Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cells of the Nervous System".

Deadline for manuscript submissions: closed (20 March 2023) | Viewed by 27359

Special Issue Editors


E-Mail Website
Guest Editor
1. Laboratory of Traslational Mucosal Immunology, Vall dHebron Institut de Recerca, Dept Gastroenterol, Hospital vall d'Hebron. Paseo Vall dHebron 119-129, 08035 Barcelona, Spain
2. Department of Gastrointestinal Health, Société des produits Nestlé S.A., Nestlé Research, 1800 Lausanne, Switzerland
Interests: mucosal immunology; gut–brain axis; functional gastrointestinal disorders

E-Mail Website
Assistant Guest Editor
Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Mucosal Permeability Lab, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
Interests: epithelial barrier function; gut–brain axis; gut vascular barrier

E-Mail Website
Assistant Guest Editor
Vascular and renal translational research group, Lleida Institute for Biomedical Research, 25198 Lleida, Spain
Interests: host–microbiome interactions; epithelial barrier function; renal function

Special Issue Information

Dear Colleagues,

During the last two decades, we have witnessed an increasing recognition of environmental factors in shaping human physiology and tissue homeostasis. The major related breakthroughs have been the association of microbiome signatures with health and disease and the better understanding of the complex cell interactions that underlie host responses at mucosal sites. Within this circuitry, the nervous and the immune systems play a central role as they are essential for sensing external threats and for mounting regulatory and defensive responses. Neural and immune crosstalk is supported by a myriad of molecules, including neurotransmitters, neuropeptides, cytokines, hormones, and growth factors, and their corresponding receptors. Upstream signaling promotes protective responses or activates inflammatory activity and neural firing, which can be translated into changes in secretory activity, contractility, and pain signaling of the organ affected. Although technological advances have significantly contributed to unveil neuro-immune mechanisms, especially under pathological conditions, the biomedical community needs sensitive biomarkers for differential diagnosis as well as novel targets that constitute effective therapies. The increasing prevalence rates of certain entities such as inflammatory diseases, allergies, and functional gastrointestinal disorders are indeed a red flag for scientists and funding agencies to combine efforts to better understand neuro-immunity at mucosal interfaces. This Special Issue will contribute to a further comprehension of these mechanisms during homeostasis and under challenges that compromise organ function.

Dr. Maria Vicario
Guest Editor
Dr. Ricard Farré
Dr. Cristina Martinez
Assistant Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • inflammation
  • epithelial barrier
  • allergy
  • neuronal and glial sensing
  • pain
  • microbial metabolites
  • chronic stress
  • tissue homeostasis

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

17 pages, 2550 KiB  
Article
Acute Stress Regulates Sex-Related Molecular Responses in the Human Jejunal Mucosa: Implications for Irritable Bowel Syndrome
by Bruno K. Rodiño-Janeiro, Marc Pigrau, Eloísa Salvo-Romero, Adoración Nieto, Elba Expósito, Ana M. González-Castro, Carmen Galán, Inés de Torres, Teodora Pribic, Laura Hernández, Beatriz Lobo, Marina Fortea, Milagros Gallart, Cristina Pardo-Camacho, Danila Guagnozzi, Javier Santos and Carmen Alonso-Cotoner
Cells 2023, 12(3), 423; https://doi.org/10.3390/cells12030423 - 27 Jan 2023
Cited by 1 | Viewed by 2126
Abstract
Irritable bowel syndrome (IBS) is a prevalent gastrointestinal disorder linked to intestinal barrier dysfunction and life stress. We have previously reported that female sex per se determines an increased susceptibility to intestinal barrier dysfunction after cold pain stress (CPS). We aimed to identify [...] Read more.
Irritable bowel syndrome (IBS) is a prevalent gastrointestinal disorder linked to intestinal barrier dysfunction and life stress. We have previously reported that female sex per se determines an increased susceptibility to intestinal barrier dysfunction after cold pain stress (CPS). We aimed to identify sex-related molecular differences in response to CPS in healthy subjects to understand the origin of sex bias predominance in IBS. In 13 healthy males and 21 females, two consecutive jejunal biopsies were obtained using Watson’s capsule, at baseline, and ninety minutes after CPS. Total mucosal RNA and protein were isolated from jejunal biopsies. Expression of genes related to epithelial barrier (CLDN1, CLDN2, OCLN, ZO-1, and ZO-3), mast cell (MC) activation (TPSAB1, SERPINA1), and the glucocorticoid receptor (NR3C1) were analyzed using RT-qPCR. NR3C1, ZO-1 and OCLN protein expression were evaluated through immunohistochemistry and western blot, and mucosal inflammation through MC, lymphocyte, and eosinophil numbering. Autonomic, hormonal, and psychological responses to CPS were monitored. We found an increase in jejunal MCs, a reduced CLDN1 and OCLN expression, and an increased CLDN2 and SERPINA1 expression 90 min after CPS. We also found a significant decrease in ZO-1, OCLN, and NR3C1 gene expression, and a decrease in OCLN protein expression only in females, when compared to males. CPS induced a significant increase in blood pressure, plasma cortisol and ACTH, and subjective stress perception in all participants. Specific and independent sex-related molecular responses in epithelial barrier regulation are unraveled by acute stress in the jejunum of healthy subjects and may partially explain female predominance in IBS. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

19 pages, 2075 KiB  
Article
Mucosal Plasma Cell Activation and Proximity to Nerve Fibres Are Associated with Glycocalyx Reduction in Diarrhoea-Predominant Irritable Bowel Syndrome: Jejunal Barrier Alterations Underlying Clinical Manifestations
by Cristina Pardo-Camacho, John-Peter Ganda Mall, Cristina Martínez, Marc Pigrau, Elba Expósito, Mercé Albert-Bayo, Elisa Melón-Ardanaz, Adoración Nieto, Bruno Rodiño-Janeiro, Marina Fortea, Danila Guagnozzi, Amanda Rodriguez-Urrutia, Inés de Torres, Ignacio Santos-Briones, Fernando Azpiroz, Beatriz Lobo, Carmen Alonso-Cotoner, Javier Santos, Ana M. González-Castro and Maria Vicario
Cells 2022, 11(13), 2046; https://doi.org/10.3390/cells11132046 - 28 Jun 2022
Cited by 6 | Viewed by 2624
Abstract
Irritable bowel syndrome (IBS) is a disorder of brain-gut interaction characterised by abdominal pain and changes in bowel habits. In the diarrhoea subtype (IBS-D), altered epithelial barrier and mucosal immune activation are associated with clinical manifestations. We aimed to further evaluate plasma cells [...] Read more.
Irritable bowel syndrome (IBS) is a disorder of brain-gut interaction characterised by abdominal pain and changes in bowel habits. In the diarrhoea subtype (IBS-D), altered epithelial barrier and mucosal immune activation are associated with clinical manifestations. We aimed to further evaluate plasma cells and epithelial integrity to gain understanding of IBS-D pathophysiology. One mucosal jejunal biopsy and one stool sample were obtained from healthy controls and IBS-D patients. Gastrointestinal symptoms, stress, and depression scores were recorded. In the jejunal mucosa, RNAseq and gene set enrichment analyses were performed. A morphometric analysis by electron microscopy quantified plasma cell activation and proximity to enteric nerves and glycocalyx thickness. Immunoglobulins concentration was assessed in the stool. IBS-D patients showed differential expression of humoral pathways compared to controls. Activation and proximity of plasma cells to nerves and IgG concentration were also higher in IBS-D. Glycocalyx thickness was lower in IBS-D compared to controls, and this reduction correlated with plasma cell activation, proximity to nerves, and clinical symptoms. These results support humoral activity and loss of epithelial integrity as important contributors to gut dysfunction and clinical manifestations in IBS-D. Additional studies are needed to identify the triggers of these alterations to better define IBS-D pathophysiology. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

15 pages, 2484 KiB  
Article
A Galactooligosaccharide Product Decreases the Rotavirus Infection in Suckling Rats
by Malén Massot-Cladera, María del Mar Rigo-Adrover, Laura Herrero, Àngels Franch, Margarida Castell, Jelena Vulevic, Francisco J. Pérez-Cano and María J. Rodríguez Lagunas
Cells 2022, 11(10), 1669; https://doi.org/10.3390/cells11101669 - 18 May 2022
Cited by 4 | Viewed by 1871
Abstract
The leading cause of gastroenteritis among young children worldwide is the Group A rotaviruses (RV), which produce a wide range of symptoms, from a limited diarrhea to severe dehydration and even death. After an RV infection, immunity is not complete and less severe [...] Read more.
The leading cause of gastroenteritis among young children worldwide is the Group A rotaviruses (RV), which produce a wide range of symptoms, from a limited diarrhea to severe dehydration and even death. After an RV infection, immunity is not complete and less severe re-infections usually occur. These infections could be ameliorated by nutritional interventions with bioactive compounds, such as prebiotics. The aim of this research was to study the impact of a particular galactooligosaccharide (B-GOS) on the RV symptomatology and immune response during two consecutive infections. Lewis neonatal rats were inoculated with SA11 (first RV infection) on day 6 of life and with EDIM (second RV infection) on day 17 of life. B-GOS group was administered by oral gavage with a daily dose of B-GOS between days three to nine of life. Clinical and immunological variables were assessed during both infective processes. In the first infection, after the prebiotic intervention with B-GOS, a lower incidence, duration, and overall severity of the diarrhea (p < 0.05) was observed. In addition, it improved another severity indicator, the fecal weight output, during the diarrhea period (p < 0.05). The second RV infection failed in provoking diarrhea in the groups studied. The immune response during first infection with SA11 was not affected by B-GOS administration and had no impact on second infection, but the prebiotic intervention significantly increased IFN-γ and TNF-α intestinal production after the second infection (p < 0.05). In summary, B-GOS supplementation is able to reduce the incidence and severity of the RV-associated diarrhea and to influence the immune response against RV infections. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

17 pages, 3438 KiB  
Article
Estrogens Play a Critical Role in Stress-Related Gastrointestinal Dysfunction in a Spontaneous Model of Disorders of Gut–Brain Interaction
by Alison Accarie, Joran Toth, Lucas Wauters, Ricard Farré, Jan Tack and Tim Vanuytsel
Cells 2022, 11(7), 1214; https://doi.org/10.3390/cells11071214 - 4 Apr 2022
Cited by 6 | Viewed by 2924
Abstract
Background: Disorders of the gut–brain interaction (DGBI), such as irritable bowel syndrome and functional dyspepsia, are more prevalent in women than in men, with a ratio of 2:1. Furthermore, stressful life events have been reported as one of the triggers for symptoms in [...] Read more.
Background: Disorders of the gut–brain interaction (DGBI), such as irritable bowel syndrome and functional dyspepsia, are more prevalent in women than in men, with a ratio of 2:1. Furthermore, stressful life events have been reported as one of the triggers for symptoms in DGBI patients. Methods: Here, we studied the effect of an early-life stressor (maternal separation (MS)) on jejunal and colonic alterations, including colonic sensitivity and immune cells infiltration and activation in a validated spontaneous model of DGBI (BBDP-N), and investigated the involvement of β-estradiol on stress-worsened intestinal alterations. Results: We found that maternal separation exacerbated colonic sensitivity and mast cell and eosinophil infiltration and activation in females only. Ovariectomy partially rescued the stress phenotype by decreasing colonic sensitivity, which was restored by β-estradiol injections and did not impact immune cells infiltration and activation. Stressed males exposed to β-estradiol demonstrated similar intestinal alterations as MS females. Conclusion: Estrogen plays a direct critical role in colonic hypersensitivity in a spontaneous animal model of DGBI, while for immune activation, estrogen seems to be involved in the first step of their recruitment and activation. Our data point towards a complex interaction between stress and β-estradiol in DGBI. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

15 pages, 2054 KiB  
Article
Increased Numbers of Enteric Glial Cells in the Peyer’s Patches and Enhanced Intestinal Permeability by Glial Cell Mediators in Patients with Ileal Crohn’s Disease
by Olga Biskou, Felipe Meira de-Faria, Susanna M. Walter, Martin E. Winberg, Staffan Haapaniemi, Pär Myrelid, Johan D. Söderholm and Åsa V. Keita
Cells 2022, 11(3), 335; https://doi.org/10.3390/cells11030335 - 20 Jan 2022
Cited by 14 | Viewed by 3496
Abstract
Enteric glial cells (EGC) are known to regulate gastrointestinal functions; however, their role in Crohn’s disease (CD) is elusive. Microscopic erosions over the ileal Peyer’s patches are early signs of CD. The aim of this work was to assess the localization of EGC [...] Read more.
Enteric glial cells (EGC) are known to regulate gastrointestinal functions; however, their role in Crohn’s disease (CD) is elusive. Microscopic erosions over the ileal Peyer’s patches are early signs of CD. The aim of this work was to assess the localization of EGC in the follicle and interfollicular region of the Peyer’s patches and in the lamina propria and study the effects of EGC mediators on barrier function in CD patients and non-inflammatory bowel disease (non-IBD) controls. EGC markers, glial fibrillary acidic protein (GFAP), and S100 calcium-binding protein β (S100β) were quantified by immunofluorescence and Western blotting. Both markers showed significantly more EGC in the Peyer’s patches and lamina propria of CD patients compared to the non-IBD controls. In CD patients there were significantly more EGC in Peyer’s patches compared to lamina propria, while the opposite pattern was seen in controls. Barrier function studies using Ussing chambers showed increased paracellular permeability by EGC mediators in CD patients, whereas permeability decreased by the mediators in controls. We show the accumulation of EGC in Peyer’s patches of CD patients. Moreover, EGC mediators induced barrier dysfunction in CD patients. Thus, EGC might have harmful impacts on ongoing inflammation and contribute to the pathophysiology of the disease. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

16 pages, 3062 KiB  
Article
A Distinct Faecal Microbiota and Metabolite Profile Linked to Bowel Habits in Patients with Irritable Bowel Syndrome
by Bani Ahluwalia, Cristina Iribarren, Maria K. Magnusson, Johanna Sundin, Egbert Clevers, Otto Savolainen, Alastair B. Ross, Hans Törnblom, Magnus Simrén and Lena Öhman
Cells 2021, 10(6), 1459; https://doi.org/10.3390/cells10061459 - 10 Jun 2021
Cited by 26 | Viewed by 3889
Abstract
Patients with irritable bowel syndrome (IBS) are suggested to have an altered intestinal microenvironment. We therefore aimed to determine the intestinal microenvironment profile, based on faecal microbiota and metabolites, and the potential link to symptoms in IBS patients. The faecal microbiota was evaluated [...] Read more.
Patients with irritable bowel syndrome (IBS) are suggested to have an altered intestinal microenvironment. We therefore aimed to determine the intestinal microenvironment profile, based on faecal microbiota and metabolites, and the potential link to symptoms in IBS patients. The faecal microbiota was evaluated by the GA-mapTM dysbiosis test, and tandem mass spectrometry (GC-MS/MS) was used for faecal metabolomic profiling in patients with IBS and healthy subjects. Symptom severity was assessed using the IBS Severity Scoring System and anxiety and depression were assessed using the Hospital Anxiety and Depression Scale. A principal component analysis based on faecal microbiota (n = 54) and metabolites (n = 155) showed a clear separation between IBS patients (n = 40) and healthy subjects (n = 18). Metabolites were the main driver of this separation. Additionally, the intestinal microenvironment profile differed between IBS patients with constipation (n = 15) and diarrhoea (n = 11), while no clustering was detected in subgroups of patients according to symptom severity or anxiety. Furthermore, ingenuity pathway analysis predicted amino acid metabolism and several cellular and molecular functions to be altered in IBS patients. Patients with IBS have a distinct faecal microbiota and metabolite profile linked to bowel habits. Intestinal microenvironment profiling, based on faecal microbiota and metabolites, may be considered as a future non-invasive diagnostic tool, alongside providing valuable insights into the pathophysiology of IBS. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

Review

Jump to: Research

20 pages, 1582 KiB  
Review
Making Sense of Quorum Sensing at the Intestinal Mucosal Interface
by Friederike Uhlig and Niall P. Hyland
Cells 2022, 11(11), 1734; https://doi.org/10.3390/cells11111734 - 24 May 2022
Cited by 10 | Viewed by 3521
Abstract
The gut microbiome can produce metabolic products that exert diverse activities, including effects on the host. Short chain fatty acids and amino acid derivatives have been the focus of many studies, but given the high microbial density in the gastrointestinal tract, other bacterial [...] Read more.
The gut microbiome can produce metabolic products that exert diverse activities, including effects on the host. Short chain fatty acids and amino acid derivatives have been the focus of many studies, but given the high microbial density in the gastrointestinal tract, other bacterial products such as those released as part of quorum sensing are likely to play an important role for health and disease. In this review, we provide of an overview on quorum sensing (QS) in the gastrointestinal tract and summarise what is known regarding the role of QS molecules such as auto-inducing peptides (AIP) and acyl-homoserine lactones (AHL) from commensal, probiotic, and pathogenic bacteria in intestinal health and disease. QS regulates the expression of numerous genes including biofilm formation, bacteriocin and toxin secretion, and metabolism. QS has also been shown to play an important role in the bacteria–host interaction. We conclude that the mechanisms of action of QS at the intestinal neuro–immune interface need to be further investigated. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

18 pages, 2177 KiB  
Review
Eosinophils in the Gastrointestinal Tract: Key Contributors to Neuro-Immune Crosstalk and Potential Implications in Disorders of Brain-Gut Interaction
by Eloísa Salvo-Romero, Bruno K. Rodiño-Janeiro, Mercé Albert-Bayo, Beatriz Lobo, Javier Santos, Ricard Farré, Cristina Martinez and María Vicario
Cells 2022, 11(10), 1644; https://doi.org/10.3390/cells11101644 - 14 May 2022
Cited by 10 | Viewed by 5440
Abstract
Eosinophils are innate immune granulocytes actively involved in defensive responses and in local and systemic inflammatory processes. Beyond these effector roles, eosinophils are fundamental to maintaining homeostasis in the tissues they reside. Gastrointestinal eosinophils modulate barrier function and mucosal immunity and promote tissue [...] Read more.
Eosinophils are innate immune granulocytes actively involved in defensive responses and in local and systemic inflammatory processes. Beyond these effector roles, eosinophils are fundamental to maintaining homeostasis in the tissues they reside. Gastrointestinal eosinophils modulate barrier function and mucosal immunity and promote tissue development through their direct communication with almost every cellular component. This is possible thanks to the variety of receptors they express and the bioactive molecules they store and release, including cytotoxic proteins, cytokines, growth factors, and neuropeptides and neurotrophines. A growing body of evidence points to the eosinophil as a key neuro-immune player in the regulation of gastrointestinal function, with potential implications in pathophysiological processes. Eosinophil–neuron interactions are facilitated by chemotaxis and adhesion molecules, and the mediators released may have excitatory or inhibitory effects on each cell type, with physiological consequences dependent on the type of innervation involved. Of special interest are the disorders of the brain–gut interaction (DBGIs), mainly functional dyspepsia (FD) and irritable bowel syndrome (IBS), in which mucosal eosinophilia and eosinophil activation have been identified. In this review, we summarize the main roles of gastrointestinal eosinophils in supporting gut homeostasis and the evidence available on eosinophil–neuron interactions to bring new insights that support the fundamental role of this neuro-immune crosstalk in maintaining gut health and contributing to the pathophysiology of DBGIs. Full article
(This article belongs to the Special Issue Neuro-Immune Crosstalk at Mucosal Interfaces in Health and Disease)
Show Figures

Figure 1

Back to TopTop