2,990
Views
5
CrossRef citations to date
0
Altmetric
Review

Fungal feelings in the irritable bowel syndrome: the intestinal mycobiome and abdominal pain

, & ORCID Icon
Article: 2168992 | Received 17 Oct 2022, Accepted 10 Jan 2023, Published online: 01 Feb 2023

ABSTRACT

Although the gut microbiota consists of bacteria, viruses, and fungi, most publications addressing the microbiota-gut-brain axis in irritable bowel syndrome (IBS) have a sole focus on bacteria. This may relate to the relatively low presence of fungi and viruses as compared to bacteria. Yet, in the field of inflammatory bowel disease research, the publication of several papers addressing the role of the intestinal mycobiome now suggested that these low numbers do not necessarily translate to irrelevance. In this review, we discuss the available clinical and preclinical IBS mycobiome data, and speculate how these recent findings may relate to earlier observations in IBS. By surveying literature from the broader mycobiome research field, we identified questions open to future IBS-oriented investigations.

This article is part of the following collections:
Gut Feelings: Microbiota and Pain

Introduction

Irritable bowel syndrome (IBS) is a common disorder of gut-brain interaction that affects an approximate 5–10% of the population. Diagnosis is based on symptoms and exclusion of structural alterations. Patients experience chronic abdominal pain in combination with altered bowel habits.Citation1 Online searches suggest that at least part of the IBS patients and clinical practitioners is convinced that intestinal yeasts are causally involved in this disorder (Google Search; “irritable bowel syndrome AND yeast”; 4,410,000 results, January 2023). Yet, scientific evidence for such an assumption is scarce. In PubMed, a search string including “irritable bowel syndrome AND (yeast OR candida OR myco*)” showed 152 results (January 2023). Although IBS-oriented gut microbiome research exploded during the last decade, investigations were mostly geared toward bacteria, leaving fungi (including yeasts) largely unexplored.Citation2,Citation3 The latter may be explained by the finding that fungi are only a minor component of the gut ecosystem. Shotgun sequencing results published by the METAgenomics of the Human Intestinal Tract (MetaHIT)-consortium indicated that fungi constitute no more than 0.1% of the human gut microbiota,Citation4 although this may have been an underestimate due to the use of non-yeast directed DNA isolation procedures and deficient fungal genome databases used for annotation. Another important consideration is that fungi are approximately 100 times larger in size than bacteria. Relative biomass is not reflected in read counts of metagenomics approaches but may be important when considering the fungal contribution to the total gut metabolome. Compiling 36 human gut mycobiome studies, Suhr and Hallen-AdamsCitation5 observed that ten out of the twelve most commonly detected fungi are yeasts, with Candida albicans and Saccharomyces cerevisiae as leading species. This may be relevant because yeast are capable of fermentation, and a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) was shown to manage IBS symptoms in the short and long term.Citation6 Importantly, whereas C. albicans is considered a gut commensal, the ubiquitous dietary presence of S. cerevisiae may explain at least part of its high frequency of detection.Citation7 In the field of inflammatory bowel disease (IBD), there is an increasing awareness that intestinal fungi/yeasts may be relevant players in IBD pathogenesis.Citation8 In IBS, mycobiome research is still in its infancy, despite patients’ perception and attitude toward the subject. Yet, some small IBS cohort studies indicated alterations of the fecal fungal community with, amongst others, enhanced relative abundance of C. albicans. In addition, using the IBS-like maternal separation model in rat, the first preclinical studies on the functional relevance of the mycobiome and modulation thereof have been published.Citation9–15 In this review, we aim to discuss the sparse IBS mycobiome literature and some of the recent developments in the broader IBS- and mycobiome research fields that may be relevant in the IBS/mycobiome context as well.

Anti-fungal and anti-food allergen antibodies

One of the early findings that sparked mycobiome research in IBD was the serological presence of the so-called anti-Saccharomyces cerevisiae antibodies (ASCAs), especially in patients with Crohn’s disease (~60%).Citation16 These antibodies recognize yeast mannans and we now know that Candida albicans is the more likely fungal immunological antigen trigger for ASCAs.Citation17 Serum titers in IBS were addressed in several studies, but ASCAs never showed increased presence over healthy volunteer sera.Citation18–20 Although this may indicate that there is no role for intestinal fungi, it could also relate to clear differences in (immune)-pathology between IBD and IBS. The latter is characterized by a general absence of overt inflammation,Citation21 whereas in Crohn’s disease, ASCA titers were shown to correlate with disease severity.Citation22,Citation23 Even so, antibodies with other specificity, i.e., dietary antigens, have been implicated in mast cell-dependent visceral pain. Mast cells and especially the mast cell mediator histamine are well-recognized drivers of abdominal pain complaints in at least a subset of IBS patients.Citation24–26 In a study conducted by the Boeckxstaens group, mucosal mast cells of all IBS patients proved to be sensitive to at least one of the tested food allergens upon local injection into rectosigmoid mucosa, despite negative skin prick tests.Citation24 The investigators suggested that bacterial gut infections and subsequent epithelial barrier disruption are the initial trigger for this localized immune response (). The latter was modeled in the ovalbumin (OVA) food allergy mouse model. While exposing mice to OVA, they were simultaneously infected with Citrobacter rodentium. When the infection was cleared, repeated OVA exposure led to an enhanced IgE-specific and mast cell-dependent visceromotor response during colorectal distension that was not observed in sham infected mice. The visceromotor response is often used in preclinical studies as readout for visceral sensitivity.Citation27,Citation28 Visceral hypersensitivity (i.e., increased perception of gastrointestinal stimuli) was shown to consistently associate with symptom severity in functional gastrointestinal disorders and is thought to be a mechanism explaining abdominal pain.Citation29 Based on the OVA/C. rodentium results, it was suggested that transient gut infection, like it is observed in post-infectious IBS (PI-IBS), can lead to IgE-dependent food allergy and abdominal pain.Citation24,Citation30 Others have shown that C. albicans is also capable of breaking oral tolerance in the OVA model and confirmed a role for IgE and mast cells, but possible effects on visceral sensitivity were not addressed in these C. albicans-focused studies.Citation31,Citation32

Figure 1. Possible mycobiome-related mechanisms of action in Irritable Bowel Syndrome. (a) Cellular and molecular mechanisms through which host and mycobiome may interact. Part of these interactions were described in IBS and IBS-like rodent models. Others were addressed in the somatic pain field and still need confirmation in IBS (see text). BLP, balloon-like protrusion; CGRP, calcitonin-related gene peptide; CRF, corticotropin-releasing factor; H1R, histamine-1-receptor; PAR2, protease activated receptor 2; PKC, protein kinase C; PLC, phospholipase C; TRPV1, transient receptor potential channel vanilloid 1. (b) Recent observations and considerations regarding the gut mycobiome in IBS. (c) Possible strategies for modulation of the fungal community in patients with IBS. Illustration created with BioRender.com.

Figure 1. Possible mycobiome-related mechanisms of action in Irritable Bowel Syndrome. (a) Cellular and molecular mechanisms through which host and mycobiome may interact. Part of these interactions were described in IBS and IBS-like rodent models. Others were addressed in the somatic pain field and still need confirmation in IBS (see text). BLP, balloon-like protrusion; CGRP, calcitonin-related gene peptide; CRF, corticotropin-releasing factor; H1R, histamine-1-receptor; PAR2, protease activated receptor 2; PKC, protein kinase C; PLC, phospholipase C; TRPV1, transient receptor potential channel vanilloid 1. (b) Recent observations and considerations regarding the gut mycobiome in IBS. (c) Possible strategies for modulation of the fungal community in patients with IBS. Illustration created with BioRender.com.

Mast cells and visceral hypersensitivity

Possible triggers for mast cell activation include, next to the aforementioned (food allergen-specific) IgE, a broad range of nonimmune signals such as substance P, nerve growth factor, calcitonin gene-related peptide (CGRP).Citation33 Because stress is an important trigger for IBS-associated complaints, the stress hormone corticotropin releasing factor (CRF) received a lot of attention. Stress-induced mast cell-dependent visceral hypersensitivity is thought to be an important mechanism explaining abdominal pain in IBS. Preclinical IBS-like animal models suggested that peripheral CRF can be responsible for mucosal mast cell degranulation, subsequent gut barrier dysfunction, and visceral hypersensitivity ().Citation33,Citation34 Yet, clinical trials with CRF-receptor antagonists were unsuccessful,Citation35,Citation36 possibly due to inadequate antagonist development.Citation37 In search for an alternative explanation, we addressed a possible flaw in the design of pioneering preclinical IBS studies that mostly administered CRF-receptor antagonists prior to stress.Citation34 When maternal separated rats were subjected to acute stress at adult age (i.e., 1 hour of water avoidance stress), they became hypersensitive to colorectal distension. This visceral hypersensitivity developed directly after water avoidance and was mediated by the activation of mucosal mast cells.Citation28,Citation38 We next showed that visceral hypersensitivity remained to be present for up to 30 days after just 1 hour of water avoidance stress, and that this long-term hypersensitivity also depended on the activation of mast cells.Citation39 Administering the CRF-receptor antagonist α-helical CRF prior to water avoidance stress prevented visceral hypersensitivity, but post-stress administration of the same antagonist could not reverse long-term hypersensitivity. In other words, acute and long-term visceral hypersensitivity both depended on mast cells, but only in the acute phase their activation was triggered by CRF. In a follow-up investigation, described in the next paragraph, we used the maternal separation model to address the possible role of intestinal fungi in post-stress mast cell activation and prolonged visceral hypersensitivity.Citation9

The mycobiome and visceral hypersensitivity

To assess whether the gut mycobiome is involved in IBS, we first compared the fecal mycobiota of healthy volunteers and patients with IBS with known visceral sensitivity status. Patient samples showed lower α-diversity and we observed differences in mycobiome signature when comparing normosensitive and hypersensitive patients.Citation9 Importantly, mycobiota dissimilarities between controls and IBS patients were later confirmed by others.Citation12–14 Subsequent analysis of fecal samples obtained from normal and maternal separated IBS-like rats showed profound mycobiome differences. In this mast cell and histamine-1-receptor dependent model, fungicide treatment reversed stress-induced visceral hypersensitivity.Citation9,Citation38,Citation40 Additional fecal transfer experiments confirmed an essential role for the maternal separation-related gut mycobiome. In relation to these results, it should be re-emphasized that despite the obvious role of intestinal fungi and mast cells, there is no overt inflammation in this IBS-like model.Citation38 The latter properly reflects the immune status in IBS.Citation21 Interestingly, although C. albicans is not considered a common colonizer of the rodent gut,Citation41 our rat mycobiome analysis indicated fecal presence of this species. This may be due to non-sterile handling procedures and the open cage system that was used for the Long-Evans in-house rat colony at the time. Although this may complicate duplication studies by other labs, we consider the use of “dirty rats” a positive attribute. Laboratory rodents are generally kept in highly sanitized environments, avoiding any contact with novel (pathogenic) microbial components. This does not reflect the everyday situation in humans and may diminish the translational value of these models.Citation8 Yeung et al. showed that controlled release of lab mice into the wild led to increased intestinal fungal load and, related to that, immune system maturation. The latter showed in enhanced differentiation of T cell populations and increased numbers of circulating granulocytes.Citation42 Although this “rewilding” setting is different from what our rats experienced, we assume that the translational value of our model was positively affected by the suboptimal level of cleanness in which experiments were conducted. Yet, we did not address whether the presence of C. albicans in maternal separated rats was essential for the hypersensitive phenotype. In IBS patients however, C. albicans was shown to be more abundant and the genus Candida positively correlated to severity of bloating and anxiety in diarrhea-predominant IBS (IBS-D).Citation12,Citation13 It has to be noted though that these sequencing results were obtained based on fecal samples. While these samples are relatively easy to obtain, they may not provide an accurate window to colonic mycobiota composition at mucosal level. Compared to healthy volunteer samples, the relative abundance of fecal Candida spp. is consistently increased in IBD cohorts, and recent data obtained by Li et al. showed that this is mirrored in ulcerative colitis mucosa.Citation43 Since there are no mucosa-associated mycobiota studies in IBS, it is not known whether the same holds true for this disorder.

Mast cells and the mycobiome in visceral hypersensitivity

Prior to our mycobiome-directed investigations, several groups already described direct fungal-induced mast cell activation involving the C-type lectin receptor Dectin-1 ().Citation44–46 This transmembrane receptor is predominantly expressed by myeloid cells and recognizes 1,3-linked β-glucans present in the fungal cell wall. Spleen tyrosine kinase (SYK) and caspase-associated recruitment domain 9 (CARD9) are both essential in mediating downstream cellular responses of this receptor.Citation47 Given the relevance of mast cells in sensory afferent activation in patients as well as in the maternal separation model,Citation24–26,Citation38–40 we also addressed this particular pathway of innate fungal recognition. Administration of inhibitory soluble β-glucans that antagonize Dectin-1, as well as a SYK inhibitor both reduced visceral hypersensitivity in maternal separated rats. Ex vivo experiments with mesenteric windows confirmed Dectin-1 dependent mast cell degranulation by particulate β-glucans.Citation9 In human, indications for a role of the Dectin-1/Syk pathway in mast cell activation were obtained by Chi et al.Citation48 When comparing IBS-D and healthy volunteer ileocecal biopsies, these investigators observed enhanced expression of CLEC7A (encoding Dectin-1), SYK, and CARD9 in patient biopsies. Which cell types were responsible for the enhanced expressions cannot be deduced from their findings. However, there was a positive correlation between plasma tryptase levels and CLEC7A expression, as well as plasma tryptase and increased visceral sensitivity of these patients. Next to mast cell derived histamine that triggers visceral hypersensitivity via the histamine-1-receptor in the maternal separation model and patients, tryptase was implicated in IBS pathophysiology by others as well.Citation26,Citation33,Citation40 This serine protease is released during mast cell degranulation. It can activate proteinase-activated receptor 2 (PAR2) on sensory afferents and enterocytes, leading to visceral hypersensitivity and barrier dysfunction ().Citation49–51 Concerning its mechanisms of action, cell culture experiments showed that activated PAR2 sensitizes, via protein kinase C, the capsaicin receptor transient receptor potential channel vanilloid 1 (TRPV1). Moreover, in an in vivo somatic pain setting, co-administration of non-hyperalgesic dosis of PAR2 agonist and capsaicin led to hyperalgesia that was absent in Trpv1−/− mice.Citation52 Since TRPV1 is also a key ion channel for visceral hypersensitivity in maternal separated rats and patients with IBS,Citation26,Citation38 these somatic findings on PAR2-mediated afferent sensitization may be relevant for IBS as well. Indeed, results obtained in an acetic enema rat model of visceral hypersensitivity suggested that the mast cell-PAR2-TRPV1 axis may play a role.Citation53 The possible relevance of tryptase/PAR2-induced intestinal permeability becomes clear knowing that barrier dysfunction observed in patients with IBS-D and PI-IBS subtypes positively associates with abdominal pain and changes in bowel function.Citation54 Moreover, when stress-induced barrier dysfunction was prevented in a rat model for IBS, this also prevented the development of visceral hypersensitivity.Citation55 Taken together, the above data suggest that mast cell recognition of intestinal fungi, via the pattern recognition receptor Dectin-1, may lead to histamine and tryptase release and consequent barrier dysfunction and visceral hypersensitivity.

Candida albicans and candidalysin dependent afferent activation

Dectin-1 dependent abdominal pain may also result from direct afferent activation. Albeit in a somatic setting, it was shown that C. albicans can interact with afferent expressed Dectin-1 to activate the neuronal phospholipase C/TRP-channel axis (i.e., TRPV1 and TRPA1; ) and cause β-glucan-dependent allodynia.Citation56 Importantly, in addition to showing a role for direct afferent activation in fungal-induced somatic pain, the same study also suggested that keratinocytes were the main cell type responsible for Dectin-1 dependent allodynia. In response to C. albicans, these skin epithelial cells released ATP that subsequently stimulated sensory afferents via P2X receptors. Whether similar mechanisms are relevant for visceral pain is not known, but functional Dectin-1 may be absent from enterocytes.Citation57 Besides addressing a role for Dectin-1, Maruyama and colleagues also investigated a possible role for candidalysin in direct afferent activation.Citation56 This pore-forming peptide was the first cytolytic toxin identified in C. albicans. Although C. albicans is a gut commensal, its ability to transform from yeast to filamentous morphology can turn it into an opportunistic pathogen. For in depth coverage and excellent graphical visualization of C. albicans interaction with, and translocation across the intestinal barrier and discussion of all virulence factors involved in this process, please refer to a recent open access publication by Sprague et al. in this journal.Citation58

Concerning candidalysin, it was shown that hypha-associated candidalysin is an essential virulence factor leading to damage of host cells and concurrent activation of anti-fungal immune responses.Citation59,Citation60 Others had shown that the bacterial pore-forming toxin α-hemolysin induces calcium flux and action potentials in nociceptor neurons.Citation61 Candidalysin however did not induce calcium fluxes in dorsal root ganglion neurons isolated from mice.Citation56 Mast cells on the other hand may be stimulated by candidalysin (). Using HMC-1 and LAD2 mast cell lines, candidalysin peptide and candidalysin-positive C. albicans strains were both shown to induce degranulation and cytokine release, which was not observed when candidalysin-null mutant C. albicans strains were used. Furthermore, intradermal (i.e., skin) injection of candidalysin in wild-type mice led to vascular permeability that was not observed in mast cell deficient KitW-sh/W-sh mice.Citation62 At present there are no data regarding in vivo candidalysin-mediated degranulation of colonic mast cells. But, some other candidalysin-focused investigations, described in the section on “C. albicans strain differences”, were performed in relation to IBS.Citation15

Candida albicans morphotype switching and IBS

As mentioned earlier, candidalysin becomes expressed upon yeast to hyphae transition which can be triggered by a diversity of signals such as temperature, pH, serum, N-acetylglucoseamine, peptidoglycan, amino acids etcetera. Because of the multiplicity and complexity of possible triggers and pathways involved in morphotype switching, Noble et al. suggested that C. albicans “continuously surveils the mammalian host, integrating a variety of signaling inputs to generate adaptive responses to the local environment”.Citation63 For IBS, the nature of these local environmental triggers is unknown. Even more, at this point there are no publications regarding the presence or absence of filamentous cells in IBS fecal and/or mucosal samples. However, in a mouse model of C. albicans colonization, yeast and hyphal morphotypes were simultaneously present, herewith challenging the dogma that these appearances strictly belong to commensal and tissue-invasive states respectively.Citation64,Citation65 In the absence of tissue invasion, both forms were observed in the intestinal lumen as well as adjacent to and in the mucus layer. Compared to stomach and small intestine, the filamentous form even predominated over yeast in cecum and colon. Further investigations showed that not yeast-to-hypha transition per se (i.e. cell morphology), but activation of virulence factors controls C. albicans intestinal commensalism. Based on their observations, Witchley et al. suggested that an anti-fungal host response to the hyphal phenotype will only be triggered when virulence factors exceed a threshold level.Citation65 This interesting “under the radar” hypothesis may also be relevant for IBS and the IBS-like maternal separation model, were fungi seem to play a role in abdominal pain without triggering a massive immune response.Citation9,Citation66–68

Candida albicans strain differences

Pilot investigations in a limited set of fecal healthy volunteer and IBS patient-derived C. albicans strains showed that yeast-to-hyphae transition could be induced in all strains, whereas simultaneous ECE1 (the gene encoding candidalysin) induction rates greatly differed.Citation15 The IBS C. albicans strains were isolated from hyper- and normosensitive patients. Genotyping by Amplified Fragment Length Polymorphisms (AFLP)-fingerprinting analysis of 63 strains showed partial clustering of strains derived from hypersensitive patients. Unfortunately, ECE1 expression analysis was performed in six strains only, thus preventing any conclusions regarding possible correlations with visceral hypersensitivity. On the other hand, clonal expansion of IBS-derived C. albicans strains distinct from healthy volunteers was also described by others, although without including the hyper- vs. normosensitive distinction.Citation14 Notably, the functional relevance of C. albicans strain differences for IBS pathophysiology and IBS clinical characteristics was not addressed so far. In IBD however, a range of C. albicans clinical isolates showed different damaging capacity toward macrophages, and induced different levels of antifungal interleukin (IL)-17A-producing T helper cell (Th17) responses. In vitro evaluation of C. albicans Ece1 knockout in high and low damaging strains suggested that candidalysin has a key role in these processes, and this was confirmed in vivo.Citation43 Candidalysin is also critical to induce epithelial damage needed for C. albicans translocation across intestinal epithelia.Citation69,Citation70 As mentioned before, our first fungal findings in the maternal separation model suggested that β-glucans are an important trigger for mast cell activation and subsequent histamine-induced visceral hypersensitivity.Citation9 In a review on host-microbe interactions, the question was raised how these fungal antigens make their way from the gut lumen.Citation71 Knowing that C. albicans was present during the referred animal experiments, we now suggest that future investigations should focus on a possible role for candidalysin in yeast or its antigen translocation in IBS.

Increased attention for strain differences between healthy volunteers and patients with IBS may also accelerate the search for biomarkers in this disorder. Due to the lack of convincing biological indicators, IBS is diagnosed by the symptom-based Rome IV criteria.Citation72 In search for alternative tools to discriminate patients with IBS from healthy individuals, or even identify patient subsets, numerous bacteria-focused microbiota analysis have been conducted on healthy volunteer and IBS fecal samples. Until now, inter study discrepancies prevented the identification of a universal “IBS microbiota” and hampered broad application of such techniques.Citation73 Recent findings within the Dutch Microbiome Project suggest that this type of biomarker-finding efforts may even be more difficult than appreciated thus far. Bacterial microbiome profiling of more than 8000 Dutch individuals showed shared dysbiosis between unrelated diseases (including IBS), which will most likely complicate the identification of disease specific microbiome signatures.Citation74 Similar inter-disease datasets are not yet available for the intestinal mycobiome, but its low abundance and high inter-individual variability already suggest that classical compositional analysis will most likely not result in satisfactory mycobiome based diagnostic and monitoring tools.Citation75 In general, a major caveat in bacterial as well as fungal compositional microbiota analysis for biomarker finding, is the complete disregard of strain differences within a species. Odds et al. studied a large set of non-IBS-related C. albicans strains that included isolates from longitudinal samples. Their results suggested clonal persistence in time.Citation76 Based on this, and the two recent publications on genetic clustering of C. albicans strains in IBS,Citation14,Citation15 we argue that strain genotyping may open up new avenues for diagnosis and identification of patient subsets.

Macrophages and the mycobiome in visceral hypersensitivity

Recent publications showed an indispensable role of intestinal CX3CR1+ (i.e., fractalkine receptor-expressing) mononuclear phagocytes in providing anti-fungal Th17 immune responses and maintaining intestinal epithelium integrity ().Citation77,Citation78 In mice, the latter protection is provided by Cx3cr1+ macrophages with high CD11c expression levels. These macrophages insert balloon-like protrusions (BLP) into colonic epithelial cells to sense fungal toxins and metabolites absorbed by these enterocytes. In the presence of fungal toxins, BLP+ macrophages instructed epithelial cells to stop fluid absorption, preventing epithelial cell death. Macrophage depletion in distal colon was associated with massive enterocyte apoptosis that was prevented by anti-fungal treatment.Citation77 Leonardi et al. showed that ablation of intestinal CD11c+/Cx3cr1+ cells led to mycobiota changes and enhanced susceptibility to dextran sodium sulfate (DSS) colitis that was rescued by fungicide treatment.Citation78 In relation to IBS not much is known about a possible role for CD11c+/CX3CR1+ cells. Microarray analysis of IBS and healthy volunteer sigmoid biopsies indicated elevated expression of CX3CR1 in IBS. But, interpretation of these results is difficult because they were not cell specific.Citation79 In addition, if and how overexpression associated with mycobiota dysbiosis is not known. Results obtained in a Trichinella spiralis PI-IBS model seem to contradict the above findings on epithelial integrity.Citation80 Transfer of post-infectious CD11c+ lamina propria mononuclear cells caused mucosal barrier dysfunction and visceral hypersensitivity in naïve recipient mice. However, donor cells were isolated from small intestine. Although, transepithelial dendrites that sample the lumen were indeed reported in small intestine, BLPs observed in colonic CD11c+/Cx3cr1+ cells do not reach the lumen, suggesting that these two cell types serve different functions.Citation77,Citation81

Importantly, Candida spp. colonization in mice induced an increase in Th17 cell frequency which dramatically decreased upon depletion of colonic Cx3cr1+ cells, confirming their role in anti-fungal immune defense.Citation78 Only few reports exist on Th17 immune response in IBS. Berg et al. assessed cytokine levels in homogenized rectal biopsies of IBS patients and controls. They observed, amongst increased presence of several other pro-inflammatory cytokines, that IL-17 was significantly increased in IBS samples.Citation82 In situ immunohistochemical stainings on descending and rectosigmoid biopsies confirmed enhanced presence of Th17 cells in IBS patients as compared to controls.Citation83 Interestingly, using TRPV1-Ai32 optogenetic mice and cutaneous light stimulation it could be shown that activation of TRPV1+ neurons is sufficient to elicit, via CGRP-release, a Th17 response. Not only was this response sufficient to increase host defense to C. albicans, it also provided regional Type-17 inflammation via a nerve-reflex arc.Citation84 These observations suggested, that direct sensory afferent activation by fungi not only signals danger by eliciting a pain response,Citation56 but also initiates early protective immunity for adjacent areas. Whether these mechanisms also apply to the intestinal environment and visceral afferents could be a topic for future IBS-related investigations.

Whether colonic CD11c+/CX3CR1+ cells directly interact with mast cells to induce degranulation is not known, but macrophages may contribute in another unexpected fashion. Macrophages themselves could be a source of histamine. Histidine decarboxylase (HDC) is the only enzyme known to catalyze the conversion of histidine to histamine. Some investigators described HDC expression and histamine release in blood monocytes and bone marrow-derived macrophages.Citation85–88 Unfortunately, at this point, there are no data on the possible in situ presence of HDC-expressing macrophages in IBS tissues. In vitro, enhanced histamine release due to luminal factors was successfully shown with LPS, but possible yeast derived triggers were not evaluated for these macrophages.Citation85,Citation86

Inter-kingdom interactions in IBS

Although inter-kingdom interactions are a two-way street, most publications seem to focus on bacteria influencing fungal traits such as biofilm formation, yeast-to-hyphae transition and mucosal invasion. On the other hand, the gut mycobiota also plays an important role in maintaining homeostasis of the bacterial microbiome and gut health. For non-IBS focused but recent overviews on the aforementioned cross-kingdom interactions in health and disease we refer to two excellent reviews.Citation89,Citation90 Hong et al. addressed positive and negative fungal-bacterial correlations in IBS.Citation13 Compared to healthy volunteers, fecal samples of IBS-D showed fewer significant correlations (43 vs. 25, resp.). In both groups, the highest number of interactions concerned the genus Candida. However, in healthy volunteers 9 out of 12 observed Candida-bacteria correlations were negative, whereas in IBS-D all 8 correlations were positive. Moreover, not one of these Candida-specific correlations overlapped between the healthy volunteer and IBS-D groups. The overall decline in the number of interactions in IBS-D and changes in specificity of Candida-interactions may reflect the microbiome’s inability to maintain homeostasis. The functional relevance of these interactions should be addressed in future investigations, perhaps by using intestinal organoid cultures and microbiota grafting in IBS-like animal models. Possibly, the opposite Candida correlations reported by Hong et al. can be explained by the earlier reports on C. albicans genetic and phenotypic diversity.Citation14,Citation15 Thus, we feel that C. albicans strain differences should be taken into account when addressing the nature and relevance of altered inter-kingdom interactions.

Mycobiome-targeted therapy

Direct targeting of the gut fungal community in order to relieve abdominal complaints has, to our knowledge, not yet been tested for patients with IBS. In the IBS-like rodent model, we have shown that classical antifungal treatment does indeed reduce visceral hypersensitivity.Citation9 But, considering the increasing fungal resistance to common antifungal medications, other methods should be focused on as well. Administration of the antiparasitic drug miltefosine reduced and prevented visceral hypersensitivity in the same animal model through inference with the gut bacterial and fungal microbial composition, and by reducing mast cell activation.Citation10 Similar observations were made for the application of herbal oil mixtures. A preparation of caraway and peppermint essential oils (Menthacarin®) modulated the mycobiome and reduced visceral hypersensitivity in maternal separated rats.Citation11 Whether there was a causal relation between modulation of the mycobiota and reduction of complaints remains to be established. In human, the mixtures effect on the gut mycobiome was not yet studied and the same holds true for the herbal mixture STW-5 (Iberogast®) that is indicated for the treatment of IBS and also contains caraway and peppermint.Citation91 Furthermore, a meta-analysis on peppermint oil studies in IBS suggested that peppermint oil was more efficacious than placebo for global IBS symptoms and abdominal pain, but mycobiome alterations were never addressed.Citation92 Large randomized controlled trials with accompanying mycobiota analysis could provide evidence as to whether herbal-mediated mycobiota modulation is feasible and relevant for patients with IBS.

Dietary restriction of FODMAPs is now widely used in the management of IBS.Citation6 Two important mechanisms seem to be involved in the successful application of the low FODMAP diet. In small intestine, FODMAPs are thought to induce enhanced water uptake due to osmotic effects. In colon, microbial fermentation of FODMAPs leads to gas production. Both mechanisms give rise to luminal distension and symptoms in those with visceral hypersensitivity.Citation93,Citation94 Despite fungi making up an approximate 1–2% of the luminal biomass,Citation95 and although yeast are the prototypical fermentation workhorses in human history, intestinal FODMAP fermentation is always discussed in the context of bacteria only. Especially when knowing that high Candida spp. abundance strongly associated with recent consumption of carbohydrates,Citation96 investigations addressing the relative involvement of yeast in the success of the low FODMAP diet are eagerly awaited. Alternatively, and based on the aforementioned inter-kingdom interactions, bacterial probiotics may be used to interfere with intestinal yeasts.Citation90 In a recent systemic meta-analysis, McFarland et al. aimed to determine, when taking into account the recommendations from the European Society for Pediatric Gastroenterology, Hepathology and Nutrition (ESPGHAN) and the American Gastroenterology Association (AGA), which probiotics are safe and effective for the treatment of IBS. Only randomized, controlled trials (RCTs) with at least one strain-specific confirmatory RCT and common IBS outcome measures such as abdominal pain scores were included.Citation97 Only 3 single strain bacterial probiotics showed significant changes in abdominal pain scores and/or frequency of abdominal pain relief: Lactobacilli plantarum 299 v, Lactobacillus rhamnosus GG (LGG) and Bacillus coagulans MTCC5260. Although their working mechanisms in IBS are largely unknown, for two of these strains (L. plantarum 299 v and LGG) in vitro experiments have shown effects on Candida growth, morphogenesis, and adhesion, and these outcomes may also be relevant to IBS.Citation98–101 We suggest, that future selection procedures of probiotics for IBS clinical trials should include in vitro yeast-related read outs. Importantly, it was shown that the efficacy of probiotics is both strain- and disease-specific.Citation102 Thus, multiple strains of the same species should be included in in vitro experiments and selected strains should be tested in preclinical models prior to clinical evaluation. In addition to probiotic bacteria, some yeasts are suggested to have probiotic effects as well. At present, only a very limited set of yeast species have a qualified presumption of safety (QPS) by the European Food Safety Agency (EFSA) for viable usage. These include Candida kefyr (also known as Kluyveromyces marxianus) and Saccharomyces cerevisiae (including S. boulardii CNCM I-7454 and S. cerevisiae CNCM I-3856).Citation103,Citation104 In the McFarland meta-analysis on probiotic RCTs in IBS, the latter two S. cerevisiae strains showed significant efficacy for abdominal pain management in ≥ 2 RCTs.Citation97 Although the responsible mechanisms remain elusive, both strains are known to affect C. albicans as well as albicans-related host immune responses.Citation104,Citation105 We suggest that the efficacy of yeast probiotics in IBS may be further enhanced when probiotic strain selection is performed with C. albicans or other IBS-suspect fungal species in mind. Relevant aspects such as C. albicans adhesion to intestinal epithelial cells, yeast-to-hypha transition and host immune responses can be targeted during such probiotic evaluations.

Perspective

Although still limited in number, recent publications indicated intestinal mycobiota alterations in IBS. Similar findings in IBD initiated a promising new field of mycobiome-oriented research, and the same may happen in IBS. Investigations performed in an IBS-like rat model already suggested that the observed mycobiota changes may be relevant for abdominal pain and addressed possible mechanisms and therapeutic approaches. In IBS there is a relative increase of intestinal C. albicans and genotyping of fecal C. albicans strains indicated clustering of IBS-derived strains as compared to healthy volunteers. Traditionally, there is a lot of attention for C. albicans in the broader mycobiome field of investigations. Recent insights on C. albicans/macrophage and C. albicans/epithelial cell interactions as well as C. albicans-mediated afferent-activation should be addressed in relation to IBS. Taken together, we suggest that fungal feelings in IBS should no longer be ignored, but investigated instead.

Disclosure statement

Authors declare no relevant financial or non-financial competing interests.

Additional information

Funding

This work was supported by the European Crohn’s and Colitis Organisation [ECCO grant 2019]; Health~Holland [PPP Allowance number LSHM20085];

References

  • Ford AC, Sperber AD, Corsetti M, Camilleri M. Irritable bowel syndrome. Lancet. 2020;396(10263):1675–14. doi:10.1016/S0140-6736(20)31548-8.
  • Algera JP, Törnblom H, Simrén M. Treatments targeting the luminal gut microbiota in patients with irritable bowel syndrome. Current Opinion in Pharmacology. 2022;66:102284. doi:10.1016/j.coph.2022.102284.
  • Pittayanon R, Lau JT, Yuan Y, Leontiadis GI, Tse F, Surette M, Moayyedi P. Gut microbiota in patients with irritable bowel syndrome-a systematic review. Gastroenterology. 2019;157(1):97–108. doi:10.1053/j.gastro.2019.03.049.
  • Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. doi:10.1038/nature08821.
  • Suhr MJ, Hallen-Adams HE. The human gut mycobiome: pitfalls and potentials–a mycologist’s perspective. Mycologia. 2015;107(6):1057–1073. doi:10.3852/15-147.
  • Whelan K, Staudacher H. Low FODMAP diet in irritable bowel syndrome: a review of recent clinical trials and meta-analyses. Curr Opin Clin Nutr Metab Care. 2022;25(5):341–347. doi:10.1097/MCO.0000000000000854.
  • Hallen-Adams HE, Suhr MJ. Fungi in the healthy human gastrointestinal tract. Virulence. 2017;8(3):352–358.
  • Underhill DM, Braun J. Fungal microbiome in inflammatory bowel disease: a critical assessment. J Clin Invest. 2022;132(5):e155786.
  • Botschuijver S, Roeselers G, Levin E, Jonkers DM, Welting O, Heinsbroek SEM, de Weerd HH, Boekhout T, Fornai M, Masclee AA, et al. Intestinal fungal dysbiosis is associated with visceral hypersensitivity in patients with irritable bowel syndrome and rats. Gastroenterology. 2017;153(4):1026–1039. doi:10.1053/j.gastro.2017.06.004.
  • Botschuijver S, van Diest SA, van Thiel IAM, Saia RS, Strik AS, Yu Z, Maria-Ferreira D, Welting O, Keszthelyi D, Jennings G, et al. Miltefosine treatment reduces visceral hypersensitivity in a rat model for irritable bowel syndrome via multiple mechanisms. Sci Rep. 2019;9(1):12530. doi:10.1038/s41598-019-49096-y.
  • Botschuijver S, Welting O, Levin E, Maria-Ferreira D, Koch E, Montijn RC, Seppen J, Hakvoort TBM, Schuren FHJ, de Jonge WJ, et al. Reversal of visceral hypersensitivity in rat by Menthacarin®, a proprietary combination of essential oils from peppermint and caraway, coincides with mycobiome modulation. Neurogastroenterology and Motility: the Official Journal of the European Gastrointestinal Motility Society. 2018;30(6):e13299. doi:10.1111/nmo.13299.
  • Das A, O’Herlihy E, Shanahan F, O’Toole PW, Jeffery IB. The fecal mycobiome in patients with irritable bowel syndrome. Sci Rep. 2021;11(1):124. doi:10.1038/s41598-020-79478-6.
  • Hong G, Li Y, Yang M, Li G, Qian W, Xiong H, Bai T, Song J, Zhang L, Hou X. Gut fungal dysbiosis and altered bacterial-fungal interaction in patients with diarrhea-predominant irritable bowel syndrome: an explorative study. Neurogastroenterology and Motility: the Official Journal of the European Gastrointestinal Motility Society. 2020;32(11):e13891. doi:10.1111/nmo.13891.
  • Sciavilla P, Strati F, Di Paola M, Modesto M, Vitali F, Cavalieri D, Prati GM, Di Vito M, Aragona G, De Filippo C, et al. Gut microbiota profiles and characterization of cultivable fungal isolates in IBS patients. Appl Microbiol Biotechnol. 2021;105(8):3277–3288. doi:10.1007/s00253-021-11264-4.
  • van Thiel IAM, Stavrou AA, de Jong A, Theelen B, Davids M, Hakvoort TBM, Admiraal-van den Berg I, Weert ICM, de Kruijs M, Vu D, et al. Genetic and phenotypic diversity of fecal Candida albicans strains in irritable bowel syndrome. Sci Rep. 2022;12(1):5391. doi:10.1038/s41598-022-09436-x.
  • Quinton J-F, Sendid B, Reumaux D, Duthilleul P, Cortot A, Grandbastien B, Charrier G, Targan SR, Colombel J-F, Poulain D. Anti- Saccharomyces cerevisiae mannan antibodies combined with antineutrophil cytoplasmic autoantibodies in inflammatory bowel disease: prevalence and diagnostic role. Gut. 1998;42(6):788–791. doi:10.1136/gut.42.6.788.
  • Poulain D, Sendid B, Standaert-Vitse A, Fradin C, Jouault T, Jawhara S, Colombel JF. Yeasts: neglected pathogens. Digestive Diseases (Basel, Switzerland). 2009;27(Suppl 1):104–110. doi:10.1159/000268129.
  • Kaila B, Orr K, Bernstein CN. The anti-Saccharomyces cerevisiae antibody assay in a province-wide practice: accurate in identifying cases of Crohn’s disease and predicting inflammatory disease. Can J Gastroenterol. 2005;19(12):717–721. doi:10.1155/2005/147681.
  • Müller S, Styner M. Anti- Saccharomyces cerevisiae antibody titers are stable over time in Crohn’s patients and are not inducible in murine models of colitis. World Journal of Gastroenterology: WJG. 2005;11(44):6988–6994. doi:10.3748/wjg.v11.i44.6988.
  • Schoepfer AM, Schaffer T, Seibold-Schmid B, Muller S, Seibold F. Antibodies to flagellin indicate reactivity to bacterial antigens in IBS patients. NeurogastroenterolMotil. 2008;20(10):1110–1118. doi:10.1111/j.1365-2982.2008.01166.x.
  • Bashashati M, Moossavi S, Cremon C, Barbaro MR, Moraveji S, Talmon G, Rezaei N, Hughes PA, Bian ZX, Choi CH, et al. Colonic immune cells in irritable bowel syndrome: a systematic review and meta-analysis. Neurogastroenterol Motil: Off J Eu Gastrointestinal Motil Soc. 2018;30(1):e13192.
  • Arnott ID, Landers CJ, Nimmo EJ, Drummond HE, Smith BK, Targan SR, Satsangi J. Sero-reactivity to microbial components in Crohn’s disease is associated with disease severity and progression, but not NOD2/CARD15 genotype. The American Journal of Gastroenterology. 2004;99(12):2376–2384. doi:10.1111/j.1572-0241.2004.40417.x.
  • Walker LJ, Aldhous MC, Drummond HE, Smith BR, Nimmo ER, Arnott ID, Satsangi J. Anti- Saccharomyces cerevisiae antibodies (ASCA) in Crohn’s disease are associated with disease severity but not NOD2/CARD15 mutations. Clinical and Experimental Immunology. 2004;135(3):490–496. doi:10.1111/j.1365-2249.2003.02392.x.
  • Aguilera-Lizarraga J, Florens MV, Viola MF, Jain P, Decraecker L, Appeltans I, Cuende-Estevez M, Fabre N, Van Beek K, Perna E, et al. Local immune response to food antigens drives meal-induced abdominal pain. Nature. 2021;590(7844):151–156. doi:10.1038/s41586-020-03118-2.
  • Klooker TK, Braak B, Koopman KE, Welting O, Wouters MM, van der Heide S, Schemann M, Bischoff SC, van den Wijngaard RM, Boeckxstaens GE. The mast cell stabiliser ketotifen decreases visceral hypersensitivity and improves intestinal symptoms in patients with irritable bowel syndrome. Gut. 2010;59(9):1213–1221. doi:10.1136/gut.2010.213108.
  • Wouters MM, Balemans D, Van Wanrooy S, Dooley J, Cibert-Goton V, Alpizar YA, Valdez-Morales EE, Nasser Y, Van Veldhoven PP, Vanbrabant W, et al. Histamine receptor H1-mediated sensitization of TRPV1 mediates visceral hypersensitivity and symptoms in patients with irritable bowel syndrome. Gastroenterology. 2016;150(4):875–87 e9. doi:10.1053/j.gastro.2015.12.034.
  • Ness TJ, Gebhart GF. Colorectal distension as a noxious visceral stimulus: physiologic and pharmacologic characterization of pseudaffective reflexes in the rat. Brain Res. 1988;450:153–169.
  • Welting O, Van Den Wijngaard RM, De Jonge WJ, Holman R, Boeckxstaens GE. Assessment of visceral sensitivity using radio telemetry in a rat model of maternal separation. NeurogastroenterolMotil. 2005;17(6):838–845. doi:10.1111/j.1365-2982.2005.00677.x.
  • Simren M, Tornblom H, Palsson OS, van Tilburg MA, Van Oudenhove L, Tack J, Whitehead WE. Visceral hypersensitivity is associated with GI symptom severity in functional GI disorders: consistent findings from five different patient cohorts. Gut. 2018;67(2):255–262.
  • Barbara G, Grover M, Bercik P, Corsetti M, Ghoshal UC, Ohman L, Rajilic-Stojanovic M. Rome foundation working team report on post-infection irritable bowel syndrome. Gastroenterology. 2019;156(1):46–58.e7. doi:10.1053/j.gastro.2018.07.011.
  • Sugita R, Hata E, Miki A, Andoh R, Umeda C, Takemura N, Sonoyama K. Gut colonization by Candida albicans Inhibits the induction of humoral immune tolerance to dietary antigen in BALB/c mice. Biosci Microbiota Food Health. 2012;31:77–84.
  • Yamaguchi N. Gastrointestinal Candida colonisation promotes sensitisation against food antigens by affecting the mucosal barrier in mice. Gut. 2006;55(7):954–960. doi:10.1136/gut.2005.084954.
  • Wouters MM, Vicario M, Santos J. The role of mast cells in functional GI disorders. Gut. 2016;65(1):155–168. doi:10.1136/gutjnl-2015-309151.
  • Van Den Wijngaard RM, Klooker TK, De Jonge WJ, Boeckxstaens GE. Peripheral relays in stress-induced activation of visceral afferents in the gut. AutonNeurosci. 2010;153:99–105.
  • Dukes GE, Mayer EA, Kelleher DL, Hicks KJ, Boardley RL, Alpers DH. A randomised double blind, placebo controlled, crossover study to evaluate the efficacy and safety of the corticotrophin releasing factor 1 (CRF1) receptor antagonist GW876008 in IBS patients. NeurogastroenterolMotil. 2009;21(Suppl):84.
  • Sweetser S, Camilleri M, Linker Nord SJ, Burton DD, Castenada L, Croop R, Tong G, Dockens R, Zinsmeister AR. Do corticotropin releasing factor-1 receptors influence colonic transit and bowel function in women with irritable bowel syndrome? AmJPhysiol GastrointestLiver Physiol. 2009;296(6):G1299–G306. doi:10.1152/ajpgi.00011.2009.
  • Lv Y, Wen J, Fang Y, Zhang H, Zhang J. Corticotropin-releasing factor receptor 1 (CRF-R1) antagonists: promising agents to prevent visceral hypersensitivity in irritable bowel syndrome. Peptides. 2022;147:170705. doi:10.1016/j.peptides.2021.170705.
  • van den Wijngaard RM, Klooker TK, Welting O, Stanisor OI, Wouters MM, van der Coelen D, Bulmer DC, Peeters PJ, Aerssens J, de Hoogt R, et al. Essential role for TRPV1 in stress-induced (mast cell-dependent) colonic hypersensitivity in maternally separated rats. Neurogastroenterol Motil: Off J Eu Gastrointestinal Motil Soc. 2009;21:1107–e94.
  • Van Den Wijngaard RM, Stanisor OI, van Diest SA, Welting O, Wouters MM, De Jonge WJ, Boeckxstaens GE. Peripheral alpha-helical CRF (9-41) does not reverse stress-induced mast cell dependent visceral hypersensitivity in maternally separated rats. NeurogastroenterolMotil. 2012;24:e111.
  • Stanisor OI, van Diest SA, Yu Z, Welting O, Bekkali N, Shi J, de Jonge WJ, Boeckxstaens GE, van den Wijngaard RM, Chowen JA. Stress-induced visceral hypersensitivity in maternally separated rats can be reversed by peripherally restricted histamine-1-receptor antagonists. PloS one. 2013;8(6):e66884. doi:10.1371/journal.pone.0066884.
  • Koh AY. Murine models of Candida gastrointestinal colonization and dissemination. Eukaryotic Cell. 2013;12(11):1416–1422. doi:10.1128/EC.00196-13.
  • Yeung F, Chen YH, Lin JD, Leung JM, McCauley C, Devlin JC, Hansen C, Cronkite A, Stephens Z, Drake-Dunn C, et al. Altered immunity of laboratory mice in the natural environment is associated with fungal colonization. Cell Host & Microbe. 2020;27(5):809–822.e6. doi:10.1016/j.chom.2020.02.015.
  • Li XV, Leonardi I, Putzel GG, Semon A, Fiers WD, Kusakabe T, Lin WY, Gao IH, Doron I, Gutierrez-Guerrero A, et al. Immune regulation by fungal strain diversity in inflammatory bowel disease. Nature. 2022;603(7902):672–678. doi:10.1038/s41586-022-04502-w.
  • Kimura Y, Chihara K, Honjoh C, Takeuchi K, Yamauchi S, Yoshiki H, Fujieda S, Sada K. Dectin-1-mediated signaling leads to characteristic gene expressions and cytokine secretion via spleen tyrosine kinase (Syk) in rat mast cells. The Journal of Biological Chemistry. 2014;289(45):31565–31575. doi:10.1074/jbc.M114.581322.
  • Nieto-Patlan A, Campillo-Navarro M, Rodriguez-Cortes O, Munoz-Cruz S, Wong-Baeza I, Estrada-Parra S, Estrada-Garcia I, Serafin-Lopez J, Chacon-Salinas R. Recognition of Candida albicans by Dectin-1 induces mast cell activation. Immunobiology. 2015;220(9):1093–1100. doi:10.1016/j.imbio.2015.05.005.
  • Pinke KH, Lima HG, Cunha FQ, Lara VS. Mast cells phagocyte Candida albicans and produce nitric oxide by mechanisms involving TLR2 and Dectin-1. Immunobiology. 2015;221(2):220–227. doi:10.1016/j.imbio.2015.09.004.
  • Reid DM, Gow NA, Brown GD. Pattern recognition: recent insights from Dectin-1. CurrOpinImmunol. 2009;21:30–37.
  • Chi Y, Li C, Wu LH, Wang HH. The relationship between dectin-1 and mast cells in patients with diarrhea-predominant irritable bowel syndrome. Scandinavian J Gastroenterol. 2020;55(7):762–768.
  • Bueno L. Protease activated receptor 2: a new target for IBS treatment. European Review for Medical and Pharmacological Sciences. 2008;12:95–102.
  • Jacob C, Yang PC, Darmoul D, Amadesi S, Saito T, Cottrell GS, Coelho AM, Singh P, Grady EF, Perdue M, et al. Mast cell tryptase controls paracellular permeability of the intestine. Role of protease-activated receptor 2 and beta-arrestins. JBiolChem. 2005;280:31936–31948.
  • Vergnolle N, Bunnett NW, Sharkey KA, Brussee V, Compton SJ, Grady EF, Cirino G, Gerard N, Basbaum AI, Andrade-Gordon P, et al. Proteinase-activated receptor-2 and hyperalgesia: a novel pain pathway. Nature Medicine. 2001;7(7):821–826. doi:10.1038/89945.
  • Amadesi S, Nie J, Vergnolle N, Cottrell GS, Grady EF, Trevisani M, Manni C, Geppetti P, McRoberts JA, Ennes H, et al. Protease-activated receptor 2 sensitizes the capsaicin receptor transient receptor potential vanilloid receptor 1 to induce hyperalgesia. JNeurosci. 2004;24(18):4300–4312. doi:10.1523/JNEUROSCI.5679-03.2004.
  • Li YJ, Dai C, Jiang M. Mechanisms of probiotic VSL#3 in a rat model of visceral hypersensitivity involves the mast cell-PAR2-TRPV1 pathway. Digestive Diseases and Sciences. 2019;64(5):1182–1192. doi:10.1007/s10620-018-5416-6.
  • Hanning N, Edwinson AL, Ceuleers H, Peters SA, De Man JG, Hassett LC, De Winter BY, Grover M. Intestinal barrier dysfunction in irritable bowel syndrome: a systematic review. Therapeutic Advances in Gastroenterology. 2021;14:1756284821993586. doi:10.1177/1756284821993586.
  • Ait-Belgnaoui A, Bradesi S, Fioramonti J, Theodorou V, Bueno L. Acute stress-induced hypersensitivity to colonic distension depends upon increase in paracellular permeability: role of myosin light chain kinase. Pain. 2005;113(1):141–147. doi:10.1016/j.pain.2004.10.002.
  • Maruyama K, Takayama Y, Sugisawa E, Yamanoi Y, Yokawa T, Kondo T, Ishibashi KI, Sahoo BR, Takemura N, Mori Y, et al. The ATP transporter VNUT mediates induction of Dectin-1-triggered candida nociception. iScience. 2018;6:306–318. doi:10.1016/j.isci.2018.08.007.
  • Volman JJ, Mensink RP, Buurman WA, Onning G, Plat J. The absence of functional dectin-1 on enterocytes may serve to prevent intestinal damage. Eur J Gastroenterol Hepatol. 2010;22(1):88–94. doi:10.1097/MEG.0b013e32832a20dc.
  • Sprague JL, Kasper L, Hube B. From intestinal colonization to systemic infections: candida albicans translocation and dissemination. Gut Microbes. 2022;14(1):2154548. doi:10.1080/19490976.2022.2154548.
  • Moyes DL, Wilson D, Richardson JP, Mogavero S, Tang SX, Wernecke J, Hofs S, Gratacap RL, Robbins J, Runglall M, et al. Candidalysin is a fungal peptide toxin critical for mucosal infection. Nature. 2016;532(7597):64–68. doi:10.1038/nature17625.
  • Naglik JR, Gaffen SL, Hube B. Candidalysin: discovery and function in Candida albicans infections. Curr Opin Microbiol. 2019;52:100–109. doi:10.1016/j.mib.2019.06.002.
  • Chiu IM, Heesters BA, Ghasemlou N, Von Hehn CA, Zhao F, Tran J, Wainger B, Strominger A, Muralidharan S, Horswill AR, et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature. 2013;501(7465):52–57. doi:10.1038/nature12479.
  • Song P, Peng G, Yue H, Ogawa T, Ikeda S, Okumura K, Ogawa H, Niyonsaba F. Candidalysin, a virulence factor of Candida albicans, stimulates mast cells by mediating cross-talk between signaling pathways activated by the dectin-1 receptor and MAPKs. J Clin Immunol. 2022;42(5):1009–1025. doi:10.1007/s10875-022-01267-9.
  • Noble SM, Gianetti BA, Witchley JN. Candida albicans cell-type switching and functional plasticity in the mammalian host. Nature Reviews Microbiology. 2017;15(2):96–108. doi:10.1038/nrmicro.2016.157.
  • Desai JV, Lionakis MS. Setting up home: fungal rules of commensalism in the mammalian gut. Cell Host & Microbe. 2019;25(3):347–349. doi:10.1016/j.chom.2019.02.012.
  • Witchley JN, Penumetcha P, Abon NV, Woolford CA, Mitchell AP, Noble SM. Candida albicans Morphogenesis Programs Control the Balance between Gut Commensalism and Invasive Infection. Cell Host & Microbe. 2019;25(3):432–43.e6. doi:10.1016/j.chom.2019.02.008.
  • Braak B, Klooker TK, Wouters MM, Welting O, van der Loos CM, Stanisor OI, van Diest S, van den Wijngaard RM, Boeckxstaens GE. Mucosal immune cell numbers and visceral sensitivity in patients with irritable bowel syndrome: is there any relationship? The American Journal of Gastroenterology. 2012;107(5):715–726. doi:10.1038/ajg.2012.54.
  • Gu Y, Zhou G, Qin X, Huang S, Wang B, Cao H. The potential role of gut mycobiome in irritable bowel syndrome. Frontiers in Microbiology. 2019;10:1894. doi:10.3389/fmicb.2019.01894.
  • Liu A, Gao W, Zhu Y, Hou X Chu H. Gut Non-Bacterial Microbiota: Emerging Link to Irritable Bowel Syndrome. Toxins (Basel). 2022;14(9):596.
  • Allert S, Forster TM, Svensson CM, Richardson JP, Pawlik T, Hebecker B, Rudolphi S, Juraschitz M, Schaller M, Blagojevic M, et al. Candida albicans-induced epithelial damage mediates translocation through intestinal barriers. mBio. 2018;9(3):e00915–18.
  • Mogavero S, Sauer FM, Brunke S, Allert S, Schulz D, Wisgott S, Jablonowski N, Elshafee O, Krüger T, Kniemeyer O, et al. Candidalysin delivery to the invasion pocket is critical for host epithelial damage induced by Candida albicans. Cellular Microbiology. 2021;23(10):e13378. doi:10.1111/cmi.13378.
  • Paterson MJ, Oh S, Underhill DM. Host-microbe interactions: commensal fungi in the gut. Current Opinion in Microbiology. 2017;40:131–137. doi:10.1016/j.mib.2017.11.012.
  • Lacy BE, Patel NK, Iwata N, Katsuyama T, Komatsubara M, Nagao R, Inagaki K, Otsuka F. Rome criteria and a diagnostic approach to irritable bowel syndrome. Journal of Clinical Medicine. 2017;6(1):6. doi:10.3390/jcm6010006.
  • Hillestad EMR, van der Meeren A, Nagaraja BH, Bjørsvik BR, Haleem N, Benitez-Paez A, Sanz Y, Hausken T, Lied GA, Lundervold A, et al. Gut bless you: the microbiota-gut-brain axis in irritable bowel syndrome. World Journal of Gastroenterology: WJG. 2022;28(4):412–431. doi:10.3748/wjg.v28.i4.412.
  • Gacesa R, Kurilshikov A, Vich Vila A, Sinha T, Klaassen MAY, Bolte LA, Andreu-Sánchez S, Chen L, Collij V, Hu S, et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature. 2022;604(7907):732–739. doi:10.1038/s41586-022-04567-7.
  • Enaud R, Vandenborght LE, Coron N, Bazin T, Prevel R, Schaeverbeke T, Berger P, Fayon M, Lamireau T. The mycobiome: a neglected component in the microbiota-gut-brain axis. Microorganisms. 2018;6(1):6. doi:10.3390/microorganisms6010006.
  • Odds FC, Davidson AD, Jacobsen MD, Tavanti A, Whyte JA, Kibbler CC, Ellis DH, Maiden MC, Shaw DJ, Gow NA. Candida albicans strain maintenance, replacement, and microvariation demonstrated by multilocus sequence typing. Journal of Clinical Microbiology. 2006;44(10):3647–3658. doi:10.1128/JCM.00934-06.
  • Chikina AS, Nadalin F, Maurin M, San-Roman M, Thomas-Bonafos T, Li XV, Lameiras S, Baulande S, Henri S, Malissen B, et al. Macrophages maintain epithelium integrity by limiting fungal product absorption. Cell. 2020;183(2):411–428.e16. doi:10.1016/j.cell.2020.08.048.
  • Leonardi I, Li X, Semon A, Li D, Doron I, Putzel G, Bar A, Prieto D, Rescigno M, McGovern DPB, et al. CX3CR1(+) mononuclear phagocytes control immunity to intestinal fungi. Science. 2018;359(2):232–236. doi:10.1126/science.aao1503.
  • Videlock EJ, Mahurkar-Joshi S, Hoffman JM, Iliopoulos D, Pothoulakis C, Mayer EA, Chang L. Sigmoid colon mucosal gene expression supports alterations of neuronal signaling in irritable bowel syndrome with constipation. Am J Physiol Gastrointestinal Liver Physiol. 2018;315(1):G140–g57. doi:10.1152/ajpgi.00288.2017.
  • Ren YJ, Zhang L, Bai T, Yu HL, Li Y, Qian W, Jin S, Xiong ZF, Wang H, Hou XH. Transfer of CD11c+ lamina propria mononuclear phagocytes from post-infectious irritable bowel syndrome causes mucosal barrier dysfunction and visceral hypersensitivity in recipient mice. International Journal of Molecular Medicine. 2017;39(6):1555–1563. doi:10.3892/ijmm.2017.2966.
  • Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, Granucci F, Kraehenbuhl JP, Ricciardi-Castagnoli P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001;2(4):361–367. doi:10.1038/86373.
  • Berg LK, Goll R, Fagerli E, Ludviksen JK, Fure H, Moen OS, Sørbye SW, Mollnes TE, Florholmen J. Intestinal inflammatory profile shows increase in a diversity of biomarkers in irritable bowel syndrome. Scandinavian J Gastroenterol. 2020;55(5):537–542. doi:10.1080/00365521.2020.1754455.
  • Singh M, Singh V, Schurman JV, Colombo JM, Friesen CA. The relationship between mucosal inflammatory cells, specific symptoms, and psychological functioning in youth with irritable bowel syndrome. Sci Rep. 2020;10(1):11988. doi:10.1038/s41598-020-68961-9.
  • Cohen JA, Edwards TN, Liu AW, Hirai T, Jones MR, Wu J, Li Y, Zhang S, Ho J, Davis BM, et al. Cutaneous TRPV1(+) neurons trigger protective innate type 17 anticipatory immunity. Cell. 2019;178(4):919–32.e14. doi:10.1016/j.cell.2019.06.022.
  • Belke K, Baron J, Schmutzler W, Zwadlo-Klarwasser G. Histidine decarboxylase expression in human monocytes, macrophages and macrophage subsets. International Archives of Allergy and Immunology. 1999;118(2–4):353–354. doi:10.1159/000024133.
  • Takamatsu S, Nakano K. Histamine synthesis by bone marrow-derived macrophages. BiosciBiotechnolBiochem. 1994;58:1918–1919.
  • Zwadlo-Klarwasser G, Braam U, Jungbluth C, Schmutzler W. Time-course of the histamine release from human peripheral blood monocytes and the influence of ketotifen and disodium cromoglycate (DSCG). Inflammation Research: Official Journal of the European Histamine Research Society [Et Al]. 1995;44(Suppl 1):S18–9. doi:10.1007/BF01674375.
  • Zwadlo-Klarwasser G, Vogts M, Hamann W, Belke K, Baron J, Schmutzler W. Generation and subcellular distribution of histamine in human blood monocytes and monocyte subsets. Inflammation Research: Official Journal of the European Histamine Research Society [Et Al]. 1998;47(11):434–439. doi:10.1007/s000110050357.
  • Lapiere A, Richard ML. Bacterial-fungal metabolic interactions within the microbiota and their potential relevance in human health and disease: a short review. Gut Microbes. 2022;14(1):2105610. doi:10.1080/19490976.2022.2105610.
  • Santus W, Devlin JR, Behnsen J. Crossing kingdoms: how the mycobiota and fungal-bacterial interactions impact host health and disease. Infection and Immunity. 2012;89(4):e00648–20.
  • Allescher HD, Burgell R, Malfertheiner P, Mearin F. Multi-target treatment for irritable bowel syndrome with STW 5: pharmacological modes of action. J Gastrointestin Liver Dis. 2020;29(2):227–233. doi:10.15403/jgld-814.
  • Ingrosso MR, Ianiro G, Nee J, Lembo AJ, Moayyedi P, Black CJ, Ford AC. Systematic review and meta-analysis: efficacy of peppermint oil in irritable bowel syndrome. Aliment Pharmacol Ther. 2022;56(6):932–941. doi:10.1111/apt.17179.
  • Major G, Pritchard S, Murray K, Alappadan JP, Hoad CL, Marciani L, Gowland P, Spiller R. Colon hypersensitivity to distension, rather than excessive gas production, produces carbohydrate-related symptoms in individuals with irritable bowel syndrome. Gastroenterology. 2017;152(1):124–33.e2. doi:10.1053/j.gastro.2016.09.062.
  • Staudacher HM, Whelan K. The low FODMAP diet: recent advances in understanding its mechanisms and efficacy in IBS. Gut. 2017;66(8):1517–1527. doi:10.1136/gutjnl-2017-313750.
  • Iliev ID, Cadwell K. Effects of intestinal fungi and viruses on immune responses and inflammatory bowel diseases. Gastroenterology. 2021;160(4):1050–1066. doi:10.1053/j.gastro.2020.06.100.
  • Hoffmann C, Dollive S, Grunberg S, Chen J, Li H, Wu GD, Lewis JD, Bushman FD. Archaea and fungi of the human gut microbiome: correlations with diet and bacterial residents. PloS one. 2013;8(6):e66019. doi:10.1371/journal.pone.0066019.
  • McFarland LV, Karakan T, Karatas A. Strain-specific and outcome-specific efficacy of probiotics for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. EClinicalMedicine. 2021;41:101154. doi:10.1016/j.eclinm.2021.101154.
  • Allonsius CN, van den Broek MFL, De Boeck I, Kiekens S, Oerlemans EFM, Kiekens F, Foubert K, Vandenheuvel D, Cos P, Delputte P, et al. Interplay between Lactobacillus rhamnosus GG and Candida and the involvement of exopolysaccharides. Microbial Biotechnology. 2017;10(6):1753–1763. doi:10.1111/1751-7915.12799.
  • Allonsius CN, Vandenheuvel D, Oerlemans EFM, Petrova MI, Donders GGG, Cos P, Delputte P, Lebeer S. Inhibition of Candida albicans morphogenesis by chitinase from Lactobacillus rhamnosus GG. Sci Rep. 2019;9(1):2900. doi:10.1038/s41598-019-39625-0.
  • Graf K, Last A, Gratz R, Allert S, Linde S, Westermann M, Gröger M, Mosig AS, Gresnigt MS, Hube B. Keeping Candida commensal: how lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model. Disease Models & Mechanisms. 2019;12(9):dmm039719.
  • Hasslöf P, Hedberg M, Twetman S, Stecksén-Blicks C. Growth inhibition of oral mutans streptococci and candida by commercial probiotic lactobacilli–an in vitro study. BMC Oral Health. 2010;10(1):18. doi:10.1186/1472-6831-10-18.
  • McFarland LV, Evans CT, Goldstein EJC. Strain-specificity and disease-specificity of probiotic efficacy: a systematic review and meta-analysis. Front Med (Lausanne). 2018;5:124. doi:10.3389/fmed.2018.00124.
  • Koutsoumanis K, Alvarez-Ordóñez A, Alvarez-Ordóñez A, Bolton D, Bover-Cid S, Chemaly M, Davies R, De Cesare A, Hilbert F, Lindqvist R, et al. Update of the list of QPS-recommended microbiological agents intentionally added to food or feed as notified to EFSA 16: suitability of taxonomic units notified to EFSA until March 2022. Efsa J. 2022;20(7):e07408. doi:10.2903/j.efsa.2022.7408.
  • Sen S, Mansell TJ. Yeasts as probiotics: mechanisms, outcomes, and future potential. Fungal Genetics and Biology: FG & B. 2020;137:103333. doi:10.1016/j.fgb.2020.103333.
  • Roselletti E, Sabbatini S, Ballet N, Perito S, Pericolini E, Blasi E, Mosci P, Cayzeele Decherf A, Monari C, Vecchiarelli A. Saccharomyces cerevisiae CNCM I-3856 as a new therapeutic agent against oropharyngeal candidiasis. Frontiers in Microbiology. 2019;10:1469. doi:10.3389/fmicb.2019.01469.