blind, placebo-controlled trial. Gastroenterology. 2003;124:1202-1209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 807]  [Cited by in F6Publishing: 673]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
122.  Gibson GR, Beatty ER, Wang X, Cummings JH. Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin. Gastroenterology. 1995;108:975-982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1062]  [Cited by in F6Publishing: 872]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
123.  Muccioli GG, Naslain D, Bäckhed F, Reigstad CS, Lambert DM, Delzenne NM, Cani PD. The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol. 2010;6:392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 447]  [Cited by in F6Publishing: 471]  [Article Influence: 33.6]  [Reference Citation Analysis (0)]
124.  Looijer-van Langen MA, Dieleman LA. Prebiotics in chronic intestinal inflammation. Inflamm Bowel Dis. 2009;15:454-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 114]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
125.  Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2937]  [Cited by in F6Publishing: 3472]  [Article Influence: 315.6]  [Reference Citation Analysis (0)]
126.  Furrie E, Macfarlane S, Kennedy A, Cummings JH, Walsh SV, O'neil DA, Macfarlane GT. Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial. Gut. 2005;54:242-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 547]  [Cited by in F6Publishing: 428]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
127.  Steed H, Macfarlane GT, Blackett KL, Bahrami B, Reynolds N, Walsh SV, Cummings JH, Macfarlane S. Clinical trial: the microbiological and immunological effects of synbiotic consumption - a randomized double-blind placebo-controlled study in active Crohn's disease. Aliment Pharmacol Ther. 2010;32:872-883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 135]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
128.  Dinan TG, Stanton C, Cryan JF. Psychobiotics: a novel class of psychotropic. Biol Psychiatry. 2013;74:720-726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 719]  [Cited by in F6Publishing: 719]  [Article Influence: 65.4]  [Reference Citation Analysis (0)]
129.  Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, Burnet PWJ. Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals. Trends Neurosci. 2016;39:763-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 485]  [Cited by in F6Publishing: 536]  [Article Influence: 67.0]  [Reference Citation Analysis (0)]
130.  Abautret-Daly Á, Dempsey E, Parra-Blanco A, Medina C, Harkin A. Gut-brain actions underlying comorbid anxiety and depression associated with inflammatory bowel disease. Acta Neuropsychiatr. 2018;30:275-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 99]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
131.  Taft TH, Ballou S, Bedell A, Lincenberg D. Psychological Considerations and Interventions in Inflammatory Bowel Disease Patient Care. Gastroenterol Clin North Am. 2017;46:847-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
132.  Hood MM, Jedel S. Mindfulness-Based Interventions in Inflammatory Bowel Disease. Gastroenterol Clin North Am. 2017;46:859-874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 87]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
133.  von Wietersheim J, Kessler H. Psychotherapy with chronic inflammatory bowel disease patients: a review. Inflamm Bowel Dis. 2006;12:1175-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 63]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
134.  Mikocka-Walus A, Bampton P, Hetzel D, Hughes P, Esterman A, Andrews JM. Cognitive-Behavioural Therapy for Inflammatory Bowel Disease: 24-Month Data from a Randomised Controlled Trial. Int J Behav Med. 2017;24:127-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
135.  Pavlov VA, Parrish WR, Rosas-Ballina M, Ochani M, Puerta M, Ochani K, Chavan S, Al-Abed Y, Tracey KJ. Brain acetylcholinesterase activity controls systemic cytokine levels through the cholinergic anti-inflammatory pathway. Brain Behav Immun. 2009;23:41-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 321]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
136.  Tracey KJ. Suppression of TNF and other proinflammatory cytokines by the tetravalent guanylhydrazone CNI-1493. Prog Clin Biol Res. 1998;397:335-343.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Kox M, Pompe JC, Gordinou de Gouberville MC, van der Hoeven JG, Hoedemaekers CW, Pickkers P. Effects of the α7 nicotinic acetylcholine receptor agonist GTS-21 on the innate immune response in humans. Shock. 2011;36:5-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 62]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
138.  The FO, Boeckxstaens GE, Snoek SA, Cash JL, Bennink R, Larosa GJ, van den Wijngaard RM, Greaves DR, de Jonge WJ. Activation of the cholinergic anti-inflammatory pathway ameliorates postoperative ileus in mice. Gastroenterology. 2007;133:1219-1228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 173]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
139.  Click BH, Greer JB, Regueiro MD, Hartman DJ, Davis PL, Siegel CA, Herfarth HH, Rosh JR, Shah SA, Koltun WA, Binion DG, Baidoo L, Szigethy E. IBD LIVE Series-Case 7: The Brain-Gut Connection and the Importance of Integrated Care in IBD. Inflamm Bowel Dis. 2017;23:681-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]