1
|
Zhang M, Wu X, Gao H, Zhang L, Li Y, Li M, Zhao C, Wei P, Ou L. Chinese Herbal Medicine for Irritable Bowel Syndrome: A Perspective of Local Immune Actions. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2079-2106. [PMID: 39663262 DOI: 10.1142/s0192415x24500800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Irritable bowel syndrome (IBS) is the functional gastrointestinal disorder, characterized by abdominal pain and altered bowel habits. The interest in intestinal immune activation as a potential disease mechanism for IBS has increased exponentially in recent years. This study was designed to summarize the Chinese herbal medicine (CHM) that potentially exert protective effects against IBS through inhibition of intestinal immune activation. We detailed the current evidence that immune activation contributes to the pathology of IBS and discussed the potential mechanisms involved. Then, therapeutic effects and possible mechanisms related to immune response of herbal medicine prescriptions, extracts, and monomers were analyzed. The reasons leading to the aberrant and persistent immune activation noted in IBS are mainly associated with the increased number of mast cells, CD3[Formula: see text] T cells, and CD4[Formula: see text] T cells. The mechanisms mainly focused on the gut microbiota disorder induced alteration of the PGE2/COX2/SERT/5-HT, TLR4/MyD88/NF-κB, and BDNF/TrkB pathways. Most of the CHM alleviated IBS through interventions of intestinal immune activation via gut microbiota related to the TLR4/MyD88/NF-κB and SCF/c-kit pathways. We hope this review will provide some clues for the further development of novel candidate agents for IBS and other intestinal immune disorders.
Collapse
Affiliation(s)
- Mengmeng Zhang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Xu Wu
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
- Engineering Technology Research Center of Shaanxi, Administration of Chinese Herbal Pieces, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Huanqing Gao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Lin Zhang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Yao Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Min Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Chongbo Zhao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
- Engineering Technology Research Center of Shaanxi, Administration of Chinese Herbal Pieces, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Peifeng Wei
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Li Ou
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| |
Collapse
|
2
|
Lv J, Ibrahim YS, Yumashev A, Hjazi A, Faraz A, Alnajar MJ, Qasim MT, Ghildiyal P, Hussein Zwamel A, Fakri Mustafa Y. A comprehensive immunobiology review of IBD: With a specific glance to Th22 lymphocytes development, biology, function, and role in IBD. Int Immunopharmacol 2024; 137:112486. [PMID: 38901239 DOI: 10.1016/j.intimp.2024.112486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
The two primary forms of inflammatory disorders of the small intestine andcolon that make up inflammatory bowel disease (IBD) are ulcerative colitis (UC) and Crohn's disease (CD). While ulcerative colitis primarily affects the colon and the rectum, CD affects the small and large intestines, as well as the esophagus,mouth, anus, andstomach. Although the etiology of IBD is not completely clear, and there are many unknowns about it, the development, progression, and recurrence of IBD are significantly influenced by the activity of immune system cells, particularly lymphocytes, given that the disease is primarily caused by the immune system stimulation and activation against gastrointestinal (GI) tract components due to the inflammation caused by environmental factors such as viral or bacterial infections, etc. in genetically predisposed individuals. Maintaining homeostasis and the integrity of the mucosal barrier are critical in stopping the development of IBD. Specific immune system cells and the quantity of secretory mucus and microbiome are vital in maintaining this stability. Th22 cells are helper T lymphocyte subtypes that are particularly important for maintaining the integrity and equilibrium of the mucosal barrier. This review discusses the most recent research on these cells' biology, function, and evolution and their involvement in IBD.
Collapse
Affiliation(s)
- Jing Lv
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, PR China
| | - Yousif Saleh Ibrahim
- Department of Chemistry and Biochemistry, College of Medicine, University of Fallujah, Fallujah, Iraq
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Ali Faraz
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Majmaah 11952, Saudi Arabia.
| | | | - Maytham T Qasim
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar 64001, Iraq
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq; Medical Laboratory Technique College, The Islamic University of Aldiwaniyah, Aldiwaniyah, Iraq; Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
3
|
Sestak SS, da Motta Lima FG, de Oliveira AP, Barateiro LGRP, Vieira-Frez FC, de Souza SRG, Guarnier FA, Perles JVCM, Zanoni JN. Effects of cancer-induced cachexia and administration of L-glutathione on the intestinal mucosa in rat. Amino Acids 2024; 56:30. [PMID: 38607556 PMCID: PMC11009745 DOI: 10.1007/s00726-024-03391-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Walker-256 tumor is an experimental model known to promote cachexia syndrome, oxidative stress, and systemic inflammation. This study evaluated the duodenal mucosa of rats with Walker-256 tumor administered with 1% L-glutathione, intending to evaluate the damage caused by cancer-associated cachexia in the gastrointestinal tract and the effects of antioxidant administration on mucosal protection. Twenty-four 55-day-old male Wistar rats were distributed into four groups: control (C); control administered with 1% L-glutathione (C-GSH); Walker-256 tumor (W) and Walker-256 tumor administered with 1% L-glutathione (W-GSH). After 14 days of treatment, the duodenum was harvested for morphometric analysis of the mucosa, proliferation, apoptosis, immunostaining of varicosities immunoreactive (IR) to vasoactive intestinal peptide (VIP) and 5-HT-IR cells, and quantification of mast cells and goblet cells. Walker-256 tumor-bearing rats showed cachexia syndrome, mucosal atrophy, reduced cell proliferation, reduced 5-HT-IR cells, and increased goblet cells and VIPergic varicosities, which were not reversed by L-glutathione. On the other hand, L-glutathione caused a reduction of cells in apoptosis and mast cell recruitment, demonstrating a partial recovery of the damage detected in the intestinal mucosa.
Collapse
Affiliation(s)
- Sabrina Silva Sestak
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil
| | - Fabiana Galvão da Motta Lima
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil
| | - Ana Paula de Oliveira
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil
| | | | | | | | | | | | - Jacqueline Nelisis Zanoni
- Department of Physiology, Laboratory of Enteric Neural Plasticity, State University of Maringá, O33 Block, Colombo Avenue, 5790, Maringá, Paraná, CEP 87020-900, Brazil.
- Department of Morphological Sciences, State University of Maringá, Maringá, Paraná, Brazil.
| |
Collapse
|
4
|
Wang Y, Ullah H, Deng T, Ren X, Zhao Z, Xin Y, Qiu J. Social isolation induces intestinal barrier disorder and imbalances gut microbiota in mice. Neurosci Lett 2024; 826:137714. [PMID: 38479554 DOI: 10.1016/j.neulet.2024.137714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
Social isolation, a known stressor, can have detrimental effects on both physical and mental health. Recent scientific attention has been drawn to the gut-brain axis, a bidirectional communication system between the central nervous system and gut microbiota, suggesting that gut microbes may influence brain function. This study aimed to explore the impact of social isolation on the intestinal barrier and gut microbiota. 40 male BALB/c mice were either individually housed or kept in groups for 8 and 15 weeks. Socially isolated mice exhibited increased anxiety-like behavior, with significant differences between the 8-week and 15-week isolation groups (P < 0.05). After 8 weeks of isolation, there was a reduction in tight junction protein expression in the intestinal mechanical barrier. Furthermore, after 15 weeks of isolation, both tight junction protein and mucin expression, key components of the intestinal chemical barrier, decreased. This was accompanied by a substantial increase in inflammatory cytokines (IL-6 mRNA, IL-10, and TNF-α) in colon tissue in the 15-week isolated group (P < 0.05). Additionally, Illumina MiSequencing revealed significant alterations in the gut microbiota of socially isolated mice, including reduced Firmicutes and Bacteroides compared to the control group. Lactobacillus levels also decreased in the socially isolated mice.
Collapse
Affiliation(s)
- Yue Wang
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Hidayat Ullah
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Ting Deng
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Xinxiu Ren
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Zinan Zhao
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Yi Xin
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Juanjuan Qiu
- Central Lab, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
5
|
La Torre D, Van Oudenhove L, Vanuytsel T, Verbeke K. Psychosocial stress-induced intestinal permeability in healthy humans: What is the evidence? Neurobiol Stress 2023; 27:100579. [PMID: 37842017 PMCID: PMC10569989 DOI: 10.1016/j.ynstr.2023.100579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023] Open
Abstract
An impaired intestinal barrier function can be detrimental to the host as it may allow the translocation of luminal antigens and toxins into the subepithelial tissue and bloodstream. In turn, this may cause local and systemic immune responses and lead to the development of pathologies. In vitro and animal studies strongly suggest that psychosocial stress is one of the factors that can increase intestinal permeability via mast-cell dependent mechanisms. Remarkably, studies have not been able to yield unequivocal evidence that such relation between stress and intestinal permeability also exists in (healthy) humans. In the current Review, we discuss the mechanisms that are involved in stress-induced intestinal permeability changes and postulate factors that influence these alterations and that may explain the translational difficulties from in vitro and animal to human studies. As human research differs highly from animal research in the extent to which stress can be applied and intestinal permeability can be measured, it remains difficult to draw conclusions about the presence of a relation between stress and intestinal permeability in (healthy) humans. Future studies should bear in mind these difficulties, and more research into in vivo methods to assess intestinal permeability are warranted.
Collapse
Affiliation(s)
- Danique La Torre
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Lukas Van Oudenhove
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Cognitive and Affective Neuroscience Lab, Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Tim Vanuytsel
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Division of Gastroenterology and Hepatology, Leuven University Hospital, Leuven, Belgium
| | - Kristin Verbeke
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Leigh SJ, Uhlig F, Wilmes L, Sanchez-Diaz P, Gheorghe CE, Goodson MS, Kelley-Loughnane N, Hyland NP, Cryan JF, Clarke G. The impact of acute and chronic stress on gastrointestinal physiology and function: a microbiota-gut-brain axis perspective. J Physiol 2023; 601:4491-4538. [PMID: 37756251 DOI: 10.1113/jp281951] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The physiological consequences of stress often manifest in the gastrointestinal tract. Traumatic or chronic stress is associated with widespread maladaptive changes throughout the gut, although comparatively little is known about the effects of acute stress. Furthermore, these stress-induced changes in the gut may increase susceptibility to gastrointestinal disorders and infection, and impact critical features of the neural and behavioural consequences of the stress response by impairing gut-brain axis communication. Understanding the mechanisms behind changes in enteric nervous system circuitry, visceral sensitivity, gut barrier function, permeability, and the gut microbiota following stress is an important research objective with pathophysiological implications in both neurogastroenterology and psychiatry. Moreover, the gut microbiota has emerged as a key aspect of physiology sensitive to the effects of stress. In this review, we focus on different aspects of the gastrointestinal tract including gut barrier function as well as the immune, humoral and neuronal elements involved in gut-brain communication. Furthermore, we discuss the evidence for a role of stress in gastrointestinal disorders. Existing gaps in the current literature are highlighted, and possible avenues for future research with an integrated physiological perspective have been suggested. A more complete understanding of the spatial and temporal dynamics of the integrated host and microbial response to different kinds of stressors in the gastrointestinal tract will enable full exploitation of the diagnostic and therapeutic potential in the fast-evolving field of host-microbiome interactions.
Collapse
Affiliation(s)
- Sarah-Jane Leigh
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Friederike Uhlig
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
| | - Lars Wilmes
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Paula Sanchez-Diaz
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Cassandra E Gheorghe
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Michael S Goodson
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, Ohio, USA
| | - Nancy Kelley-Loughnane
- Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, Ohio, USA
| | - Niall P Hyland
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| |
Collapse
|
7
|
Wu W, Ma M, Ibarra AE, Lu G, Bakshi VP, Li L. Global Neuropeptidome Profiling in Response to Predator Stress in Rat: Implications for Post-Traumatic Stress Disorder. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:1549-1558. [PMID: 37405781 PMCID: PMC11731200 DOI: 10.1021/jasms.3c00027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Traumatic stress triggers or exacerbates multiple psychiatric illnesses, including post-traumatic stress disorder (PTSD). Nevertheless, the neurophysiological mechanisms underlying stress-induced pathology remain unclear, in part due to the limited understanding of neuronal signaling molecules, such as neuropeptides, in this process. Here, we developed mass spectrometry (MS)-based qualitative and quantitative analytical strategies to profile neuropeptides in rats exposed to predator odor (an ethologically relevant analogue of trauma-like stress) versus control subjects (no odor) to determine peptidomic alterations induced by trauma. In total, 628 unique neuropeptides were identified across 5 fear-circuitry-related brain regions. Brain-region-specific changes of several neuropeptide families, including granin, ProSAAS, opioids, cholecystokinin, and tachykinin, were also observed in the stressed group. Neuropeptides from the same protein precursor were found to vary across different brain regions, indicating the site-specific effects of predator stress. This study reveals for the first time the interaction between neuropeptides and traumatic stress, providing insights into the molecular mechanisms of stress-induced psychopathology and suggesting putative novel therapeutic strategies for disorders such as PTSD.
Collapse
Affiliation(s)
- Wenxin Wu
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Min Ma
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Angel Erbey Ibarra
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Gaoyuan Lu
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Vaishali P. Bakshi
- Department of Psychiatry, University of Wisconsin–Madison, Madison, WI 53719, United States
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53705, United States
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705, United States
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States
| |
Collapse
|
8
|
González-Vergara A, Benavides B, Julio-Pieper M. Mapping and quantifying neuropeptides in the enteric nervous system. J Neurosci Methods 2023; 393:109882. [PMID: 37172914 DOI: 10.1016/j.jneumeth.2023.109882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Neuropeptides are a highly diverse group of signaling molecules found in the central nervous system (CNS) and peripheral organs, including the enteric nervous system (ENS). Increasing efforts have been focused on dissecting the role of neuropeptides in both neural- and non-neural-related diseases, as well as their potential therapeutic value. In parallel, accurate knowledge on their source of production and pleiotropic functions is still needed to fully understand their implications in biological processes. This review will focus on the analytical challenges involved in studying neuropeptides, particularly in the ENS, a tissue where their abundance is low, together with opportunities for further technical development.
Collapse
Affiliation(s)
- Alex González-Vergara
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Benjamín Benavides
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
9
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
10
|
Kanamori A, Tanaka F, Takashima S, Sawada A, Ominami M, Nadatani Y, Fukunaga S, Otani K, Hosomi S, Kamata N, Nagami Y, Taira K, Watanabe T, Fujiwara Y. Esophageal mast cells may be associated with the perception of symptoms in patients with eosinophilic esophagitis. Esophagus 2023; 20:333-341. [PMID: 36342564 DOI: 10.1007/s10388-022-00967-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a type 2 helper T-cell (Th2)-mediated allergic disease that involves mast cells. This study aimed to clarify the relationship between perception of symptoms and mast cell levels in patients with EoE. METHODS We enrolled patients with asymptomatic esophageal eosinophilia (aEE) and those with symptomatic EoE. Immunofluorescence staining was performed on esophageal biopsy specimens to quantify mast cell-related molecules, such as tryptase, proteinase-activated receptor (PAR)-2, and vasoactive intestinal peptide receptor (VPAC)-1. RESULTS We evaluated 28 and 58 patients with aEE and EoE, respectively. There were no significant differences in clinical and endoscopic features and peak eosinophil counts between both groups. Mast cell tryptase-positive areas were significantly higher in EoE than in aEE (4.9 [3.5-6.2] vs. 2.0 [1.2-3.4] %, p < 0.01). The number of PAR-2-positive cells was significantly higher in EoE than in aEE (14 [8.8-20.0] vs. 4 [2.8-8.0] cells/high-power field [HPF], p < 0.01). The number of VPAC-1-positive cells was significantly higher in the EoE group than in the aEE group (13 [8.8-16.0] vs. 6 [3.0-9.3] cells/HPF, p < 0.01). A positive correlation was observed between the numbers of PAR-2-positive cells and VPAC-1-positive cells (r = 0.851, p < 0.01). Moreover, mast cell tryptase-positive areas positively correlated with the number of PAR-2- and VPAC-1-positive cells (r = 0.352, p < 0.01; r = 0.355, p < 0.01, respectively). CONCLUSIONS Esophageal mast cells and their receptors, PAR-2 and VPAC-1, may contribute to the perception of symptoms in patients with EoE.
Collapse
Affiliation(s)
- Atsushi Kanamori
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Fumio Tanaka
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Shingo Takashima
- Department of Premier Preventive Medicine/MedCity21, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Akinari Sawada
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masaki Ominami
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yuji Nadatani
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
- Department of Premier Preventive Medicine/MedCity21, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Shusei Fukunaga
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Koji Otani
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shuhei Hosomi
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Noriko Kamata
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yasuaki Nagami
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Koichi Taira
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Toshio Watanabe
- Department of Premier Preventive Medicine/MedCity21, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
11
|
Hanč P, Messou MA, Wang Y, von Andrian UH. Control of myeloid cell functions by nociceptors. Front Immunol 2023; 14:1127571. [PMID: 37006298 PMCID: PMC10064072 DOI: 10.3389/fimmu.2023.1127571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
The immune system has evolved to protect the host from infectious agents, parasites, and tumor growth, and to ensure the maintenance of homeostasis. Similarly, the primary function of the somatosensory branch of the peripheral nervous system is to collect and interpret sensory information about the environment, allowing the organism to react to or avoid situations that could otherwise have deleterious effects. Consequently, a teleological argument can be made that it is of advantage for the two systems to cooperate and form an “integrated defense system” that benefits from the unique strengths of both subsystems. Indeed, nociceptors, sensory neurons that detect noxious stimuli and elicit the sensation of pain or itch, exhibit potent immunomodulatory capabilities. Depending on the context and the cellular identity of their communication partners, nociceptors can play both pro- or anti-inflammatory roles, promote tissue repair or aggravate inflammatory damage, improve resistance to pathogens or impair their clearance. In light of such variability, it is not surprising that the full extent of interactions between nociceptors and the immune system remains to be established. Nonetheless, the field of peripheral neuroimmunology is advancing at a rapid pace, and general rules that appear to govern the outcomes of such neuroimmune interactions are beginning to emerge. Thus, in this review, we summarize our current understanding of the interaction between nociceptors and, specifically, the myeloid cells of the innate immune system, while pointing out some of the outstanding questions and unresolved controversies in the field. We focus on such interactions within the densely innervated barrier tissues, which can serve as points of entry for infectious agents and, where known, highlight the molecular mechanisms underlying these interactions.
Collapse
Affiliation(s)
- Pavel Hanč
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- *Correspondence: Pavel Hanč, ; Ulrich H. von Andrian,
| | - Marie-Angèle Messou
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Yidi Wang
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Ulrich H. von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- *Correspondence: Pavel Hanč, ; Ulrich H. von Andrian,
| |
Collapse
|
12
|
Gerdin L, González‐Castro AM, Ericson A, Persborn M, Santos J, Walter SA, Keita ÅV, Vicario M, Söderholm JD. Acute psychological stress increases paracellular permeability and modulates immune activity in rectal mucosa of healthy volunteers. United European Gastroenterol J 2022; 11:31-41. [PMID: 36314901 PMCID: PMC9892435 DOI: 10.1002/ueg2.12329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Psychological stress and increased permeability are implicated as contributing factors in the initiation and worsening of gastrointestinal diseases. A link between stress and intestinal permeability has been shown in animal models as well as in human small intestine, but stress effects on the human colorectal mucosal barrier has not been reported. OBJECTIVE To investigate the potential effects of acute psychological stress on colorectal mucosal barrier function and to explore stress-induced molecular events in the rectal mucosa under healthy conditions. METHODS Endoscopic biopsies were taken from the rectosigmoid region of healthy volunteers, who had been subjected to dichotomous listening stress and after a control session, respectively. Paracellular and transcellular permeability were assessed in modified Ussing chambers. RNA expression (microarray technology confirmed by quantitative real-time polymerase chain reaction) and biological pathway analysis were used to investigate the local mucosal response to acute stress. RESULTS Dichotomous listening stress induced a subjective and objective stress response, and significantly increased paracellular but not transcellular permeability. We also identified a stress-induced reduction in RNA expression of genes related to immune cell activation and maturation (CR2, CD20, TCLA1, BANK1, CD22, FDCSP), signaling molecules of homing of immune cells to the gut (chemokines: CCL21, CXCL13, and CCL19, and receptors: CCR7, CXCR5), and innate immunity (DUOX2). Eight of the 10 top down-regulated genes are directly involved in B cell activation, signaling and migration. The systemic stress response correlated positively with paracellular permeability and negatively with DUOX2 expression. CONCLUSION Dichotomous listening stress increases paracellular permeability and modulates immune cell activity in the rectal mucosa. Further studies are warranted to identify the primary mechanisms of stress-mediated reduction of mucosal defensive activity and barrier dysfunction, and their potential implications for gastrointestinal disorders.
Collapse
Affiliation(s)
- Linda Gerdin
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden,Department of SurgeryLinköping UniversityLinköpingSweden,Surgical Clinic of Jönköping CountyJönköpingSweden
| | - Ana M. González‐Castro
- Laboratory of Translational Mucosal ImmunologyDigestive Diseases Research UnitVall d'Hebron Institut de RecercaHospital Universitari Vall d'Hebron & Facultat de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Ann‐Charlott Ericson
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden,Department of SurgeryLinköping UniversityLinköpingSweden
| | | | - Javier Santos
- Laboratory of Neuro‐immuno‐gastroenterologyDigestive Diseases Research UnitVall d'Hebron Institut de RecercaHospital Universitari Vall d'Hebron & Facultat de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain
| | - Susanna A. Walter
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden,Department of SurgeryLinköping UniversityLinköpingSweden,Department of GastroenterologyLinköping UniversityLinköpingSweden
| | - Åsa V. Keita
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden,Department of SurgeryLinköping UniversityLinköpingSweden
| | - Maria Vicario
- Laboratory of Translational Mucosal ImmunologyDigestive Diseases Research UnitVall d'Hebron Institut de RecercaHospital Universitari Vall d'Hebron & Facultat de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain,Department of Gastrointestinal HealthNestlé Institute of Health SciencesNestlé Research, Société des Produits Nestlé S.A.LausanneSwitzerland
| | - Johan D. Söderholm
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden,Department of SurgeryLinköping UniversityLinköpingSweden
| |
Collapse
|
13
|
Xu D, Wu F, Yu Y, Lou X, Ye M, Zhang H, Zhao Y. Sympathetic activation leads to Schlemm's canal expansion via increasing vasoactive intestinal polypeptide secretion from trabecular meshwork. Exp Eye Res 2022; 224:109235. [PMID: 36049555 DOI: 10.1016/j.exer.2022.109235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/04/2022]
Abstract
We previously demonstrated vasoactive intestinal polypeptide (VIP) eyedrops reduce intraocular pressure (IOP) and stabilize cytoskeleton of the Schlemm's canal (SC) endothelium in a chronic ocular hypertension rat model. Here we determine if the trabecular meshwork (TM) releases endogenous VIP and affect SC in paracrine manner, and whether this cellular interaction via VIP is strengthened under stimulated sympathetic activity. A rat model of moderate-intensity exercise was established to stimulate sympathetic activation. IOP post exercise was measured by a rebound tonometer. Sympathetic nerve activity at the TM was immunofluorescence-stained with DβH and PGP9.5. Morphological changes of TM and SC were quantitatively measured by hematoxylin-eosin (HE) staining. Further, epinephrine was applied to mimic sympathetic excitation on primary rat TM cells, and ELISA to measure VIP levels in the medium. The cytoskeleton protective effect of VIP in the epinephrine-stimulated conditioned medium (Epi-CM) was evaluated in oxidative stressed human umbilical vein endothelial cells (HUVECs). Elevated sympathetic nerve activity was found at TM post exercise. Changes accompanying the sympathetic excitation included thinned TM, expanded SC and decreased IOP, which were consistent with epinephrine treatment. Epinephrine decreased TM cell size, enhanced VIP expression and release in the medium in vitro. Epi-CM restored linear F-actin and cell junction integrity in H2O2 treated HUVECs. Blockage of VIP receptor by PG99-465 attenuated the protective capability of Epi-CM. VIP expression was upregulated at TM and the inner wall of SC post exercise in vivo. PG99-465 significantly attenuated exercise-induced SC expansion and IOP reduction. Thus, the sympathetic activation promoted VIP release from TM cells and subsequently expanded SC via stabilizing cytoskeleton, which resulted in IOP reduction.
Collapse
Affiliation(s)
- Dingwen Xu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feipeng Wu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yixian Yu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaotong Lou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng Ye
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
14
|
Biskou O, Meira de-Faria F, Walter SM, Winberg ME, Haapaniemi S, Myrelid P, Söderholm JD, Keita ÅV. 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. Cells 2022; 11:335. [PMID: 35159145 PMCID: PMC8833935 DOI: 10.3390/cells11030335] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/22/2022] Open
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 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.
Collapse
Affiliation(s)
- Olga Biskou
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden; (O.B.); (F.M.d.-F.); (S.M.W.); (M.E.W.); (P.M.); (J.D.S.)
| | - Felipe Meira de-Faria
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden; (O.B.); (F.M.d.-F.); (S.M.W.); (M.E.W.); (P.M.); (J.D.S.)
| | - Susanna M. Walter
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden; (O.B.); (F.M.d.-F.); (S.M.W.); (M.E.W.); (P.M.); (J.D.S.)
- Department of Gastroenterology, Linköping University, 58185 Linköping, Sweden
| | - Martin E. Winberg
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden; (O.B.); (F.M.d.-F.); (S.M.W.); (M.E.W.); (P.M.); (J.D.S.)
| | | | - Pär Myrelid
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden; (O.B.); (F.M.d.-F.); (S.M.W.); (M.E.W.); (P.M.); (J.D.S.)
- Department of Surgery, Linköping University Hospital, 58185 Linköping, Sweden
| | - Johan D. Söderholm
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden; (O.B.); (F.M.d.-F.); (S.M.W.); (M.E.W.); (P.M.); (J.D.S.)
- Department of Surgery, Linköping University Hospital, 58185 Linköping, Sweden
| | - Åsa V. Keita
- Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden; (O.B.); (F.M.d.-F.); (S.M.W.); (M.E.W.); (P.M.); (J.D.S.)
| |
Collapse
|
15
|
Struffert M, Maier C, Neid M, Schäfer HL, Tannapfel A, Schmidt-Choudhury A. Duodenal mast cells and eosinophils in children with celiac disease: occurrence and distribution pattern. Scand J Gastroenterol 2022; 57:22-30. [PMID: 34618623 DOI: 10.1080/00365521.2021.1985601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The aim of this study was to characterize duodenal mast cell (MC) and eosinophil (EO) numbers, their distribution within the lamina propria and possible impact on disease severity of paediatric celiac patients compared to children without celiac disease (CD). METHODS We analysed duodenal samples of 215 children (109 CD, 106 controls) who underwent esophagogastroduodenoscopy from 2010 to 2018. After immunohistochemical staining, average MC and EO counts were histologically examined in ten high-power-fields. Additionally, cell-distribution within the lamina propria was analysed. Possible influence of relevant clinical parameters was evaluated. STATISTICS Student's-t-test, Mann-Whitney U-test, Chi-square-test, ANOVA, significance-level <.05. Trial registration-number: DRKS00024669. RESULTS MC-density was higher in CD-patients compared to the control-group (23.7 (±12.1)/HPF versus 19.7 (±9.1)/HPF; p = .008), varying in number interindividually. Eosinophils were also increased in the duodenum of celiac patients (23.3 (±9.3)/HPF versus 12.2 (±6.3)/HPF; p= <.001). MCs were distributed more often homogenously in all parts of CD lamina propria (44 biopsies (40.4%), residing more distant from the intestinal lumen in controls (0 biopsies with homogenous distribution-pattern (0%); p= <.001). Regarding EOs no polarity was observable. Atopic diseases did not occur significantly more often in patients with elevated EO-counts. CONCLUSION MC- and EO-numbers were increased in the duodenum of CD-patients and MCs showed a different distribution-pattern in the lamina propria of celiac patients. These findings support the concept that both cell-types contribute to disease-pathogenesis. However, functional studies highlighting both cell-types' and their mediators' role regarding mucosal alterations during the course of the inflammatory process in celiac patients are needed. TRIAL REGISTRATION NUMBER AND URL DRKS00024669; https://www.drks.de/drks_web/.
Collapse
Affiliation(s)
- Marie Struffert
- Department of Pediatric Gastroenterology, St. Josef-Hospital, University Hospital of Pediatrics and Adolescent Medicine, Ruhr-University, Bochum, Germany
| | - Christoph Maier
- Department of Pediatric Gastroenterology, St. Josef-Hospital, University Hospital of Pediatrics and Adolescent Medicine, Ruhr-University, Bochum, Germany
| | - Matthias Neid
- Institute of Pathology, Ruhr-University, Bochum, Germany
| | - Hannah-Lena Schäfer
- Department of Pediatric Gastroenterology, St. Josef-Hospital, University Hospital of Pediatrics and Adolescent Medicine, Ruhr-University, Bochum, Germany
| | | | - Anjona Schmidt-Choudhury
- Department of Pediatric Gastroenterology, St. Josef-Hospital, University Hospital of Pediatrics and Adolescent Medicine, Ruhr-University, Bochum, Germany
| |
Collapse
|
16
|
Song X, Pi S, Gao Y, Zhou F, Yan S, Chen Y, Qiao L, Dou X, Shao D, Xu C. The Role of Vasoactive Intestinal Peptide and Mast Cells in the Regulatory Effect of Lactobacillus casei ATCC 393 on Intestinal Mucosal Immune Barrier. Front Immunol 2021; 12:723173. [PMID: 34899686 PMCID: PMC8657605 DOI: 10.3389/fimmu.2021.723173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/09/2021] [Indexed: 01/07/2023] Open
Abstract
Vasoactive intestinal peptide (VIP) plays an important role in the neuro-endocrine-immune system. Mast cells (MCs) are important immune effector cells. This study was conducted to investigate the protective effect of L. casei ATCC 393 on Enterotoxigenic Escherichia coli (ETEC) K88-induced intestinal mucosal immune barrier injury and its association with VIP/MC signaling by in vitro experiments in cultures of porcine mucosal mast cells (PMMCs) and in vivo experiments using VIP receptor antagonist (aVIP) drug. The results showed that compared with the ETEC K88 and lipopolysaccharides (LPS)-induced model groups, VIP pretreatment significantly inhibited the activation of MCs and the release of β-hexosaminidase (β-hex), histamine and tryptase. Pretreatment with aVIP abolished the protective effect of L. casei ATCC 393 on ETEC K88-induced intestinal mucosal immune barrier dysfunction in C57BL/6 mice. Also, with the blocking of VIP signal transduction, the ETEC K88 infection increased serum inflammatory cytokines, and the numbers of degranulated MCs in ileum, which were decreased by administration of L. casei ATCC 393. In addition, VIP mediated the regulatory effect of L. casei ATCC 393 on intestinal microbiota in mice. These findings suggested that VIP may mediate the protective effect of L.casei ATCC 393 on intestinal mucosal immune barrier dysfunction via MCs.
Collapse
Affiliation(s)
- Xiaofan Song
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Shanyao Pi
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yueming Gao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Fengxia Zhou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Shuqi Yan
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yue Chen
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Dongyan Shao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
17
|
Meira de-Faria F, Casado-Bedmar M, Mårten Lindqvist C, Jones MP, Walter SA, Keita ÅV. Altered interaction between enteric glial cells and mast cells in the colon of women with irritable bowel syndrome. Neurogastroenterol Motil 2021; 33:e14130. [PMID: 33797165 DOI: 10.1111/nmo.14130] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Enteric glial cells (EGC) and mast cells (MC) are intimately associated with gastrointestinal physiological functions. We aimed to investigate EGC-MC interaction in irritable bowel syndrome (IBS), a gut-brain disorder linked to increased intestinal permeability, and MC. METHODS Parallel approaches were used to quantify EGC markers in colonic biopsies from healthy controls (HC) and patients with IBS. Data were correlated with MC, vasoactive intestinal polypeptide (VIP) and VIP receptors (VPAC1/VPAC2) expressions, and bacterial translocation through biopsies mounted in Ussing chambers. In addition, we investigated the effects of EGC mediators on colonic permeability and the pharmacological-induced responses of EGC and MC cell lines. KEY RESULTS Immunofluorescence of IBS colonic mucosa, as well as Western blotting and ELISA of IBS biopsy lysates, revealed increased glial fibrillary intermediate filament (GFAP) expression, indicating EGC activation. Mucosal GFAP correlated with increased MC and VPAC1+ MC numbers and decreased VIP+ MC, which seemed to control bacterial translocation in HC. In the contrary, EGC activation in IBS correlated with less MC and VPAC1+ MC numbers, and more VIP+ MC. In vitro, MC and EGC cell lines showed intracellular calcium responses to each other's mediators. Furthermore, EGC mediators prevented VIP-induced MC degranulation, while MC mediators induced a reactive EGC phenotype. In Ussing chambers, EGC mediators decreased paracellular passage through healthy colonic biopsies. CONCLUSIONS & INFERENCES Findings suggest the involvement of EGC and MC in the control of barrier function in the human colon and indicate a potential EGC-MC interaction that seems altered in IBS, with detrimental consequences to colonic permeability. Altogether, results suggest that imbalanced EGC-MC communication contributes to the pathophysiology of IBS.
Collapse
Affiliation(s)
- Felipe Meira de-Faria
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maite Casado-Bedmar
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Carl Mårten Lindqvist
- Department of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | | | - Susanna A Walter
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Gastroenterology, Linköping University, Linköping, Sweden
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
18
|
Fortea M, Albert-Bayo M, Abril-Gil M, Ganda Mall JP, Serra-Ruiz X, Henao-Paez A, Expósito E, González-Castro AM, Guagnozzi D, Lobo B, Alonso-Cotoner C, Santos J. Present and Future Therapeutic Approaches to Barrier Dysfunction. Front Nutr 2021; 8:718093. [PMID: 34778332 PMCID: PMC8582318 DOI: 10.3389/fnut.2021.718093] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
There is converging and increasing evidence, but also uncertainty, for the role of abnormal intestinal epithelial barrier function in the origin and development of a growing number of human gastrointestinal and extraintestinal inflammatory disorders, and their related complaints. Despite a vast literature addressing factors and mechanisms underlying changes in intestinal permeability in humans, and its connection to the appearance and severity of clinical symptoms, the ultimate link remains to be established in many cases. Accordingly, there are no directives or clinical guidelines related to the therapeutic management of intestinal permeability disorders that allow health professionals involved in the management of these patients to carry out a consensus treatment based on clinical evidence. Instead, there are multiple pseudoscientific approaches and commercial propaganda scattered on the internet that confuse those affected and health professionals and that often lack scientific rigor. Therefore, in this review we aim to shed light on the different therapeutic options, which include, among others, dietary management, nutraceuticals and medical devices, microbiota and drugs, and epigenetic and exosomes-manipulation, through an objective evaluation of the scientific publications in this field. Advances in the knowledge and management of intestinal permeability will sure enable better options of dealing with this group of common disorders to enhance quality of life of those affected.
Collapse
Affiliation(s)
- Marina Fortea
- Laboratory for Enteric NeuroScience, Translational Research Center for GastroIntestinal Disorders, University of Leuven, Leuven, Belgium
| | - Mercé Albert-Bayo
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Mar Abril-Gil
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - John-Peter Ganda Mall
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Xavier Serra-Ruiz
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Alejandro Henao-Paez
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Elba Expósito
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Ana María González-Castro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Danila Guagnozzi
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHED), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Lobo
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Carmen Alonso-Cotoner
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHED), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Santos
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Gastroenterology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Facultad de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Schwerdtfeger LA, Tobet SA. Sex differences in anatomic plasticity of gut neuronal-mast cell interactions. Physiol Rep 2021; 9:e15066. [PMID: 34605201 PMCID: PMC8488573 DOI: 10.14814/phy2.15066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 11/24/2022] Open
Abstract
The gut wall houses mast cells that are anatomically situated near enteric neuronal fibers. Roles of specific neuropeptides in modulating function of immune components like mast cells in response to challenge with bacterial components are relatively unknown. Investigating such interactions requires models that include diverse cellular elements in native anatomic arrangements. Using an organotypic slice model that maintains gut wall cellular diversity ex vivo, the present study compared responses between tissues derived from male and female mice to examine neural-immune signaling in the gut wall after selected treatments. Ileum slices were treated with pharmacological reagents that block neuronal function (e.g., tetrodotoxin) or vasoactive intestinal peptide (VIP) receptors prior to challenge with lipopolysaccharide (LPS) to assess their influence on anatomic plasticity of VIP fibers and activation of mast cells. Sex differences were observed in the number of mucosal mast cells (c-kit/ACK2 immunoreactive) at baseline, regardless of treatment, with female ileum tissue having 46% more ACK2-IR mast cells than males. After challenge with LPS, male mast cell counts rose to female levels. Furthermore, sex differences were observed in the percentage of ACK2-IR cells within 1 µm of a VIP+ neuronal fiber, and mast cell size, a metric previously tied to activation, with females having larger cells at baseline. Male mast cell sizes reached female levels after LPS challenge. This study suggests sex differences in neural-immune plasticity and in mast cell activation both basally and in response to challenge with LPS. These sex differences could potentially impact functional neuroimmune response to pathogens.
Collapse
Affiliation(s)
| | - Stuart A. Tobet
- Department of Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
- School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|
20
|
Muntjewerff EM, Tang K, Lutter L, Christoffersson G, Nicolasen MJT, Gao H, Katkar GD, Das S, ter Beest M, Ying W, Ghosh P, El Aidy S, Oldenburg B, van den Bogaart G, Mahata SK. Chromogranin A regulates gut permeability via the antagonistic actions of its proteolytic peptides. Acta Physiol (Oxf) 2021; 232:e13655. [PMID: 33783968 DOI: 10.1111/apha.13655] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
AIM A "leaky" gut barrier has been implicated in the initiation and progression of a multitude of diseases, for example, inflammatory bowel disease (IBD), irritable bowel syndrome and celiac disease. Here we show how pro-hormone Chromogranin A (CgA), produced by the enteroendocrine cells, and Catestatin (CST: hCgA352-372 ), the most abundant CgA-derived proteolytic peptide, affect the gut barrier. METHODS Colon tissues from region-specific CST-knockout (CST-KO) mice, CgA-knockout (CgA-KO) and WT mice were analysed by immunohistochemistry, western blot, ultrastructural and flowcytometry studies. FITC-dextran assays were used to measure intestinal barrier function. Mice were supplemented with CST or CgA fragment pancreastatin (PST: CgA250-301 ). The microbial composition of cecum was determined. CgA and CST levels were measured in blood of IBD patients. RESULTS Plasma levels of CST were elevated in IBD patients. CST-KO mice displayed (a) elongated tight, adherens junctions and desmosomes similar to IBD patients, (b) elevated expression of Claudin 2, and (c) gut inflammation. Plasma FITC-dextran measurements showed increased intestinal paracellular permeability in the CST-KO mice. This correlated with a higher ratio of Firmicutes to Bacteroidetes, a dysbiotic pattern commonly encountered in various diseases. Supplementation of CST-KO mice with recombinant CST restored paracellular permeability and reversed inflammation, whereas CgA-KO mice supplementation with CST and/or PST in CgA-KO mice showed that intestinal paracellular permeability is regulated by the antagonistic roles of these two peptides: CST reduces and PST increases permeability. CONCLUSION The pro-hormone CgA regulates the intestinal paracellular permeability. CST is both necessary and sufficient to reduce permeability and primarily acts by antagonizing PST.
Collapse
Affiliation(s)
- Elke M. Muntjewerff
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
| | - Kechun Tang
- VA San Diego Healthcare System San Diego CA USA
| | - Lisanne Lutter
- Center for Translational Immunology Utrecht University Medical Center Utrecht the Netherlands
- Department of Gastroenterology and Hepatology Utrecht University Medical Center Utrecht the Netherlands
| | - Gustaf Christoffersson
- Science for Life Laboratory Uppsala University Uppsala Sweden
- Department of Medical Cell biology Uppsala University Uppsala Sweden
| | - Mara J. T. Nicolasen
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
| | - Hong Gao
- Department of Medicine University of California San Diego La Jolla CA USA
| | - Gajanan D. Katkar
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla CA USA
| | - Soumita Das
- Department of Pathology University of California San Diego La Jolla CA USA
| | - Martin ter Beest
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
| | - Wei Ying
- Department of Medicine University of California San Diego La Jolla CA USA
| | - Pradipta Ghosh
- Department of Medicine University of California San Diego La Jolla CA USA
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla CA USA
| | - Sahar El Aidy
- Department of Molecular Immunology and Microbiology Groningen Biomolecular Sciences and Biotechnology Institute University of Groningen Groningen the Netherlands
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology Utrecht University Medical Center Utrecht the Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
- Department of Molecular Immunology and Microbiology Groningen Biomolecular Sciences and Biotechnology Institute University of Groningen Groningen the Netherlands
| | - Sushil K. Mahata
- VA San Diego Healthcare System San Diego CA USA
- Department of Medicine University of California San Diego La Jolla CA USA
| |
Collapse
|
21
|
Peripheral Corticotropin-Releasing Factor Triggers Jejunal Mast Cell Activation and Abdominal Pain in Patients With Diarrhea-Predominant Irritable Bowel Syndrome. Am J Gastroenterol 2020; 115:2047-2059. [PMID: 32740086 DOI: 10.14309/ajg.0000000000000789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION To determine the effect of peripheral CRF on intestinal barrier function in diarrhea-predominant IBS (IBS-D). Irritable bowel syndrome (IBS) pathophysiology has been linked to life stress, epithelial barrier dysfunction, and mast cell activation. Corticotropin-releasing factor (CRF) is a major mediator of stress responses in the gastrointestinal tract, yet its role on IBS mucosal function remains largely unknown. METHODS Intestinal response to sequential i.v. 5-mL saline solution (placebo) and CRF (100 μg) was evaluated in 21 IBS-D and 17 healthy subjects (HSs). A 20-cm jejunal segment was perfused with an isosmotic solution and effluents collected at baseline, 30 minutes after placebo, and 60 minutes after CRF. We measured water flux, albumin output, tryptase release, stress hormones, cardiovascular and psychological responses, and abdominal pain. A jejunal biopsy was obtained for CRF receptor expression assessment. RESULTS Water flux did not change after placebo in IBS-D and HS but significantly increased after CRF in IBS-D (P = 0.007). Basal luminal output of albumin was higher in IBS-D and increased further after CRF in IBS-D (P = 0.042). Basal jejunal tryptase release was higher in IBS-D, and CRF significantly increased it in both groups (P = 0.004), the response being higher in IBS-D than in HS (P = 0.0023). Abdominal pain worsened only in IBS-D after CRF and correlated with jejunal tryptase release, water flux, and albumin output. IBS-D displayed jejunal up-regulation of CRF2 and down-regulation of CRF1 compared with HS. DISCUSSION Stress via CRF-driven mast cell activation seems to be relevant in the pathophysiology of IBS-D.
Collapse
|
22
|
Takashima S, Tanaka F, Kawaguchi Y, Usui Y, Fujimoto K, Nadatani Y, Otani K, Hosomi S, Nagami Y, Kamata N, Taira K, Tanigawa T, Watanabe T, Imoto S, Uematsu S, Fujiwara Y. Proton pump inhibitors enhance intestinal permeability via dysbiosis of gut microbiota under stressed conditions in mice. Neurogastroenterol Motil 2020; 32:e13841. [PMID: 32319196 DOI: 10.1111/nmo.13841] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Intestinal permeability and psychological stress are considered the key mechanism(s) in functional dyspepsia (FD). Although proton pump inhibitors (PPIs) are commonly used for the treatment of FD, the effect of PPIs on intestinal permeability has not been elucidated. This study investigated the effect of PPI on intestinal permeability under stressed conditions. METHODS C57BL/6J mice were subjected to water avoidance stress (WAS) and administered rabeprazole (40 mg/kg) or vehicle treatment (VT). We then evaluated intestinal permeability both in vivo and ex vivo using plasma fluorescein isothiocyanate-dextran and by assessing the paracellular permeability and transepithelial electrical resistance (TEER) in an Ussing chamber, respectively. Furthermore, we evaluated the effect of PPI-treated fecal microbiota transplant (FMT) on intestinal permeability in vivo. Microbiota profiles of donor feces were assessed by 16S rRNA gene analysis using MiSeq and QIIME2. KEY RESULTS In the WAS treatment, PPI significantly enhanced intestinal permeability in vivo compared to that in VT. Moreover, PPI significantly increased paracellular permeability and decreased TEER in the duodenum and jejunum, respectively, compared to those in VT under stressed conditions. Moreover, both vasoactive intestinal peptide (VIP) receptor antagonist and ketotifen significantly reversed the effect of PPI on intestinal permeability. Furthermore, PPI-treated FMT significantly increased the intestinal permeability in vivo compared to that in vehicle-treated FMT. Proton pump inhibitors treatment altered the gut microbiota composition, indicating that PPI induced dysbiosis. CONCLUSIONS AND INFERENCES Under stressed conditions, PPI enhances intestinal permeability via dysbiosis of gut microbiota. Vasoactive intestinal peptide and mast cells are also implicated in the underlying mechanisms.
Collapse
Affiliation(s)
- Shingo Takashima
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Fumio Tanaka
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yunosuke Kawaguchi
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuki Usui
- Division of Systems Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kosuke Fujimoto
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuji Nadatani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Otani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shuhei Hosomi
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasuaki Nagami
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Noriko Kamata
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koichi Taira
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tetsuya Tanigawa
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Toshio Watanabe
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Seiya Imoto
- Division of Health Medical Data Science, Health Intelligence Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
23
|
Wauters L, Burns G, Ceulemans M, Walker MM, Vanuytsel T, Keely S, Talley NJ. Duodenal inflammation: an emerging target for functional dyspepsia? Expert Opin Ther Targets 2020; 24:511-523. [PMID: 32249629 DOI: 10.1080/14728222.2020.1752181] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Functional dyspepsia (FD) is one of the most common functional gastrointestinal disorders and is classified into postprandial distress and epigastric pain syndrome. Despite the recognition of duodenal inflammation as a potential trigger of symptoms, only limited anti-inflammatory therapies exist.Areas covered: This narrative review summarizes the recent advances in the pathophysiology and treatment of FD; it identifies potential therapeutic targets and gaps in the field. An electronic literature search was conducted in Pubmed up to 31st of December 2019.Expert opinion: There is compelling evidence for the role of duodenal inflammation and the eosinophil-mast cell axis in the pathogenesis of dyspeptic symptoms. Traditional prokinetic drugs and neuromodulators target gastric dysmotility and visceral hypersensitivity but are hampered by limited efficacy and side effects. Independent of acid suppression, the anti-inflammatory action of proton pump inhibitors, which remain the first-line therapy in FD, may also explain their therapeutic effect. Other existing and newly established anti-inflammatory drugs should be investigated while trials including probiotics and selective antibiotics should examine the host microbiome and immune activation. Targeted treatments for potential causes of duodenal pathology, such as impaired permeability and dysbiosis, are likely to emerge in the future.
Collapse
Affiliation(s)
- Lucas Wauters
- Translational Research in Gastrointestinal Diseases (TARGID), KU Leuven, Leuven, Belgium.,Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Grace Burns
- Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia
| | - Matthias Ceulemans
- Translational Research in Gastrointestinal Diseases (TARGID), KU Leuven, Leuven, Belgium
| | - Marjorie M Walker
- Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia
| | - Tim Vanuytsel
- Translational Research in Gastrointestinal Diseases (TARGID), KU Leuven, Leuven, Belgium.,Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Simon Keely
- Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia.,Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute, Callaghan, Australia
| | - Nicholas J Talley
- Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia
| |
Collapse
|
24
|
Casado-Bedmar M, Keita ÅV. Potential neuro-immune therapeutic targets in irritable bowel syndrome. Therap Adv Gastroenterol 2020; 13:1756284820910630. [PMID: 32313554 PMCID: PMC7153177 DOI: 10.1177/1756284820910630] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/11/2020] [Indexed: 02/04/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal (GI) disorder characterized by recurring abdominal pain and disturbed bowel habits. The aetiology of IBS is unknown but there is evidence that genetic, environmental and immunological factors together contribute to the development of the disease. Current treatment of IBS includes lifestyle and dietary interventions, laxatives or antimotility drugs, probiotics, antispasmodics and antidepressant medication. The gut-brain axis comprises the central nervous system, the hypothalamic pituitary axis, the autonomic nervous system and the enteric nervous system. Within the intestinal mucosa there are close connections between immune cells and nerve fibres of the enteric nervous system, and signalling between, for example, mast cells and nerves has shown to be of great importance during GI disorders such as IBS. Communication between the gut and the brain is most importantly routed via the vagus nerve, where signals are transmitted by neuropeptides. It is evident that IBS is a disease of a gut-brain axis dysregulation, involving altered signalling between immune cells and neurotransmitters. In this review, we analyse the most novel and distinct neuro-immune interactions within the IBS mucosa in association with already existing and potential therapeutic targets.
Collapse
Affiliation(s)
- Maite Casado-Bedmar
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Åsa V. Keita
- Department of Biomedical and Clinical Sciences, Medical Faculty, Linköping University, Campus US, Linköping, 581 85, Sweden
| |
Collapse
|
25
|
Keita ÅV, Alkaissi LY, Holm EB, Heil SDS, Chassaing B, Darfeuille-Michaud A, McKay DM, Söderholm JD. Enhanced E. coli LF82 Translocation through the Follicle-associated Epithelium in Crohn's Disease is Dependent on Long Polar Fimbriae and CEACAM6 expression, and Increases Paracellular Permeability. J Crohns Colitis 2020; 14:216-229. [PMID: 31393983 PMCID: PMC7008151 DOI: 10.1093/ecco-jcc/jjz144] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Patients with Crohn's disease [CD] harbour an increased number of adherent-invasive E. coli [AIEC]. The strain LF82, identified in the ileal mucosa of CD patients, has been extensively studied for pathogenic mechanisms. However, understanding of the interaction of LF82 with the intestinal mucosa of CD patients is lacking. METHODS Here, we investigated the importance of long polar fimbriae [LPF] type 1 pili and the carcinoembryonic antigen-related cell-adhesion molecule 6 [CEACAM6] for translocation of LF82 in an in vitro model of follicle-associated epithelium [FAE], and in the FAE and villus epithelium [VE] of patients with CD and controls, using Ussing chambers. RESULTS Significantly greater LF82 passage occurred in the FAE model compared with in the VE Caco-2cl1 mono-culture. Moreover, bacterial translocation was inhibited by either LPF disruption or pre-incubation with anti-CEACAM6 antibody. Tissue mounted in Ussing chambers showed significantly higher LF82 passage in FAE from patients with CD compared with control FAE, that was diminished in LF82 lacking LPF and by blocking host CEACAM6. Interestingly, addition of LF82 to the CD FAE tissues significantly increased paracellular permeability [of 51Chromium-EDTA] compared with baseline, and the increase was inhibited by anti-CEACAM6. Immunofluorescence and immunoblots showed higher expression of CEACAM6 in FAE of patients with CD compared with in FAE from controls. CONCLUSIONS These data suggest that the FAE of CD patients is a site of vulnerability for invasion by LF82 via a mechanism that requires both bacterial LPF and host CEACAM6. Further, LF82 has the ability to increase paracellular passage through the FAE of patients with CD. These data can help define novel therapeutic targets in CD for the prevention of clinical recurrence.
Collapse
Affiliation(s)
- Åsa V Keita
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Medical Faculty, Linköping University, Linköping, Sweden
| | - Lina Yakymenko Alkaissi
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Medical Faculty, Linköping University, Linköping, Sweden
| | - Elin B Holm
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Medical Faculty, Linköping University, Linköping, Sweden
| | - Stéphanie D S Heil
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Medical Faculty, Linköping University, Linköping, Sweden
| | - Benoit Chassaing
- Neuroscience Institute and Institute for Biomedical Sciences, Georgia State University, Atlanta, USA
| | | | - Derek M McKay
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Johan D Söderholm
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Medical Faculty, Linköping University, Linköping, Sweden
- Department of Surgery, County Council of Östergötland, Linköping, Sweden
| |
Collapse
|
26
|
Schwerdtfeger LA, Tobet SA. Vasoactive intestinal peptide regulates ileal goblet cell production in mice. Physiol Rep 2020; 8:e14363. [PMID: 32026594 PMCID: PMC7002535 DOI: 10.14814/phy2.14363] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/14/2022] Open
Abstract
Innervation of the intestinal mucosa has gained more attention with demonstrations of tuft and enteroendocrine cell innervation. However, the role(s) these fibers play in maintaining the epithelial and mucus barriers are still poorly understood. This study therefore examines the proximity of mouse ileal goblet cells to neuronal fibers, and the regulation of goblet cell production by vasoactive intestinal peptide (VIP). An organotypic intestinal slice model that maintains the cellular diversity of the intestinal wall ex vivo was used. An ex vivo copper-free click-reaction to label glycosaminoglycans was used to identify goblet cells. Pharmacological treatment of slices was used to assess the influence of VIP receptor antagonism on goblet cell production and neuronal fiber proximity. Goblet cells were counted and shown to have at least one peripherin immunoreactive fiber within 3 µm of the cell, 51% of the time. Treatment with a VIP receptor type I and II antagonist (VPACa) resulted in an increase in the percentage of goblet cells with peripherin fibers. Pharmacological treatments altered goblet cell counts in intestinal crypts and villi, with tetrodotoxin and VPACa substantially decreasing goblet cell counts. When cultured with 5-Ethynyl-2'-deoxyuridine (EdU) as an indicator of cell proliferation, colocalization of labeled goblet cells and EdU in ileal crypts was decreased by 77% when treated with VPACa. This study demonstrates a close relationship of intestinal goblet cells to neuronal fibers. By using organotypic slices from mouse ileum, vasoactive intestinal peptide receptor regulation of gut wall goblet cell production was revealed.
Collapse
Affiliation(s)
| | - Stuart A. Tobet
- Department of Biomedical SciencesColorado State UniversityFort CollinsCOUSA
- School of Biomedical EngineeringColorado State UniversityFort CollinsCOUSA
| |
Collapse
|
27
|
Accarie A, Vanuytsel T. Animal Models for Functional Gastrointestinal Disorders. Front Psychiatry 2020; 11:509681. [PMID: 33262709 PMCID: PMC7685985 DOI: 10.3389/fpsyt.2020.509681] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Functional gastrointestinal disorders (FGID), such as functional dyspepsia (FD) and irritable bowel syndrome (IBS) are characterized by chronic abdominal symptoms in the absence of an organic, metabolic or systemic cause that readily explains these complaints. Their pathophysiology is still not fully elucidated and animal models have been of great value to improve the understanding of the complex biological mechanisms. Over the last decades, many animal models have been developed to further unravel FGID pathophysiology and test drug efficacy. In the first part of this review, we focus on stress-related models, starting with the different perinatal stress models, including the stress of the dam, followed by a discussion on neonatal stress such as the maternal separation model. We also describe the most commonly used stress models in adult animals which brought valuable insights on the brain-gut axis in stress-related disorders. In the second part, we focus more on models studying peripheral, i.e., gastrointestinal, mechanisms, either induced by an infection or another inflammatory trigger. In this section, we also introduce more recent models developed around food-related metabolic disorders or food hypersensitivity and allergy. Finally, we introduce models mimicking FGID as a secondary effect of medical interventions and spontaneous models sharing characteristics of GI and anxiety-related disorders. The latter are powerful models for brain-gut axis dysfunction and bring new insights about FGID and their comorbidities such as anxiety and depression.
Collapse
Affiliation(s)
- Alison Accarie
- Department of Chronic Diseases, Metabolism and Ageing (ChroMetA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Department of Chronic Diseases, Metabolism and Ageing (ChroMetA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.,Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Magrone T, Magrone M, Jirillo E. Mast Cells as a Double-Edged Sword in Immunity: Their Function in Health and Disease. First of Two Parts. Endocr Metab Immune Disord Drug Targets 2019; 20:654-669. [PMID: 31789135 DOI: 10.2174/1871530319666191202120301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/08/2019] [Accepted: 11/21/2019] [Indexed: 11/22/2022]
Abstract
Mast cells (MCs) have recently been re-interpreted in the context of the immune scenario in the sense that their pro-allergic role is no longer exclusive. In fact, MCs even in steady state conditions maintain homeostatic functions, producing mediators and intensively cross-talking with other immune cells. Here, emphasis will be placed on the array of receptors expressed by MCs and the variety of cytokines they produce. Then, the bulk of data discussed will provide readers with a wealth of information on the dual ability of MCs not only to defend but also to offend the host. This double attitude of MCs relies on many variables, such as their subsets, tissues of residency and type of stimuli ranging from microbes to allergens and food antigens. Finally, the relationship between MCs with basophils and eosinophils will be discussed.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Manrico Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
29
|
Iwasaki M, Akiba Y, Kaunitz JD. Recent advances in vasoactive intestinal peptide physiology and pathophysiology: focus on the gastrointestinal system. F1000Res 2019; 8. [PMID: 31559013 PMCID: PMC6743256 DOI: 10.12688/f1000research.18039.1] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
Vasoactive intestinal peptide (VIP), a gut peptide hormone originally reported as a vasodilator in 1970, has multiple physiological and pathological effects on development, growth, and the control of neuronal, epithelial, and endocrine cell functions that in turn regulate ion secretion, nutrient absorption, gut motility, glycemic control, carcinogenesis, immune responses, and circadian rhythms. Genetic ablation of this peptide and its receptors in mice also provides new insights into the contribution of VIP towards physiological signaling and the pathogenesis of related diseases. Here, we discuss the impact of VIP on gastrointestinal function and diseases based on recent findings, also providing insight into its possible therapeutic application to diabetes, autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Mari Iwasaki
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Yasutada Akiba
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jonathan D Kaunitz
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA.,Departments of Medicine and Surgery, UCLA School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
30
|
Casado-Bedmar M, Heil SDS, Myrelid P, Söderholm JD, Keita ÅV. Upregulation of intestinal mucosal mast cells expressing VPAC1 in close proximity to vasoactive intestinal polypeptide in inflammatory bowel disease and murine colitis. Neurogastroenterol Motil 2019; 31:e13503. [PMID: 30407703 DOI: 10.1111/nmo.13503] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/24/2018] [Accepted: 10/07/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Mast cells (MCs) and vasoactive intestinal polypeptide (VIP) have been proposed as regulators of the intestinal barrier and inflammation. Our aim was to map the distribution in inflammatory bowel disease (IBD) and murine colitis. METHODS MCs, VIP, and VIP-receptors (VPACs) were quantified by immunofluorescence and enzyme-immunoassay (EIA) in ileal tissues (villus epithelium (VE) and adjacent VE, ie, VE next to the follicle-associated epithelium, (FAE)) from Crohn's disease (CD; n = 16) and non-IBD patients, and in colonic specimens of ulcerative colitis (UC; n = 12) and healthy controls (HCs). In addition, VIP levels were measured in plasma from HCs, non-IBD, and IBD in remission (CD n = 30; UC n = 30). Colon, ileum, and plasma from mice with dextran sulfate sodium (DSS)-induced colitis and control mice were analyzed likewise. KEY RESULTS FAE-adjacent VE in ileum of CD possessed more MCs (P < 0.05) and MCs expressing VPAC1 (P < 0.05), but not VPAC2, compared to controls. Both adjacent and regular VE of CD had more MCs co-localizing/in close proximity to VIP (P < 0.05). In UC colon, more MCs (P < 0.0005), MCs close to VIP (P < 0.0005), and MCs expressing VPAC1 (P < 0.05) were found compared to controls. VIP levels were elevated in plasma from CD and UC compared to controls (P < 0.0005). Colon of DSS mice showed more MCs and MCs close to VIP (P < 0.05) compared to control mice. In vitro experiments revealed MCs expressing VPACs and internalized VIP after 120 minutes of VIP-stimulation. CONCLUSIONS AND INFERENCES Communication between MCs and VIP is upregulated during IBD and mice colitis. In CD patients, the epithelium next to FAE seems to be more involved than the surrounding VE, suggesting increased MC-VIP-interactions in this intestinal region.
Collapse
Affiliation(s)
- Maite Casado-Bedmar
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Linköping University, Linköping, Sweden
| | - Stéphanie D S Heil
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Linköping University, Linköping, Sweden
| | - Pär Myrelid
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Linköping University, Linköping, Sweden
- Department of Surgery, County Council of Östergötland, Linköping, Sweden
| | - Johan D Söderholm
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Linköping University, Linköping, Sweden
- Department of Surgery, County Council of Östergötland, Linköping, Sweden
| | - Åsa V Keita
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Linköping University, Linköping, Sweden
| |
Collapse
|
31
|
Schoultz I, Keita ÅV. Cellular and Molecular Therapeutic Targets in Inflammatory Bowel Disease-Focusing on Intestinal Barrier Function. Cells 2019; 8:193. [PMID: 30813280 PMCID: PMC6407030 DOI: 10.3390/cells8020193] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023] Open
Abstract
The human gut relies on several cellular and molecular mechanisms to allow for an intact and dynamical intestinal barrier. Normally, only small amounts of luminal content pass the mucosa, however, if the control is broken it can lead to enhanced passage, which might damage the mucosa, leading to pathological conditions, such as inflammatory bowel disease (IBD). It is well established that genetic, environmental, and immunological factors all contribute in the pathogenesis of IBD, and a disturbed intestinal barrier function has become a hallmark of the disease. Genetical studies support the involvement of intestinal barrier as several susceptibility genes for IBD encode proteins with key functions in gut barrier and homeostasis. IBD patients are associated with loss in bacterial diversity and shifts in the microbiota, with a possible link to local inflammation. Furthermore, alterations of immune cells and several neuro-immune signaling pathways in the lamina propria have been demonstrated. An inappropriate immune activation might lead to mucosal inflammation, with elevated secretion of pro-inflammatory cytokines that can affect the epithelium and promote a leakier barrier. This review will focus on the main cells and molecular mechanisms in IBD and how these can be targeted in order to improve intestinal barrier function and reduce inflammation.
Collapse
Affiliation(s)
- Ida Schoultz
- School of Medical Sciences, Örebro University, 703 62 Örebro, Sweden.
| | - Åsa V Keita
- Department of Clinical and Experimental Medicine, Division of Surgery, Orthopedics & Oncology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden.
| |
Collapse
|
32
|
Sun Y, Li L, Xie R, Wang B, Jiang K, Cao H. Stress Triggers Flare of Inflammatory Bowel Disease in Children and Adults. Front Pediatr 2019; 7:432. [PMID: 31709203 PMCID: PMC6821654 DOI: 10.3389/fped.2019.00432] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 10/07/2019] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an idiopathic inflammatory disease characterized by chronic and relapsing manifestations. It is noteworthy that the prevalence of IBD is gradually increasing in both children and adults. Currently, the pathogenesis of IBD remains to be completely elucidated. IBD is believed to occur through interactions among genetics, environmental factors, and the gut microbiota. However, the relapsing and remitting course of IBD underlines the importance of other modifiers, such as psychological stress. Growing evidence from clinical and experimental studies suggests that stress acts as a promoting or relapsing factor for IBD. Importantly, recent studies have reported an increasing incidence of anxiety or depression in both children and adults with IBD. In this article, we review the mechanisms by which stress affects IBD, such as via impaired intestinal barrier function, disturbance of the gut microbiota, intestinal dysmotility, and immune and neuroendocrine dysfunction. With regard to both children and adults, we provide recent evidence to describe how stress can affect IBD at various stages. Furthermore, we emphasize the importance of mental healing and discuss the value of approaches targeting stress in clinical management to develop enhanced strategies for the prevention and treatment of IBD.
Collapse
Affiliation(s)
- Yue Sun
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Lu Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Runxiang Xie
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
33
|
Mucosal permeability and mast cells as targets for functional gastrointestinal disorders. Curr Opin Pharmacol 2018; 43:66-71. [PMID: 30216901 DOI: 10.1016/j.coph.2018.08.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/22/2018] [Accepted: 08/25/2018] [Indexed: 02/08/2023]
Abstract
The intestinal mucosa is constantly exposed to harmful luminal content, and uptake is closely controlled and regulated by neuro-immune factors. If control is broken, it might lead to ongoing enhanced mucosal permeability, potentially resulting in functional gastrointestinal disorders. The importance of mast cells in the regulation of the mucosal barrier has become obvious, and increased numbers and more activated mast cells have been observed in irritable bowel syndrome, functional dyspepsia and gastroesophageal reflux disease. To target the disturbed mucosal permeability, directly or via mast cells, is therefore currently of major interest. For example, administration of mast cell stabilizers and probiotics have shown promising effects in patients with functional gastrointestinal disorders.
Collapse
|
34
|
Creekmore AL, Hong S, Zhu S, Xue J, Wiley JW. Chronic stress-associated visceral hyperalgesia correlates with severity of intestinal barrier dysfunction. Pain 2018; 159:1777-1789. [PMID: 29912860 PMCID: PMC6097612 DOI: 10.1097/j.pain.0000000000001271] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In humans, chronic psychological stress is associated with increased intestinal paracellular permeability and visceral hyperalgesia, which is recapitulated in the chronic intermittent water avoidance stress (WAS) rat model. However, it is unknown whether enhanced visceral pain and permeability are intrinsically linked and correlate. Treatment of rats with lubiprostone during WAS significantly reduced WAS-induced changes in intestinal epithelial paracellular permeability and visceral hyperalgesia in a subpopulation of rats. Lubiprostone also prevented WAS-induced decreases in the epithelial tight junction protein, occludin (Ocln). To address the question of whether the magnitude of visceral pain correlates with the extent of altered intestinal permeability, we measured both end points in the same animal because of well-described individual differences in pain response. Our studies demonstrate that visceral pain and increased colon permeability positively correlate (0.6008, P = 0.0084). Finally, exposure of the distal colon in control animals to Ocln siRNA in vivo revealed that knockdown of Ocln protein inversely correlated with increased paracellular permeability and enhanced visceral pain similar to the levels observed in WAS-responsive rats. These data support that Ocln plays a potentially significant role in the development of stress-induced increased colon permeability. We believe this is the first demonstration that the level of chronic stress-associated visceral hyperalgesia directly correlates with the magnitude of altered colon epithelial paracellular permeability.
Collapse
Affiliation(s)
| | | | | | | | - John W. Wiley
- Corresponding Author: John W Wiley, MD, University of Michigan Medical School, 1150 W Medical Center Drive, 9301A MSRB III, Ann Arbor MI 48109-5648, 734-615-6621,
| |
Collapse
|
35
|
Yakabi S, Wang L, Karasawa H, Yuan PQ, Koike K, Yakabi K, Taché Y. VIP is involved in peripheral CRF-induced stimulation of propulsive colonic motor function and diarrhea in male rats. Am J Physiol Gastrointest Liver Physiol 2018; 314:G610-G622. [PMID: 29420068 PMCID: PMC6008061 DOI: 10.1152/ajpgi.00308.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/31/2018] [Accepted: 02/06/2018] [Indexed: 01/31/2023]
Abstract
We investigated whether vasoactive intestinal peptide (VIP) and/or prostaglandins contribute to peripheral corticotropin-releasing factor (CRF)-induced CRF1 receptor-mediated stimulation of colonic motor function and diarrhea in rats. The VIP antagonist, [4Cl-D-Phe6, Leu17]VIP injected intraperitoneally completely prevented CRF (10 µg/kg ip)-induced fecal output and diarrhea occurring within the first hour after injection, whereas pretreatment with the prostaglandins synthesis inhibitor, indomethacin, had no effect. In submucosal plexus neurons, CRF induced significant c-Fos expression most prominently in the terminal ileum compared with duodenum and jejunum, whereas no c-Fos was observed in the proximal colon. c-Fos expression in ileal submucosa was colocalized in 93.4% of VIP-positive neurons and 31.1% of non-VIP-labeled neurons. CRF1 receptor immunoreactivity was found on the VIP neurons. In myenteric neurons, CRF induced only a few c-Fos-positive neurons in the ileum and a robust expression in the proximal colon (17.5 ± 2.4 vs. 0.4 ± 0.3 cells/ganglion in vehicle). The VIP antagonist prevented intraperitoneal CRF-induced c-Fos induction in the ileal submucosal plexus and proximal colon myenteric plexus. At 60 min after injection, CRF decreased VIP levels in the terminal ileum compared with saline (0.8 ± 0.3 vs. 2.5 ± 0.7 ng/g), whereas VIP mRNA level detected by qPCR was not changed. These data indicate that intraperitoneal CRF activates intestinal submucosal VIP neurons most prominently in the ileum and myenteric neurons in the colon. It also implicates VIP signaling as part of underlying mechanisms driving the acute colonic secretomotor response to a peripheral injection of CRF, whereas prostaglandins do not play a role. NEW & NOTEWORTHY Corticotropin-releasing factor (CRF) in the gut plays a physiological role in the stimulation of lower gut secretomotor function induced by stress. We showed that vasoactive intestinal peptide (VIP)-immunoreactive neurons in the ileal submucosal plexus expressed CRF1 receptor and were prominently activated by CRF, unlike colonic submucosal neurons. VIP antagonist abrogated CRF-induced ileal submucosal and colonic myenteric activation along with functional responses (defecation and diarrhea). These data point to VIP signaling in ileum and colon as downstream effectors of CRF.
Collapse
Affiliation(s)
- Seiichi Yakabi
- CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California, Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo , Tokyo , Japan
| | - Lixin Wang
- CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California, Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Hiroshi Karasawa
- CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California, Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Pu-Qing Yuan
- CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California, Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo , Tokyo , Japan
| | - Koji Yakabi
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University , Saitama , Japan
| | - Yvette Taché
- CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California, Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| |
Collapse
|
36
|
Redegeld FA, Yu Y, Kumari S, Charles N, Blank U. Non-IgE mediated mast cell activation. Immunol Rev 2018; 282:87-113. [DOI: 10.1111/imr.12629] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Frank A. Redegeld
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Yingxin Yu
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Sangeeta Kumari
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Nicolas Charles
- INSERM U1149; Centre de Recherche sur l'Inflammation; Paris France
- CNRS ERL8252; Paris France
- Université Paris-Diderot; Sorbonne Paris Cité; Faculté de Médecine; Site Xavier Bichat; Paris France
| | - Ulrich Blank
- INSERM U1149; Centre de Recherche sur l'Inflammation; Paris France
- CNRS ERL8252; Paris France
- Université Paris-Diderot; Sorbonne Paris Cité; Faculté de Médecine; Site Xavier Bichat; Paris France
- Inflamex Laboratory of Excellence; Paris France
| |
Collapse
|
37
|
Ganda Mall JP, Casado-Bedmar M, Winberg ME, Brummer RJ, Schoultz I, Keita ÅV. A β-Glucan-Based Dietary Fiber Reduces Mast Cell-Induced Hyperpermeability in Ileum From Patients With Crohn's Disease and Control Subjects. Inflamm Bowel Dis 2017; 24:166-178. [PMID: 29272475 PMCID: PMC6166688 DOI: 10.1093/ibd/izx002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Administration of β-glucan has shown immune-enhancing effects. Our aim was to investigate whether β-glucan could attenuate mast cell (MC)-induced hyperpermeability in follicle-associated epithelium (FAE) and villus epithelium (VE) of patients with Crohn's disease (CD) and in noninflammatory bowel disease (IBD)-controls. Further, we studied mechanisms of β-glucan uptake and effects on MCs in vitro. METHODS Segments of FAE and VE from 8 CD patients and 9 controls were mounted in Ussing chambers. Effects of the MC-degranulator compound 48/80 (C48/80) and yeast-derived β-1,3/1,6 glucan on hyperpermeability were investigated. Translocation of β-glucan and colocalization with immune cells were studied by immunofluorescence. Caco-2-cl1- and FAE-cultures were used to investigate β-glucan-uptake using endocytosis inhibitors and HMC-1.1 to study effects on MCs. RESULTS β-glucan significantly attenuated MC-induced paracellular hyperpermeability in CD and controls. Transcellular hyperpermeability was only significantly attenuated in VE. Baseline paracellular permeability was higher in FAE than VE in both groups, P<0.05, and exhibited a more pronounced effect by C48/80 and β-glucan P<0.05. No difference was observed between CD and controls. In vitro studies showed increased passage, P<0.05, of β-glucan through FAE-culture compared to Caco-2-cl1. Passage was mildly attenuated by the inhibitor methyl-β-cyclodextrin. HMC-1.1 experiments showed a trend to decreasing MC-degranulation and levels of TNF-α but not IL-6 by β-glucan. Immunofluorescence revealed more β-glucan-uptake and higher percentage of macrophages and dendritic cells close to β-glucan in VE of CD compared to controls. CONCLUSIONS We demonstrated beneficial effects of β-glucan on intestinal barrier function and increased β-glucan-passage through FAE model. Our results provide important and novel knowledge on possible applications of β-glucan in health disorders and diseases characterized by intestinal barrier dysfunction.
Collapse
Affiliation(s)
- John-Peter Ganda Mall
- School of Medical Sciences, Nutrition-Gut-Brain Interactions Research Centre, Örebro University, Örebro, Sweden
| | - Maite Casado-Bedmar
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Martin E Winberg
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Robert J Brummer
- School of Medical Sciences, Nutrition-Gut-Brain Interactions Research Centre, Örebro University, Örebro, Sweden
| | - Ida Schoultz
- School of Medical Sciences, Nutrition-Gut-Brain Interactions Research Centre, Örebro University, Örebro, Sweden
| | - Åsa V Keita
- School of Medical Sciences, Nutrition-Gut-Brain Interactions Research Centre, Örebro University, Örebro, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
38
|
Guan XF, Duan ZJ. Protective effects of brain-gut peptides against intestinal barrier injury and mechanisms involved. Shijie Huaren Xiaohua Zazhi 2017; 25:2805-2812. [DOI: 10.11569/wcjd.v25.i31.2805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Brain-gut peptides, a group of small molecule polypeptides, have been found to distribute widely in the brain and the gastrointestinal system and act as both neurotransmitters and hormones. Intestinal barrier injury has a serious impact on the prognosis of critical diseases. Brain-gut peptides can modulate tight junction proteins, promote epithelial cell proliferation, and inhibit apoptosis and inflammatory cytokines, thus playing an important role in the maintenance of intestinal barrier and mucosal immunity. In this review, we discuss the protective effects of brain-gut peptides against intestinal barrier injury and the underlying mechanisms.
Collapse
Affiliation(s)
- Xing-Fang Guan
- Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| | - Zhi-Jun Duan
- Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| |
Collapse
|
39
|
Bednarska O, Walter SA, Casado-Bedmar M, Ström M, Salvo-Romero E, Vicario M, Mayer EA, Keita ÅV. Vasoactive Intestinal Polypeptide and Mast Cells Regulate Increased Passage of Colonic Bacteria in Patients With Irritable Bowel Syndrome. Gastroenterology 2017; 153:948-960.e3. [PMID: 28711627 PMCID: PMC5623149 DOI: 10.1053/j.gastro.2017.06.051] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 06/13/2017] [Accepted: 06/30/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Irritable bowel syndrome (IBS) is associated with intestinal dysbiosis and symptoms of IBS develop following gastroenteritis. We aimed to study the passage of live bacteria through the colonic epithelium, and determine the role of mast cells (MCs) and vasoactive intestinal polypeptide (VIP) in barrier regulation in IBS and healthy individuals. METHODS Colon biopsies from 32 women with IBS and 15 age-matched healthy women (controls) were mounted in Ussing chambers; we measured numbers of fluorescently labeled Escherichia coli HS and Salmonella typhimurium that passed through from the mucosal side to the serosal side of the tissue. Some biopsies were exposed to agents that block the VIP receptors (VPAC1 and VPAC2) or MCs. Levels of VIP and tryptase were measured in plasma and biopsy lysates. Number of MCs and MCs that express VIP or VIP receptors were quantified by immunofluorescence. Biopsies from an additional 5 patients with IBS and 4 controls were mounted in chambers and Salmonella were added; we studied passage routes through the epithelium by transmission electron microscopy and expression of tight junctions by confocal microscopy. RESULTS In colon biopsies from patients with IBS, larger numbers of E coli HS and S typhimurium passed through the epithelium than in biopsies from controls (P < .0005). In transmission electron microscopy analyses, bacteria were found to cross the epithelium via only the transcellular route. Bacterial passage was reduced in biopsies from patients with IBS and controls after addition of antibodies against VPACs or ketotifen, which inhibits MCs. Plasma samples from patients with IBS had higher levels of VIP than plasma samples from controls. Biopsies from patients with IBS had higher levels of tryptase, larger numbers of MCs, and a higher percentage of MCs that express VPAC1 than biopsies from controls. In biopsies from patients with IBS, addition of Salmonella significantly reduced levels of occludin; subsequent addition of ketotifen significantly reversed this effect. CONCLUSIONS We found that colonic epithelium tissues from patients with IBS have increased translocation of commensal and pathogenic live bacteria compared with controls. The mechanisms of increased translocation include MCs and VIP.
Collapse
Affiliation(s)
- Olga Bednarska
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Department of Gastroenterology, Linköping University, Linköping, Sweden
| | - Susanna A Walter
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Department of Gastroenterology, Linköping University, Linköping, Sweden
| | - Maite Casado-Bedmar
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Magnus Ström
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Department of Gastroenterology, Linköping University, Linköping, Sweden
| | - Eloísa Salvo-Romero
- Laboratory of Translational Mucosal Immunology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain
| | - Maria Vicario
- Laboratory of Translational Mucosal Immunology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain
| | - Emeran A Mayer
- G Oppenheimer Center for Neurobiology of Stress & Resilience, Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Åsa V Keita
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
40
|
Verma AK, Manohar M, Upparahalli Venkateshaiah S, Mishra A. Neuroendocrine cells derived chemokine vasoactive intestinal polypeptide (VIP) in allergic diseases. Cytokine Growth Factor Rev 2017; 38:37-48. [PMID: 28964637 DOI: 10.1016/j.cytogfr.2017.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 12/20/2022]
Abstract
Worldwide increase incidences of allergic diseases have heightened the interest of clinicians and researchers to understand the role of neuroendocrine cells in the recruitment and activation of inflammatory cells. Several pieces of evidence revealed the association of neuropeptides in the pathogenesis of allergic diseases. Importantly, one such peptide that is secreted by neuronal cells and immune cells exerts a wide spectrum of immunological functions as cytokine/chemokine is termed as Vasoactive Intestinal Peptide (VIP). VIP mediates immunological function through interaction with specific receptors namely VPAC-1, VPAC-2, CRTH2 and PAC1 that are expressed on several immune cells such as eosinophils, mast cells, neutrophils, and lymphocytes; therefore, provide the basis for the action of VIP on the immune system. Additionally, VIP mediated action varies according to target organ depending upon the presence of specific VIP associated receptor, involved immune cells and the microenvironment of the organ. Herein, we present an integrative review of the current understanding on the role of VIP and associated receptors in allergic diseases, the presence of VIP receptors on various immune cells with particular emphasis on the role of VIP in the pathogenesis of allergic diseases such as asthma, allergic rhinitis, and atopic dermatitis. Being crucial signal molecule of the neuroendocrine-immune network, the development of stable VIP analogue and/or antagonist may provide the future therapeutic drug alternative for the better treatment of these allergic diseases. Taken together, our current review summarizes the current understandings of VIP biology and further explore the significance of neuroendocrine cells derived VIP in the recruitment of inflammatory cells in allergic diseases that may be helpful to the investigators for planning the experiments and accordingly predicting new therapeutic strategies for combating allergic diseases. Summarized graphical abstract will help the readers to understand the significance of VIP in allergic diseases.
Collapse
Affiliation(s)
- Alok K Verma
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Murli Manohar
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sathisha Upparahalli Venkateshaiah
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Anil Mishra
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorders Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
41
|
Zhang L, Song J, Bai T, Qian W, Hou XH. Stress induces more serious barrier dysfunction in follicle-associated epithelium than villus epithelium involving mast cells and protease-activated receptor-2. Sci Rep 2017; 7:4950. [PMID: 28694438 PMCID: PMC5503989 DOI: 10.1038/s41598-017-05064-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/23/2017] [Indexed: 02/08/2023] Open
Abstract
Psychological stress has been associated with intestinal epithelial hyperpermeability, the basic process in various functional and organic bowel diseases. In the present study, we aimed to clarify the differences and underlining mechanisms in stress-induced barrier disruption in functionally and structurally distinct epitheliums, including the villus epithelium (VE) and follicle-associated epithelium (FAE), a specialized epithelium overlaid the domes of Peyer's lymphoid follicles. Employing an Ussing Chamber system, the epithelial permeability was assessed in rats following water avoidance stress (WAS) in vivo and in mucosa tissues exposed to corticotropin-releasing factor (CRF) ex vivo. Decreased transepithelial resistance (TER) and increased paracellular and transcellular macromolecular permeability in colon, ileal VE and FAE had been observed in WAS rats and in CRF-exposed mucosa. Especially, the barrier dysfunction was more serious in the FAE. Moreover, WAS upregulated the expression of mast cell tryptase and protease-activated receptor-2 (PAR2), which positively correlated with epithelial conductance. Mast cell stabilizer cromolyn sodium obviously alleviated the barrier disruption induced by WAS in vivo and CRF in vitro. Serine protease inhibitor aprotinin and FUT-175, and selective PAR2 antagonist ENMD-1068 effectively inhibited the CRF-induced FAE hyperpermeability. Altogether, it concluded that the FAE was more susceptible to stress, and the mast cells and PAR2 signaling played crucial roles in this process.
Collapse
Affiliation(s)
- Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jun Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Hua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
42
|
Abstract
The acute phase of IBD with inflamed gut and often ulcerated mucosa is clearly different from the apparently normal mucosa characteristic of IBS. However, more detailed assessment has detected immune activation, increased gut permeability, increased mucosal serotonin availability, abnormalities of enteric nerve structure and function, and dysbiosis in gut microbiota in IBS - all features seen in IBD. Furthermore, as treatments for inflammation in IBD have become more effective it is now apparent that ∼1 in 3 patients with IBD in remission from inflammation still have persistent abnormalities of sensation, motility and gut microbiota, which might cause IBS-like symptoms. This Perspective explores the overlap between IBS and IBD and their treatments, proposing future directions for research in this stimulating area.
Collapse
Affiliation(s)
- Robin Spiller
- NIHR Biomedical Research Unit in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust and The University of Nottingham, Queens Medical Centre, E Floor West Block, Nottingham NG7 2UH, UK
| | - Giles Major
- NIHR Biomedical Research Unit in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust and The University of Nottingham, Queens Medical Centre, E Floor West Block, Nottingham NG7 2UH, UK
| |
Collapse
|
43
|
Zhang L, Song J, Hou X. Mast Cells and Irritable Bowel Syndrome: From the Bench to the Bedside. J Neurogastroenterol Motil 2016; 22:181-192. [PMID: 26755686 PMCID: PMC4819856 DOI: 10.5056/jnm15137] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/07/2015] [Accepted: 12/26/2015] [Indexed: 12/11/2022] Open
Abstract
Irritable bowel syndrome (IBS) is traditionally defined as a functional disorder since it lacks demonstrable pathological abnormalities. However, in recent years, low grade inflammatory infiltration, often rich in mast cells, in both the small and large bowel, has been observed in some patients with IBS. The close association of mast cells with major intestinal functions, such as epithelial secretion and permeability, neuroimmune interactions, visceral sensation, and peristalsis, makes researchers and gastroenterologists to focus attention on the key roles of mast cells in the pathogenesis of IBS. Numerous studies have been carried out to identify the mechanisms in the development, infiltration, activation, and degranulation of intestinal mast cells, as well as the actions of mast cells in the processes of mucosal barrier disruption, mucosal immune dysregulation, visceral hypersensitivity, dysmotility, and local and central stress in IBS. Moreover, therapies targeting mast cells, such as mast cell stabilizers (cromoglycate and ketotifen) and antagonists of histamine and serotonin receptors, have been tried in IBS patients, and have partially exhibited considerable efficacy. This review focuses on recent advances in the role of mast cells in IBS, with particular emphasis on bridging experimental data with clinical therapeutics for IBS patients.
Collapse
Affiliation(s)
- Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,
China
| | - Jun Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,
China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,
China
| |
Collapse
|
44
|
Morampudi V, Conlin VS, Dalwadi U, Wu X, Marshall KC, Nguyen C, Vallance BA, Jacobson K. Vasoactive intestinal peptide prevents PKCε-induced intestinal epithelial barrier disruption during EPEC infection. Am J Physiol Gastrointest Liver Physiol 2015; 308:G389-402. [PMID: 25501546 DOI: 10.1152/ajpgi.00195.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We previously showed that vasoactive intestinal peptide (VIP) protects against bacterial pathogen-induced epithelial barrier disruption and colitis, although the mechanisms remain poorly defined. The aim of the current study was to identify cellular pathways of VIP-mediated protection with use of pharmacological inhibitors during enteropathogenic Escherichia coli (EPEC) infection of Caco-2 cell monolayers and during Citrobacter rodentium-induced colitis. EPEC-induced epithelial barrier disruption involved the PKC pathway but was independent of functional cAMP, Rho, and NF-κB pathways. VIP mediated its protective effects by inhibiting EPEC-induced PKC activity and increasing expression of the junctional protein claudin-4. Short-term treatment with TPA, which is known to activate PKC, was inhibited by VIP pretreatment, while PKC degradation via long-term treatment with TPA mimicked the protective actions of VIP. Immunostaining for specific PKC isotypes showed upregulated expression of PKCθ and PKCε during EPEC infection. Treatment with specific inhibitors revealed a critical role for PKCε in EPEC-induced barrier disruption. Furthermore, activation of PKCε and loss of barrier integrity correlated with claudin-4 degradation. In contrast, inhibition of PKCε by VIP pretreatment or the PKCε inhibitor maintained membrane-bound claudin-4 levels, along with barrier function. Finally, in vivo treatment with the PKCε inhibitor protected mice from C. rodentium-induced colitis. In conclusion, EPEC infection increases intracellular PKCε levels, leading to decreased claudin-4 levels and compromising epithelial barrier integrity. VIP inhibits PKCε activation, thereby attenuating EPEC-induced barrier disruption.
Collapse
Affiliation(s)
- V Morampudi
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - V S Conlin
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - U Dalwadi
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - X Wu
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - K C Marshall
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - C Nguyen
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - B A Vallance
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - K Jacobson
- Child and Family Research Institute, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; Division of Gastroenterology, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
45
|
Abstract
The aetiology and pathology of IBS, a functional bowel disorder thought to lack an organic cause, is largely unknown. However, studies suggest that various features, such as altered composition of the gut microbiota, together with increased intestinal permeability, a changed balance in the enteroendocrine system and a dysregulated immune system in the gut, most likely have an important role in IBS. Exactly how these entities act together and give rise to symptoms is still unknown, but an altered gut microbiota composition could lead to dysregulation of the intestinal barrier as well as the enteroendocrine and the immune systems, which (through interactions with the nervous system) might generate symptoms. This Review highlights the crosstalk between the gut microbiota, the enteroendocrine system, the immune system and the role of intestinal permeability in patients with IBS.
Collapse
|
46
|
Retrofitting the battlements: tight junction remodeling as a novel antimicrobial approach. Future Med Chem 2015; 7:9-13. [DOI: 10.4155/fmc.14.146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
47
|
Sharkey KA, Savidge TC. Reprint of: Role of enteric neurotransmission in host defense and protection of the gastrointestinal tract. Auton Neurosci 2014; 182:70-82. [PMID: 24674836 DOI: 10.1016/j.autneu.2014.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/11/2013] [Indexed: 12/11/2022]
Abstract
Host defense is a vital role played by the gastrointestinal tract. As host to an enormous and diverse microbiome, the gut has evolved an elaborate array of chemical and physicals barriers that allow the digestion and absorption of nutrients without compromising the mammalian host. The control of such barrier functions requires the integration of neural, humoral, paracrine and immune signaling, involving redundant and overlapping mechanisms to ensure, under most circumstances, the integrity of the gastrointestinal epithelial barrier. Here we focus on selected recent developments in the autonomic neural control of host defense functions used in the protection of the gut from luminal agents, and discuss how the microbiota may potentially play a role in enteric neurotransmission. Key recent findings include: the important role played by subepithelial enteric glia in modulating intestinal barrier function, identification of stress-induced mechanisms evoking barrier breakdown, neural regulation of epithelial cell proliferation, the role of afferent and efferent vagal pathways in regulating barrier function, direct evidence for bacterial communication to the enteric nervous system, and microbial sources of enteric neurotransmitters. We discuss these new and interesting developments in our understanding of the role of the autonomic nervous system in gastrointestinal host defense.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| | - Tor C Savidge
- Texas Children's Microbiome Center, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
48
|
Role of enteric neurotransmission in host defense and protection of the gastrointestinal tract. Auton Neurosci 2013; 181:94-106. [PMID: 24412639 DOI: 10.1016/j.autneu.2013.12.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/11/2013] [Indexed: 12/24/2022]
Abstract
Host defense is a vital role played by the gastrointestinal tract. As host to an enormous and diverse microbiome, the gut has evolved an elaborate array of chemical and physicals barriers that allow the digestion and absorption of nutrients without compromising the mammalian host. The control of such barrier functions requires the integration of neural, humoral, paracrine and immune signaling, involving redundant and overlapping mechanisms to ensure, under most circumstances, the integrity of the gastrointestinal epithelial barrier. Here we focus on selected recent developments in the autonomic neural control of host defense functions used in the protection of the gut from luminal agents, and discuss how the microbiota may potentially play a role in enteric neurotransmission. Key recent findings include: the important role played by subepithelial enteric glia in modulating intestinal barrier function, identification of stress-induced mechanisms evoking barrier breakdown, neural regulation of epithelial cell proliferation, the role of afferent and efferent vagal pathways in regulating barrier function, direct evidence for bacterial communication to the enteric nervous system, and microbial sources of enteric neurotransmitters. We discuss these new and interesting developments in our understanding of the role of the autonomic nervous system in gastrointestinal host defense.
Collapse
|