1
|
Gawey BJ, Mars RA, Kashyap PC. The role of the gut microbiome in disorders of gut-brain interaction. FEBS J 2025; 292:1357-1377. [PMID: 38922780 PMCID: PMC11664017 DOI: 10.1111/febs.17200] [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: 01/14/2024] [Revised: 04/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Disorders of Gut-Brain Interaction (DGBI) are widely prevalent and commonly encountered in gastroenterology practice. While several peripheral and central mechanisms have been implicated in the pathogenesis of DGBI, a recent body of work suggests an important role for the gut microbiome. In this review, we highlight how gut microbiota and their metabolites affect physiologic changes underlying symptoms in DGBI, with a particular focus on their mechanistic influence on GI transit, visceral sensitivity, intestinal barrier function and secretion, and CNS processing. This review emphasizes the complexity of local and distant effects of microbial metabolites on physiological function, influenced by factors such as metabolite concentration, duration of metabolite exposure, receptor location, host genetics, and underlying disease state. Large-scale in vitro work has elucidated interactions between host receptors and the microbial metabolome but there is a need for future research to integrate such preclinical findings with clinical studies. The development of novel, targeted therapeutic strategies for DGBI hinges on a deeper understanding of these metabolite-host interactions, offering exciting possibilities for the future of treatment of DGBI.
Collapse
Affiliation(s)
- Brent J Gawey
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ruben A Mars
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
2
|
Trebesova H, Monaco M, Baldassari S, Ailuno G, Lancellotti E, Caviglioli G, Pittaluga AM, Grilli M. Unveiling Niaprazine's Potential: Behavioral Insights into a Re-Emerging Anxiolytic Agent. Biomedicines 2024; 12:2087. [PMID: 39335600 PMCID: PMC11428487 DOI: 10.3390/biomedicines12092087] [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: 07/18/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Ongoing global research actions seek to comprehensively understand the adverse impact of stress and anxiety on the physical and mental health of both human beings and animals. Niaprazine (NIA) is a chemical compound that belongs to the class of piperazine derivatives. This compound has recently gained renewed attention due to its potential therapeutic properties for treating certain conditions such as anxiety. Despite its potential benefits, the behavioral effects of NIA have not been thoroughly investigated. This study aimed to examine NIA's potential as an anti-anxiety and anti-stress agent. After administering either vehicle or NIA in their drinking water to mice for 14 days, we conducted behavioral analyses using the Marble Burying Test and the Elevated Plus Maze test. NIA-treated mice spend more time in the open arms and bury fewer marbles. Moreover, a stability study confirmed the linear relationship between NIA concentration and its response across concentrations encompassing the NIA mother solution and the NIA solutions administered to mice. Also, a preliminary synaptic toxicity analysis showed no direct damage to cortical nerve endings. Here, we show that NIA can modulate anxiety-related behaviors without significantly impacting exploratory activity or adverse effects. Our work describes new findings that contribute to the research on safer and more tolerable anxiety management options.
Collapse
Affiliation(s)
- Hanna Trebesova
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genova, 16148 Genoa, Italy
| | - Martina Monaco
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genova, 16148 Genoa, Italy
| | - Sara Baldassari
- Pharmaceutical Technology Unit, Department of Pharmacy, University of Genova, 16148 Genoa, Italy
| | - Giorgia Ailuno
- Pharmaceutical Technology Unit, Department of Pharmacy, University of Genova, 16148 Genoa, Italy
| | | | - Gabriele Caviglioli
- Pharmaceutical Technology Unit, Department of Pharmacy, University of Genova, 16148 Genoa, Italy
| | - Anna Maria Pittaluga
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genova, 16148 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Massimo Grilli
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genova, 16148 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
3
|
Nozu T, Arie H, Miyagishi S, Ishioh M, Takakusaki K, Okumura T. Tranilast alleviates visceral hypersensitivity and colonic hyperpermeability by suppressing NLRP3 inflammasome activation in irritable bowel syndrome rat models. Int Immunopharmacol 2024; 133:112099. [PMID: 38643709 DOI: 10.1016/j.intimp.2024.112099] [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: 02/14/2024] [Revised: 03/30/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
Visceral hypersensitivity resulting from compromised gut barrier with activated immune system is a key feature of irritable bowel syndrome (IBS). Corticotropin-releasing factor (CRF) and Toll-like receptor 4 (TLR4) activate proinflammatory cytokine signaling to induce these changes, which is one of the mechanisms of IBS. As activation of the NLRP3 inflammasome by lipopolysaccharide (LPS) or TLR4 leads to release interleukin (IL)-1β, the NLRP3 inflammasome may be involved in the pathophysiology of IBS. Tranilast, an anti-allergic drug has been demonstrated to inhibit the NLRP3 inflammasome, and we evaluated the impact of tranilast on visceral hypersensitivity and colonic hyperpermeability induced by LPS or CRF (IBS rat model). Visceral pain threshold caused by colonic balloon distention was measured by monitoring abdominal muscle contractions electrophysiologically. Colonic permeability was determined by quantifying the absorbed Evans blue within the colonic tissue. Colonic protein levels of NLRP3 and IL-1β were assessed by immunoblot or ELISA. Intragastric administration of tranilast (20-200 mg/kg) for 3 days inhibited LPS (1 mg/kg)-induced visceral hypersensitivity and colonic hyperpermeability in a dose-dependent manner. Simultaneously, tranilast also abolished these alterations induced by CRF (50 µg/kg). LPS increased colonic protein levels of NLRP3 and IL-1β, and tranilast inhibited these changes. β-hydroxy butyrate, an NLRP3 inhibitor, also abolished visceral hypersensitivity and colonic hyperpermeability caused by LPS. In contrast, IL-1β induced similar GI alterations to LPS, which were not modified by tranilast. In conclusion, tranilast improved visceral pain and colonic barrier by suppression of the NLRP3 inflammasome in IBS rat models. Tranilast may be useful for IBS treating.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Center for Medical Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Hideyuki Arie
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Saori Miyagishi
- Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Masatomo Ishioh
- Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Kaoru Takakusaki
- Division of Neuroscience, Department of Physiology, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Toshikatsu Okumura
- Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| |
Collapse
|
4
|
Decraecker L, De Looze D, Hirsch DP, De Schepper H, Arts J, Caenepeel P, Bredenoord AJ, Kolkman J, Bellens K, Van Beek K, Pia F, Peetermans W, Vanuytsel T, Denadai-Souza A, Belmans A, Boeckxstaens G. Treatment of non-constipated irritable bowel syndrome with the histamine 1 receptor antagonist ebastine: a randomised, double-blind, placebo-controlled trial. Gut 2024; 73:459-469. [PMID: 38191268 DOI: 10.1136/gutjnl-2023-331634] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
OBJECTIVE We evaluated the histamine 1 receptor antagonist ebastine as a potential treatment for patients with non-constipated irritable bowel syndrome (IBS) in a randomised, placebo-controlled phase 2 study. METHODS Non-constipated patients with IBS fulfilling the Rome III criteria were randomly assigned to 20 mg ebastine or placebo for 12 weeks. Subjects scored global relief of symptoms (GRS) and abdominal pain intensity (API). A subject was considered a weekly responder for GRS if total or obvious relief was reported and a responder for API if the weekly average pain score was reduced by at least 30% vs baseline. The primary endpoints were the proportion of subjects who were weekly responders for at least 6 out of the 12 treatment weeks for both GRS and API ('GRS+API', composite endpoint) and for GRS and API separately. RESULTS 202 participants (32±11 years, 68% female) were randomly allocated to receive ebastine (n=101) or placebo (n=101). Treatment with ebastine resulted in significantly more responders (12%, 12/92) for GRS+API compared with placebo (4%, 4/87, p=0.047) while the proportion of responders for GRS and API separately was higher for ebastine compared with placebo, although not statistically significant (placebo vs ebastine, GRS: 7% (6/87) vs 15% (14/91), p=0.072; API: 25% (20/85) vs 37% (34/92), p=0.081). CONCLUSIONS Our study shows that ebastine is superior to placebo and should be further evaluated as novel treatment for patients with non-constipated IBS. TRIAL REGISTRATION NUMBER The study protocol was approved by the local ethics committee of each study site (EudraCT number: 2013-001199-39; ClinicalTrials.gov identifier: NCT01908465).
Collapse
Affiliation(s)
- Lisse Decraecker
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven, Belgium
| | - Danny De Looze
- Department of Gastroenterology and Hepatology, UZ Gent, Gent, Belgium
| | - David P Hirsch
- Department of Gastroenterology and Hepatology, Academic Medical Centre, Amsterdam, The Netherlands
| | - Heiko De Schepper
- Department of Translational Research in Immunology and Inflammation, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Edegem, Belgium
| | - Joris Arts
- Department of Gastroenterology, AZ Sint-Lucas Brugge, Brugge, Belgium
- Department of Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Belgium
| | - Philip Caenepeel
- Department of Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Belgium
- Department of Gastroenterology, Ziekenhuis Oost-Limburg - Campus Sint Jan, Genk, Belgium
| | - Albert J Bredenoord
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jeroen Kolkman
- Department of Gastroenterology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Koen Bellens
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Belgium
| | - Kim Van Beek
- Clinical Trial Center, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Belgium
| | - Fedrica Pia
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven, Belgium
| | - Willy Peetermans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Internal Medicine, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Belgium
| | - Tim Vanuytsel
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Belgium
| | | | - Ann Belmans
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Department of Chronic Diseases, Metabolism, and Ageing, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, KU Leuven University Hospitals Leuven Gasthuisberg Campus, Leuven, Belgium
| |
Collapse
|
5
|
Lashgari NA, Roudsari NM, Momtaz S, Niazi Shahraki F, Zandi N, Pazoki B, Farzaei MH, Ghasemi M, Abdollahi M, Abdolghaffari AH. Systematic Review on Herbal Preparations for Controlling Visceral Hypersensitivity in Functional Gastrointestinal Disorders. Curr Pharm Biotechnol 2024; 25:1632-1650. [PMID: 38258770 DOI: 10.2174/0113892010261502231102040149] [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: 05/09/2023] [Revised: 08/24/2023] [Accepted: 09/19/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Visceral hypersensitivity (VH) is an overreaction of the gastrointestinal (GI) tract to various stimuli and is characterized by hyperalgesia and/or allodynia. VH contributes to the etiology of many GI dysfunctions, particularly irritable bowel syndrome (IBS). Although the exact mechanisms underlying VH are yet to be found, inflammation and oxidative stress, psychosocial factors, and sensorimotor alterations may play significant roles in it. OBJECTIVE In this review, we provide an overview of VH and its pathophysiological function in GI disorders. Adverse effects of synthetic drugs may make herbal agents a good candidate for pain management. Therefore, in this review, we will discuss the efficacy of herbal agents in the management of VH with a focus on their anti-inflammatory and antioxidant potentials. METHODS Data were extracted from clinical and animal studies published in English between 2004 and June, 2020, which were collected from PubMed, Google Scholar, Scopus, and Cochrane Library. RESULTS Overall, Radix, Melissia, Glycyrrhizae, Mentha, and Liquorice were the most efficient herbals for VH management in IBS and dyspepsia, predominantly through modulation of the mRNA expression of transient receptor potential vanilloid type-1 (TRPV1) and suppression of 5- hydroxytryptamine 3 (5-HT3) or the serotonin receptors. CONCLUSION Considering the positive effects of herbal formulations in VH management, further research on novel herbal and/or herbal/chemical preparations is warranted.
Collapse
Affiliation(s)
- Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Department of Pharmacology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Faezeh Niazi Shahraki
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nadia Zandi
- Department of Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| | - Benyamin Pazoki
- Department of Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts School of Medicine, Worcester, MA 01655, USA
| | - Mohammad Abdollahi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
6
|
Liu XY, Wu SD. Fecal microbiota transplantation for treatment of irritable bowel syndrome: Current advances and future perspectives. Shijie Huaren Xiaohua Zazhi 2023; 31:922-932. [DOI: 10.11569/wcjd.v31.i22.922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a kind of functional gastroin-testinal disorder, characterized by recurrent abdominal pain and altered bowel habits. IBS adversely affects the quality of life of patients for the lack of effective treatment. The etiology of IBS remains poorly known. Previous studies suggested a possible role of gut dysbiosis in IBS pathogenesis. Fecal microbiota transplantation (FMT), which aims to reverse the gut dysbiosis, is a promising strategy in IBS management. In this review, we summarize the role of the gut microbiota in IBS pathogenesis from different aspects. We also review recent studies on efficacy evaluation of FMT in IBS. Besides, we discuss factors affecting the efficacy of FMT, hoping to provide a reference for future IBS treatment strategies targeting the gut microbiota.
Collapse
Affiliation(s)
- Xin-Yi Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Sheng-Di Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| |
Collapse
|
7
|
van Thiel I, de Jonge W, van den Wijngaard R. Fungal feelings in the irritable bowel syndrome: the intestinal mycobiome and abdominal pain. Gut Microbes 2023; 15:2168992. [PMID: 36723172 PMCID: PMC9897793 DOI: 10.1080/19490976.2023.2168992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Although the gut microbiota consists of bacteria, viruses, and fungi, most publications addressing the microbiota-gut-brain axis in irritable bowel syndrome (IBS) have a sole focus on bacteria. This may relate to the relatively low presence of fungi and viruses as compared to bacteria. Yet, in the field of inflammatory bowel disease research, the publication of several papers addressing the role of the intestinal mycobiome now suggested that these low numbers do not necessarily translate to irrelevance. In this review, we discuss the available clinical and preclinical IBS mycobiome data, and speculate how these recent findings may relate to earlier observations in IBS. By surveying literature from the broader mycobiome research field, we identified questions open to future IBS-oriented investigations.
Collapse
Affiliation(s)
- Iam van Thiel
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Wj de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Department of General, Visceral-, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Rm van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands,CONTACT RM van den Wijngaard Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Meibergdreef 69-71, Amsterdam1105 BK, The Netherlands
| |
Collapse
|
8
|
Liu A, Gao W, Zhu Y, Hou X, Chu H. Gut Non-Bacterial Microbiota: Emerging Link to Irritable Bowel Syndrome. Toxins (Basel) 2022; 14:596. [PMID: 36136534 PMCID: PMC9503233 DOI: 10.3390/toxins14090596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022] Open
Abstract
As a common functional gastrointestinal disorder, irritable bowel syndrome (IBS) significantly affects personal health and imposes a substantial economic burden on society, but the current understanding of its occurrence and treatment is still inadequate. Emerging evidence suggests that IBS is associated with gut microbial dysbiosis, but most studies focus on the bacteria and neglect other communities of the microbiota, including fungi, viruses, archaea, and other parasitic microorganisms. This review summarizes the latest findings that link the nonbacterial microbiota with IBS. IBS patients show less fungal and viral diversity but some alterations in mycobiome, virome, and archaeome, such as an increased abundance of Candida albicans. Moreover, fungi and methanogens can aid in diagnosis. Fungi are related to distinct IBS symptoms and induce immune responses, intestinal barrier disruption, and visceral hypersensitivity via specific receptors, cells, and metabolites. Novel therapeutic methods for IBS include fungicides, inhibitors targeting fungal pathogenic pathways, probiotic fungi, prebiotics, and fecal microbiota transplantation. Additionally, viruses, methanogens, and parasitic microorganisms are also involved in the pathophysiology and treatment. Therefore, the gut nonbacterial microbiota is involved in the pathogenesis of IBS, which provides a novel perspective on the noninvasive diagnosis and precise treatment of this disease.
Collapse
Affiliation(s)
- Ao Liu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yixin Zhu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
9
|
Hasler WL, Grabauskas G, Singh P, Owyang C. Mast cell mediation of visceral sensation and permeability in irritable bowel syndrome. Neurogastroenterol Motil 2022; 34:e14339. [PMID: 35315179 PMCID: PMC9286860 DOI: 10.1111/nmo.14339] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/09/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022]
Abstract
Abnormalities of mast cell structure or function may play prominent roles in irritable bowel syndrome (IBS) symptom genesis. Mast cells show close apposition to sensory nerves and release bioactive substances in response to varied stimuli including infection, stress, and other neuroendocrine factors. Most studies focus on patients who develop IBS after enteric infection or who report diarrhea-predominant symptoms. Three topics underlying IBS pathogenesis have been emphasized in recent investigations. Visceral hypersensitivity to luminal stimulation is found in most IBS patients and may contribute to abdominal pain. Mast cell dysfunction also may disrupt epithelial barrier function which alters mucosal permeability potentially leading to altered bowel function and pain. Mast cell products including histamine, proteases, prostaglandins, and cytokines may participate in hypersensitivity and permeability defects, especially with diarrhea-predominant IBS. Recent experimental evidence indicates that the pronociceptive effects of histamine and proteases are mediated by the generation of prostaglandins in the mast cell. Enteric microbiome interactions including increased mucosal bacterial translocation may activate mast cells to elicit inflammatory responses underlying some of these pathogenic effects. Therapies to alter mast cell activity (mast cell stabilizers) or function (histamine antagonists) have shown modest benefits in IBS. Future investigations will seek to define patient subsets with greater potential to respond to therapies that address visceral hypersensitivity, epithelial permeability defects, and microbiome alterations secondary to mast cell dysfunction in IBS.
Collapse
Affiliation(s)
- William L. Hasler
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Gintautas Grabauskas
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Prashant Singh
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Chung Owyang
- Division of Gastroenterology and HepatologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| |
Collapse
|
10
|
van Thiel IAM, Stavrou AA, de Jong A, Theelen B, Davids M, Hakvoort TBM, Admiraal-van den Berg I, Weert ICM, de Kruijs MAMHV, Vu D, Moissl-Eichinger C, Heinsbroek SEM, Jonkers DMAE, Hagen F, Boekhout T, de Jonge WJ, van den Wijngaard RM. Genetic and phenotypic diversity of fecal Candida albicans strains in irritable bowel syndrome. Sci Rep 2022; 12:5391. [PMID: 35354908 PMCID: PMC8967921 DOI: 10.1038/s41598-022-09436-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 03/21/2022] [Indexed: 12/16/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a common disorder characterized by chronic abdominal pain and changes in bowel movements. Visceral hypersensitivity is thought to be responsible for pain complaints in a subset of patients. In an IBS-like animal model, visceral hypersensitivity was triggered by intestinal fungi, and lower mycobiota α-diversity in IBS patients was accompanied by a shift toward increased presence of Candida albicans and Saccharomyces cerevisiae. Yet, this shift was observed in hypersensitive as well as normosensitive patients and diversity did not differ between IBS subgroups. The latter suggests that, when a patient changes from hyper- to normosensitivity, the relevance of intestinal fungi is not necessarily reflected in compositional mycobiota changes. We now confirmed this notion by performing ITS1 sequencing on an existing longitudinal set of fecal samples. Since ITS1 methodology does not recognize variations within species, we next focused on heterogeneity within cultured healthy volunteer and IBS-derived C. albicans strains. We observed inter- and intra-individual genomic variation and partial clustering of strains from hypersensitive patients. Phenotyping showed differences related to growth, yeast-to-hyphae morphogenesis and gene expression, specifically of the gene encoding fungal toxin candidalysin. Our investigations emphasize the need for strain-specific cause-and-effect studies within the realm of IBS research.
Collapse
Affiliation(s)
- Isabelle A M van Thiel
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Aimilia A Stavrou
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands.,Institute of Biodiversity and Ecosystem Dynamics (IBED), University of Amsterdam, Amsterdam, The Netherlands
| | - Auke de Jong
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands.,Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bart Theelen
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Mark Davids
- Laboratory of Experimental Vascular Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands.,Microbiota Center Amsterdam, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Theodorus B M Hakvoort
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.,Microbiota Center Amsterdam, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Iris Admiraal-van den Berg
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.,Microbiota Center Amsterdam, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Isabelle C M Weert
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Martine A M Hesselink-van de Kruijs
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Duong Vu
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Center for Microbiome Research, Medical University Graz, Graz, Austria
| | - Sigrid E M Heinsbroek
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.,Gastroenterology and Hepatology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Daisy M A E Jonkers
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ferry Hagen
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands.,Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Teun Boekhout
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands.,Institute of Biodiversity and Ecosystem Dynamics (IBED), University of Amsterdam, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.,Gastroenterology and Hepatology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands.,Department of General, Visceral, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - René M van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands. .,Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands. .,Gastroenterology and Hepatology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Louwies T, Meerveld BGV. Abdominal Pain. COMPREHENSIVE PHARMACOLOGY 2022:132-163. [DOI: 10.1016/b978-0-12-820472-6.00037-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Aguilera-Lizarraga J, Florens M, Hussein H, Boeckxstaens G. Local immune response as novel disease mechanism underlying abdominal pain in patients with irritable bowel syndrome. Acta Clin Belg 2021; 77:889-896. [PMID: 34709996 DOI: 10.1080/17843286.2021.1996069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Irritable bowel syndrome (IBS) is the most frequently diagnosed functional gastrointestinal disorder, with a prevalence of up to 25% of the global population. IBS patients suffer from abnormal abdominal pain, or visceral hypersensitivity (VHS), associated with altered bowel habits in the absence of an organic detectable cause. The pathophysiology of the disease is incompletely understood, but the dysregulation of the brain-gut axis is well established in IBS. METHODS IBS onset is mainly triggered by infectious gastroenteritis, psychological factors, and dietary factors, but genetic predispositions and intestinal dysbiosis might also play a role. Additionally, immune activation, and particularly chronic mast cell activation, have been shown to underlie the development of abdominal pain in IBS. RESULTS By releasing increased levels of mediators, including histamine, mast cells sensitize enteric nociceptors and lead to VHS development. The mechanisms underlying aberrant mast cell activation in IBS are still under investigation, but we recently showed that a local break in oral tolerance to food antigens led to IgE-mediated mast cell activation and food-induced abdominal pain in preclinical models and in IBS patients. CONCLUSION The concept of food-mediated VHS highlights the potential of therapies targeting upstream mechanisms of mast cell sensitization to treat IBS.
Collapse
Affiliation(s)
- J. Aguilera-Lizarraga
- Center of Intestinal Neuro-immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Belgium
| | - M. Florens
- Center of Intestinal Neuro-immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Belgium
| | - H. Hussein
- Center of Intestinal Neuro-immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Belgium
| | - G. Boeckxstaens
- Center of Intestinal Neuro-immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Belgium
- Department of Gastroenterology & Hepatology, UZ Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Shulpekova YO, Nechaev VM, Popova IR, Deeva TA, Kopylov AT, Malsagova KA, Kaysheva AL, Ivashkin VT. Food Intolerance: The Role of Histamine. Nutrients 2021; 13:3207. [PMID: 34579083 PMCID: PMC8469513 DOI: 10.3390/nu13093207] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/16/2022] Open
Abstract
Histamine is a natural amine derived from L-histidine. Although it seems that our knowledge about this molecule is wide and diverse, the importance of histamine in many regulatory processes is still enigmatic. The interplay between different types of histamine receptors and the compound may cause ample effects, including histamine intoxication and so-called histamine intolerance or non-allergic food intolerance, leading to disturbances in immune regulation, manifestation of gastroenterological symptoms, and neurological diseases. Most cases of clinical manifestations of histamine intolerance are non-specific due to tissue-specific distribution of different histamine receptors and the lack of reproducible and reliable diagnostic markers. The diagnosis of histamine intolerance is fraught with difficulties, in addition to challenges related to the selection of a proper treatment strategy, the regular course of recovery, and reduced amelioration of chronic symptoms due to inappropriate treatment prescription. Here, we reviewed a history of histamine uptake starting from the current knowledge about its degradation and the prevalence of histamine precursors in daily food, and continuing with the receptor interactions after entering and the impacts on the immune, central nervous, and gastrointestinal systems. The purpose of this review is to build an extraordinarily specific method of histamine cycle assessment in regard to non-allergic intolerance and its possible dire consequences that can be suffered.
Collapse
Affiliation(s)
- Yulia O. Shulpekova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.O.S.); (V.M.N.); (I.R.P.); (V.T.I.)
| | - Vladimir M. Nechaev
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.O.S.); (V.M.N.); (I.R.P.); (V.T.I.)
| | - Irina R. Popova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.O.S.); (V.M.N.); (I.R.P.); (V.T.I.)
| | - Tatiana A. Deeva
- Department of Biological Chemistry, Sechenov University, 119991 Moscow, Russia;
| | - Arthur T. Kopylov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 123098 Moscow, Russia; (A.T.K.); (A.L.K.)
| | - Kristina A. Malsagova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 123098 Moscow, Russia; (A.T.K.); (A.L.K.)
| | - Anna L. Kaysheva
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 123098 Moscow, Russia; (A.T.K.); (A.L.K.)
| | - Vladimir T. Ivashkin
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.O.S.); (V.M.N.); (I.R.P.); (V.T.I.)
| |
Collapse
|
14
|
Mahurkar-Joshi S, Rankin CR, Videlock EJ, Soroosh A, Verma A, Khandadash A, Iliopoulos D, Pothoulakis C, Mayer EA, Chang L. The Colonic Mucosal MicroRNAs, MicroRNA-219a-5p, and MicroRNA-338-3p Are Downregulated in Irritable Bowel Syndrome and Are Associated With Barrier Function and MAPK Signaling. Gastroenterology 2021; 160:2409-2422.e19. [PMID: 33617890 PMCID: PMC8169529 DOI: 10.1053/j.gastro.2021.02.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 02/03/2021] [Accepted: 02/17/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Alterations in microRNA (miRNA) and in the intestinal barrier are putative risk factors for irritable bowel syndrome (IBS). We aimed to identify differentially expressed colonic mucosal miRNAs, their targets in IBS compared to healthy controls (HCs), and putative downstream pathways. METHODS Twenty-nine IBS patients (15 IBS with constipation [IBS-C], 14 IBS with diarrhea [IBS-D]), and 15 age-matched HCs underwent sigmoidoscopy with biopsies. A nCounter array was used to assess biopsy specimen-associated miRNA levels. A false discovery rate (FDR) < 10% was considered significant. Real-time polymerase chain reaction (PCR) was used to validate differentially expressed genes. To assess barrier function, trans-epithelial electrical resistance (TEER) and dextran flux assays were performed on Caco-2 intestinal epithelial cells that were transfected with miRNA-inhibitors or control inhibitors. Protein expression of barrier function associated genes was confirmed using western blots. RESULTS Four out of 247 miRNAs tested were differentially expressed in IBS compared to HCs (FDR < 10%). Real-time PCR validation suggested decreased levels of miR-219a-5p and miR-338-3p in IBS (P = .026 and P = .004), and IBS-C (P = .02 and P = .06) vs. HCs as the strongest associations. Inhibition of miR-219a-5p resulted in altered expression of proteasome/barrier function genes. Functionally, miR-219a-5p inhibition enhanced the permeability of intestinal epithelial cells as TEER was reduced (25-50%, P < .05) and dextran flux was increased (P < .01). Additionally, inhibition of miR-338-3p in cells caused alterations in the mitogen-activated protein kinase (MAPK) signaling pathway genes. CONCLUSION Two microRNAs that potentially affect permeability and visceral nociception were identified to be altered in IBS patients. MiR-219a-5p and miR-338-3p potentially alter barrier function and visceral hypersensitivity via neuronal and MAPK signaling and could be therapeutic targets in IBS.
Collapse
Affiliation(s)
- Swapna Mahurkar-Joshi
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Carl Robert Rankin
- UCLA Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Elizabeth Jane Videlock
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Artin Soroosh
- UCLA Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Abhishek Verma
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ariela Khandadash
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Dimitrios Iliopoulos
- UCLA Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Charalabos Pothoulakis
- UCLA Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Lin Chang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
15
|
van Thiel IAM, de Jonge WJ, Chiu IM, van den Wijngaard RM. Microbiota-neuroimmune cross talk in stress-induced visceral hypersensitivity of the bowel. Am J Physiol Gastrointest Liver Physiol 2020; 318:G1034-G1041. [PMID: 32308040 PMCID: PMC7642838 DOI: 10.1152/ajpgi.00196.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Visceral hypersensitivity of the lower gastrointestinal tract, defined as an increased response to colorectal distension, frequently prompts episodes of debilitating abdominal pain in irritable bowel syndrome (IBS). Although the pathophysiology of IBS is not yet fully elucidated, it is well known that stress is a major risk factor for development and acts as a trigger of pain sensation. Stress modulates both immune responses as well as the gut microbiota and vice versa. Additionally, either microbes themselves or through involvement of the immune system, activate or sensitize afferent nociceptors. In this paper, we review current knowledge on the influence of stress along the gut-brain-microbiota axis and exemplify relevant neuroimmune cross talk mechanisms in visceral hypersensitivity, working toward understanding how gut microbiota-neuroimmune cross talk contributes to visceral pain sensation in IBS patients.
Collapse
Affiliation(s)
- Isabelle A. M. van Thiel
- 1Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,2Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Wouter J. de Jonge
- 1Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,2Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands,3Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands,4Department of General, Visceral, Thoracic, and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Isaac M. Chiu
- 5Department of Immunology, Harvard Medical School. Boston, Massachusetts
| | - Rene M. van den Wijngaard
- 1Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,2Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands,3Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Siah KTH, Santosa A, Cheung CKY, Soh AYS, Bigliardi PL. Atopic Patients Who Fulfilled Rome III Criteria for Irritable Bowel Syndrome Had Higher Animal Danders Sensitization. J Neurogastroenterol Motil 2020; 26:267-273. [PMID: 32235034 PMCID: PMC7176502 DOI: 10.5056/jnm19181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/06/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
Background/Aims The relationship between animal exposure and irritable bowel syndrome (IBS) is debated. Epidemiological studies have shown that atopy is more prevalent in IBS patients and vice versa. We set out to examine the association between animal danders sensitization and IBS-like symptoms in atopic patients. Methods We recruited 69 consecutive atopic patients from the allergy clinic of a tertiary hospital. Subjects completed validated bowel questionnaires, underwent skin prick test, blood was collected for serum total immunoglobulin E, and ImmunoCAP immune solid-phase allergen chip (ISAC) IgE multiplex assay. Results Twenty-eight (41.0%) atopic patients fulfilled the Rome III IBS criteria (atopy-IBS). There were no differences in gender, age, pet ownership, total serum IgE, or food allergen sensitization between atopy-IBS group and atopy-non-IBS group. We found that atopy-IBS group had significantly higher number of positive skin prick test for cat dander (64.3% vs 24.4%, P < 0.001), dog dander (64.3% vs 41.5%, P = 0.015) and weed pollens (32.1% vs 14.6%, P = 0.050) compared to atopy-non-IBS group. Out of 112 components from 51 allergen sources (both aeroallergen and food allergens), only Fel d1 (a major cat dander antigen) IgE is significantly higher in atopy-IBS group than atopy-non-IBS group (21.4% vs 2.4%, P = 0.029). Majority of atopy-IBS patients had mixed-type IBS. Conclusions We demonstrated an association between animal danders sensitization, in particular cat dander sensitization, and IBS-like symptoms in atopic patients. Future studies are needed to explore the relationship between aeroallergen and functional gastrointestinal disorders. Sensitization may be related to the pathophysiology of IBS or it could be that we are missing aeroallergen-induced gut allergy.
Collapse
Affiliation(s)
- Kewin T H Siah
- Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Health System, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Amelia Santosa
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore
| | | | - Alex Y S Soh
- Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Health System, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Paul L Bigliardi
- Department of Dermatology, Division of Dermato-Allergy, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
van Thiel IAM, Botschuijver S, de Jonge WJ, Seppen J. Painful interactions: Microbial compounds and visceral pain. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165534. [PMID: 31634534 DOI: 10.1016/j.bbadis.2019.165534] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/18/2022]
Abstract
Visceral pain, characterized by abdominal discomfort, originates from organs in the abdominal cavity and is a characteristic symptom in patients suffering from irritable bowel syndrome, vulvodynia or interstitial cystitis. Most organs in which visceral pain originates are in contact with the external milieu and continuously exposed to microbes. In order to maintain homeostasis and prevent infections, the immune- and nervous system in these organs cooperate to sense and eliminate (harmful) microbes. Recognition of microbial components or products by receptors expressed on cells from the immune and nervous system can activate immune responses but may also cause pain. We review the microbial compounds and their receptors that could be involved in visceral pain development.
Collapse
Affiliation(s)
- I A M van Thiel
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - S Botschuijver
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - W J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - J Seppen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands.
| |
Collapse
|
20
|
Lomax AE, Pradhananga S, Sessenwein JL, O'Malley D. Bacterial modulation of visceral sensation: mediators and mechanisms. Am J Physiol Gastrointest Liver Physiol 2019; 317:G363-G372. [PMID: 31290688 DOI: 10.1152/ajpgi.00052.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The potential role of the intestinal microbiota in modulating visceral pain has received increasing attention during recent years. This has led to the identification of signaling pathways that have been implicated in communication between gut bacteria and peripheral pain pathways. In addition to the well-characterized impact of the microbiota on the immune system, which in turn affects nociceptor excitability, bacteria can modulate visceral afferent pathways by effects on enterocytes, enteroendocrine cells, and the neurons themselves. Proteases produced by bacteria, or by host cells in response to bacteria, can increase or decrease the excitability of nociceptive dorsal root ganglion (DRG) neurons depending on the receptor activated. Short-chain fatty acids generated by colonic bacteria are involved in gut-brain communication, and intracolonic short-chain fatty acids have pronociceptive effects in rodents but may be antinociceptive in humans. Gut bacteria modulate the synthesis and release of enteroendocrine cell mediators, including serotonin and glucagon-like peptide-1, which activate extrinsic afferent neurons. Deciphering the complex interactions between visceral afferent neurons and the gut microbiota may lead to the development of improved probiotic therapies for visceral pain.
Collapse
Affiliation(s)
- Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Sabindra Pradhananga
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Jessica L Sessenwein
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Dervla O'Malley
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
21
|
Botschuijver S, van Diest SA, van Thiel IAM, Saia RS, Strik AS, Yu Z, Maria-Ferreira D, Welting O, Keszthelyi D, Jennings G, Heinsbroek SEM, Elferink RPO, Schuren FHJ, de Jonge WJ, van den Wijngaard RM. Miltefosine treatment reduces visceral hypersensitivity in a rat model for irritable bowel syndrome via multiple mechanisms. Sci Rep 2019; 9:12530. [PMID: 31467355 PMCID: PMC6715706 DOI: 10.1038/s41598-019-49096-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/02/2019] [Indexed: 12/17/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a heterogenic, functional gastrointestinal disorder of the gut-brain axis characterized by altered bowel habit and abdominal pain. Preclinical and clinical results suggested that, in part of these patients, pain may result from fungal induced release of mast cell derived histamine, subsequent activation of sensory afferent expressed histamine-1 receptors and related sensitization of the nociceptive transient reporter potential channel V1 (TRPV1)-ion channel. TRPV1 gating properties are regulated in lipid rafts. Miltefosine, an approved drug for the treatment of visceral Leishmaniasis, has fungicidal effects and is a known lipid raft modulator. We anticipated that miltefosine may act on different mechanistic levels of fungal-induced abdominal pain and may be repurposed to IBS. In the IBS-like rat model of maternal separation we assessed the visceromotor response to colonic distension as indirect readout for abdominal pain. Miltefosine reversed post-stress hypersensitivity to distension (i.e. visceral hypersensitivity) and this was associated with differences in the fungal microbiome (i.e. mycobiome). In vitro investigations confirmed fungicidal effects of miltefosine. In addition, miltefosine reduced the effect of TRPV1 activation in TRPV1-transfected cells and prevented TRPV1-dependent visceral hypersensitivity induced by intracolonic-capsaicin in rat. Miltefosine may be an attractive drug to treat abdominal pain in IBS.
Collapse
Affiliation(s)
- Sara Botschuijver
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Sophie A van Diest
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Isabelle A M van Thiel
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Rafael S Saia
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Anne S Strik
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Zhumei Yu
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Department of Neurobiology, Tongji Medical College, HUST, Wuhan, People's Republic of China.,State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Daniele Maria-Ferreira
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Departamento de Farmacologia, Setor de Ciências Biológicas, Universidade Federal do Paraná, Curitiba, Brazil
| | - Olaf Welting
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Daniel Keszthelyi
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Gary Jennings
- Business Development, Redivia, Technische Universität, Dresden, Germany
| | - Sigrid E M Heinsbroek
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Ronald P Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Frank H J Schuren
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - René M van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands. .,Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Russell JP, Mohammadi E, Ligon CO, Johnson AC, Gershon MD, Rao M, Shen Y, Chan CC, Eidam HS, DeMartino MP, Cheung M, Oliff AI, Kumar S, Greenwood-Van Meerveld B. Exploring the Potential of RET Kinase Inhibition for Irritable Bowel Syndrome: A Preclinical Investigation in Rodent Models of Colonic Hypersensitivity. J Pharmacol Exp Ther 2019; 368:299-307. [PMID: 30413627 PMCID: PMC6346376 DOI: 10.1124/jpet.118.252973] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
Abdominal pain represents a significant complaint in patients with irritable bowel syndrome (IBS). While the etiology of IBS is incompletely understood, prior exposure to gastrointestinal inflammation or psychologic stress is frequently associated with the development of symptoms. Inflammation or stress-induced expression of growth factors or cytokines may contribute to the pathophysiology of IBS. Here, we aimed to investigate the therapeutic potential of inhibiting the receptor of glial cell line-derived neurotrophic factor, rearranged during transfection (RET), in experimental models of inflammation and stress-induced visceral hypersensitivity resembling IBS sequelae. In RET-cyan fluorescent protein [(CFP) RetCFP/+] mice, thoracic and lumbosacral dorsal root ganglia were shown to express RET, which colocalized with calcitonin gene-related peptide. To understand the role of RET in visceral nociception, we employed GSK3179106 as a potent, selective, and gut-restricted RET kinase inhibitor. Colonic hyperalgesia, quantified as exaggerated visceromotor response to graded pressures (0-60 mm Hg) of isobaric colorectal distension (CRD), was produced in multiple rat models induced 1) by colonic irritation, 2) following acute colonic inflammation, 3) by adulthood stress, and 4) by early life stress. In all the rat models, RET inhibition with GSK3179106 attenuated the number of abdominal contractions induced by CRD. Our findings identify a role for RET in visceral nociception. Inhibition of RET kinase with a potent, selective, and gut-restricted small molecule may represent a novel therapeutic strategy for the treatment of IBS through the attenuation of post-inflammatory and stress-induced visceral hypersensitivity.
Collapse
Affiliation(s)
- John P Russell
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Ehsan Mohammadi
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Casey O Ligon
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Anthony C Johnson
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Michael D Gershon
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Meenakshi Rao
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Yuhong Shen
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Chi-Chung Chan
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Hilary S Eidam
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Michael P DeMartino
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Mui Cheung
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Allen I Oliff
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Sanjay Kumar
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| | - Beverley Greenwood-Van Meerveld
- Virtual Proof of Concept Discovery Performance Unit, GlaxoSmithKline, King of Prussia, Pennsylvania (J.P.R., H.S.E., M.P.D., M.C., A.I.O., S.K.); Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (E.M., C.O.L., A.C.J., B.G.-V.M.); Department of Pathology and Cell Biology, College of Physicians and Surgeons (M.D.G.) and Department of Pediatrics (M.R.), Columbia University, New York, New York; and WuXi AppTec Co., Ltd., Waigaoqiao Free Trade Zone, Shanghai, China (Y.S., C.-C.C.)
| |
Collapse
|
23
|
Contribution of membrane receptor signalling to chronic visceral pain. Int J Biochem Cell Biol 2018; 98:10-23. [DOI: 10.1016/j.biocel.2018.02.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 12/18/2022]
|
24
|
Lilli NL, Quénéhervé L, Haddara S, Brochard C, Aubert P, Rolli-Derkinderen M, Durand T, Naveilhan P, Hardouin JB, De Giorgio R, Barbara G, Bruley des Varannes S, Coron E, Neunlist M. Glioplasticity in irritable bowel syndrome. Neurogastroenterol Motil 2018; 30:e13232. [PMID: 29027719 DOI: 10.1111/nmo.13232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/17/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Growing evidence indicates a wide array of cellular remodeling in the mucosal microenvironment during irritable bowel syndrome (IBS), which possibly contributes to pathophysiology and symptom generation. Here, we investigated whether enteric glial cells (EGC) may be altered, and which factors/mechanisms lead to these changes. METHODS Colonic mucosal biopsies of IBS patients (13 IBS-Constipation [IBS-C]; 10 IBS-Diarrhea [IBS-D]; 11 IBS-Mixed [IBS-M]) and 24 healthy controls (HC) were analyzed. Expression of S100β and GFAP was measured. Cultured rat EGC were incubated with supernatants from mucosal biopsies, then proliferation and Ca2+ response to ATP were analyzed using flow cytometry and Ca2+ imaging. Histamine and histamine 1-receptor (H1R) involvement in the effects of supernatant upon EGC was analyzed. KEY RESULTS Compared to HC, the mucosal area immunoreactive for S100β was significantly reduced in biopsies of IBS patients, independently of the IBS subtype. IBS-C supernatants reduced EGC proliferation and IBS-D and IBS-M supernatants reduced Ca2+ response to ATP in EGC. EGC expressed H1R and the effects of supernatant upon Ca2+ response to ATP in EGC were blocked by pyrilamine and reproduced by histamine via H1R. IBS supernatants reduced mRNA expression of connexin-43. The S100β-stained area was negatively correlated with the frequency and intensity of pain and bloating. CONCLUSION AND INFERENCES Changes in EGC occur in IBS, involving mucosal soluble factors. Histamine, via activation of H1R-dependent pathways, partly mediates altered Ca2+ response to ATP in EGC. These changes may contribute to the pathophysiology and the perception of pain and bloating in patients with IBS.
Collapse
Affiliation(s)
- N L Lilli
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - L Quénéhervé
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - S Haddara
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - C Brochard
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France
| | - P Aubert
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - M Rolli-Derkinderen
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - T Durand
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - P Naveilhan
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - J-B Hardouin
- Université de Nantes, INSERM, SPHERE, Université Bretagne Loire, Nantes, France
| | - R De Giorgio
- Department of Medical and Surgical Sciences, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - G Barbara
- Department of Medical and Surgical Sciences, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - S Bruley des Varannes
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - E Coron
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - M Neunlist
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France
| |
Collapse
|
25
|
Mujagic Z, Jonkers DMAE, Ludidi S, Keszthelyi D, Hesselink MA, Weerts ZZRM, Kievit RN, Althof JF, Leue C, Kruimel JW, van Schooten FJ, Masclee AAM. Biomarkers for visceral hypersensitivity in patients with irritable bowel syndrome. Neurogastroenterol Motil 2017; 29. [PMID: 28675524 DOI: 10.1111/nmo.13137] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Increased visceral sensitivity is observed in up to 60% of patients with Irritable Bowel Syndrome (IBS). Mucosal inflammation, altered neuroendocrine activity and intraluminal metabolic processes may contribute to the development of visceral hypersensitivity. Previously, we demonstrated that biomarkers, indicative for these biological processes, were altered in IBS patients compared to healthy controls. However, how these processes relate to visceral hypersensitivity is unknown. AIM The aim of this study was to provide insight in biological processes associated with visceral hypersensitivity. Fecal and plasma biomarkers were measured in normosensitive and hypersensitive IBS patients. METHODS A total of 167 IBS patients underwent a rectal barostat procedure to assess visceral sensitivity to pain. Based on the outcome, patients were classified into a normosensitive or hypersensitive group. Calprotectin, human β-defensin 2 (HBD2), chromogranin A (CgA), and short chain fatty acids (SCFAs) were measured in feces, citrulline in plasma, and serotonin and its main metabolite 5-hydroxyindoleacetic acid (5-HIAA) in platelet-poor plasma. KEY RESULTS Fecal markers and plasma citrulline were measured in 83 hypersensitive and 84 normosensitive patients, while platelet-poor plasma for the assessment of serotonin and 5-HIAA was available for a subgroup, i.e. 53 hypersensitive and 42 normosensitive patients. No statistically significant differences were found in concentrations of biomarkers between groups. Adjustment of the analyses for potential confounders, such as medication use, did not alter this conclusion. CONCLUSIONS & INFERENCES Our findings do not support a role for the biological processes as ascertained by biomarkers in visceral hypersensitivity in IBS patients. This study is registered in the US National Library of Medicine (clinicaltrials.gov, NCT00775060).
Collapse
Affiliation(s)
- Z Mujagic
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
- Top Institute Food & Nutrition (TiFN), Wageningen, The Netherlands
| | - D M A E Jonkers
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
- Top Institute Food & Nutrition (TiFN), Wageningen, The Netherlands
| | - S Ludidi
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - D Keszthelyi
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - M A Hesselink
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Z Z R M Weerts
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - R N Kievit
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J F Althof
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - C Leue
- Department of Psychiatry and Psychology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J W Kruimel
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - F J van Schooten
- Top Institute Food & Nutrition (TiFN), Wageningen, The Netherlands
- Department of Pharmacology and Toxicology, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - A A M Masclee
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
26
|
Greenwood-Van Meerveld B, Johnson AC. Stress-Induced Chronic Visceral Pain of Gastrointestinal Origin. Front Syst Neurosci 2017; 11:86. [PMID: 29213232 PMCID: PMC5702626 DOI: 10.3389/fnsys.2017.00086] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
Visceral pain is generally poorly localized and characterized by hypersensitivity to a stimulus such as organ distension. In concert with chronic visceral pain, there is a high comorbidity with stress-related psychiatric disorders including anxiety and depression. The mechanisms linking visceral pain with these overlapping comorbidities remain to be elucidated. Evidence suggests that long term stress facilitates pain perception and sensitizes pain pathways, leading to a feed-forward cycle promoting chronic visceral pain disorders such as irritable bowel syndrome (IBS). Early life stress (ELS) is a risk-factor for the development of IBS, however the mechanisms responsible for the persistent effects of ELS on visceral perception in adulthood remain incompletely understood. In rodent models, stress in adult animals induced by restraint and water avoidance has been employed to investigate the mechanisms of stress-induce pain. ELS models such as maternal separation, limited nesting, or odor-shock conditioning, which attempt to model early childhood experiences such as neglect, poverty, or an abusive caregiver, can produce chronic, sexually dimorphic increases in visceral sensitivity in adulthood. Chronic visceral pain is a classic example of gene × environment interaction which results from maladaptive changes in neuronal circuitry leading to neuroplasticity and aberrant neuronal activity-induced signaling. One potential mechanism underlying the persistent effects of stress on visceral sensitivity could be epigenetic modulation of gene expression. While there are relatively few studies examining epigenetically mediated mechanisms involved in visceral nociception, stress-induced visceral pain has been linked to alterations in DNA methylation and histone acetylation patterns within the brain, leading to increased expression of pro-nociceptive neurotransmitters. This review will discuss the potential neuronal pathways and mechanisms responsible for stress-induced exacerbation of chronic visceral pain. Additionally, we will review the importance of specific experimental models of adult stress and ELS in enhancing our understanding of the basic molecular mechanisms of pain processing.
Collapse
Affiliation(s)
- Beverley Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
- VA Medical Center, Oklahoma City, OK, United States
| | | |
Collapse
|
27
|
Abstract
Epidemiological studies indicate sex-related differences among functional gastrointestinal disorders (FGIDs) wherein females are more likely to receive a diagnosis than their male counterparts. However, the mechanism by which females exhibit an increased vulnerability for development of these pathophysiologies remains largely unknown, and therapeutic treatments are limited. The current chapter focuses on clinical research outlining our current knowledge of factors that contribute to the female predominance among FGID patients such as the menstrual cycle and sex hormones. In addition, we will discuss progress in preclinical research, including animal models, which serve as valuable tools for the investigation of the development and long term manifestation of symptoms observed within the patient population. Although much progress has been made, additional longitudinal studies in both clinical and preclinical research are necessary to identify more specific mechanisms underlying sex-related differences in FGIDs as well as targets for improved therapeutic approaches.
Collapse
|
28
|
Abstract
Preclinical research remains an important tool for discovery and validation of novel therapeutics for gastrointestinal disorders. While in vitro assays can be used to verify receptor-ligand interactions and test for structural activity of new compounds, only whole-animal studies can demonstrate drug efficacy within the gastrointestinal system. Most major gastrointestinal disorders have been modeled in animals; however the translational relevance of each model is not equal. The purpose of this chapter is to provide a critical evaluation of common animal models that are being used to develop pharmaceuticals for gastrointestinal disorders. For brevity, the models are presented for upper gastrointestinal disorders involving the esophagus, stomach, and small intestine and lower gastrointestinal disorders that focus on the colon. Particular emphasis is used to explain the face and construct validity of each model, and the limitations of each model, including data interpretation, are highlighted. This chapter does not evaluate models that rely on surgical or other non-pharmacological interventions for treatment.
Collapse
|
29
|
Botschuijver S, Roeselers G, Levin E, Jonkers DM, Welting O, Heinsbroek SEM, de Weerd HH, Boekhout T, Fornai M, Masclee AA, Schuren FHJ, de Jonge WJ, Seppen J, van den Wijngaard RM. Intestinal Fungal Dysbiosis Is Associated With Visceral Hypersensitivity in Patients With Irritable Bowel Syndrome and Rats. Gastroenterology 2017. [PMID: 28624575 DOI: 10.1053/j.gastro.2017.06.004] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Visceral hypersensitivity is one feature of irritable bowel syndrome (IBS). Bacterial dysbiosis might be involved in the activation of nociceptive sensory pathways, but there have been few studies of the role of the mycobiome (the fungal microbiome) in the development of IBS. We analyzed intestinal mycobiomes of patients with IBS and a rat model of visceral hypersensitivity. METHODS We used internal transcribed spacer 1-based metabarcoding to compare fecal mycobiomes of 18 healthy volunteers with those of 39 patients with IBS (with visceral hypersensitivity or normal levels of sensitivity). We also compared the mycobiomes of Long-Evans rats separated from their mothers (hypersensitive) with non-handled (normally sensitive) rats. We investigated whether fungi can cause visceral hypersensitivity using rats exposed to fungicide (fluconazole and nystatin). The functional relevance of the gut mycobiome was confirmed in fecal transplantation experiments: adult maternally separated rats were subjected to water avoidance stress (to induce visceral hypersensitivity), then given fungicide and donor cecum content via oral gavage. Other rats subjected to water avoidance stress were given soluble β-glucans, which antagonize C-type lectin domain family 7 member A (CLEC7A or DECTIN1) signaling via spleen-associated tyrosine kinase (SYK), a SYK inhibitor to reduce visceral hypersensitivity, or vehicle (control). The sensitivity of mast cells to fungi was tested with mesenteric windows (ex vivo) and the human mast cell line HMC-1. RESULTS α diversity (Shannon index) and mycobiome signature (stability selection) of both groups of IBS patients differed from healthy volunteers, and the mycobiome signature of hypersensitive patients differed from that of normally sensitive patients. We observed mycobiome dysbiosis in rats that had been separated from their mothers compared with non-handled rats. Administration of fungicide to hypersensitive rats reduced their visceral hypersensitivity to normal levels of sensitivity. Administration of cecal mycobiomes from rats that had been separated from their mothers (but not non-handled mycobiome) restored hypersensitivity to distension. Administration of soluble β-glucans or a SYK inhibitor reduced visceral hypersensitivity, compared with controls. Particulate β-glucan (a DECTIN-1 agonist) induced mast cell degranulation in mesenteric windows and HMC-1 cells responded to fungal antigens by release of histamine. CONCLUSIONS In an analysis of patients with IBS and controls, we associated fungal dysbiosis with IBS. In studies of rats, we found fungi to promote visceral hypersensitivity, which could be reduced by administration of fungicides, soluble β-glucans, or a SYK inhibitor. The intestinal fungi might therefore be manipulated for treatment of IBS-related visceral hypersensitivity.
Collapse
Affiliation(s)
- Sara Botschuijver
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Guus Roeselers
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Evgeni Levin
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Daisy M Jonkers
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Olaf Welting
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Sigrid E M Heinsbroek
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Heleen H de Weerd
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Teun Boekhout
- Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands; Institute for Biodiversity and Ecosystems Dynamics (IBED), University of Amsterdam, Amsterdam, The Netherlands
| | - Matteo Fornai
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands; Division of Pharmacology, Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ad A Masclee
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition, and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Frank H J Schuren
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jurgen Seppen
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - René M van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
30
|
Fabisiak A, Włodarczyk J, Fabisiak N, Storr M, Fichna J. Targeting Histamine Receptors in Irritable Bowel Syndrome: A Critical Appraisal. J Neurogastroenterol Motil 2017; 23:341-348. [PMID: 28551943 PMCID: PMC5503283 DOI: 10.5056/jnm16203] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/10/2017] [Accepted: 04/07/2017] [Indexed: 12/19/2022] Open
Abstract
Irritable bowel syndrome is a group of functional gastrointestinal disorders with not yet fully clarified etiology. Recent evidence suggesting that mast cells may play a central role in the pathogenesis of irritable bowel syndrome paves the way for agents targeting histamine receptors as a potential therapeutic option in clinical treatment. In this review, the role of histamine and histamine receptors is debated. Moreover, the clinical evidence of anti-histamine therapeutics in irritable bowel syndrome is discussed.
Collapse
Affiliation(s)
- Adam Fabisiak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz,
Poland
| | - Jakub Włodarczyk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz,
Poland
| | - Natalia Fabisiak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz,
Poland
| | - Martin Storr
- Center of Endoscopy, Starnberg,
Germany
- Walter Brendel Center of Experimental Medicine, Ludwig Maximilians University of Munich, Munich,
Germany
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz,
Poland
- Correspondence: Jakub Fichna, PhD, DSc, Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland, Tel: +48-42-272-5707, Fax: +48-42-272-5694, E-mail:
| |
Collapse
|
31
|
Boeckxstaens GE, Wouters MM. Neuroimmune factors in functional gastrointestinal disorders: A focus on irritable bowel syndrome. Neurogastroenterol Motil 2017; 29. [PMID: 28027594 DOI: 10.1111/nmo.13007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/11/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Abnormal abdominal pain perception is the most bothersome and difficult to treat symptom of functional gastrointestinal disorders (FGIDs). Visceral pain stimuli are perceived and transmitted by afferent neurons residing in the dorsal root ganglia that have sensory nerve endings in the gut wall and mesentery. Accumulating evidence indicates that peripheral activation and sensitization of these sensory nerve endings by bioactive mediators released by activated immune cells, in particular mast cells, can lead to aberrant neuroimmune interactions and the development and maintenance of visceral hypersensitivity. Besides direct neuronal activation, low concentrations of proteases, histamine, and serotonin can chronically sensitize nociceptors, such as TRP channels, leading to persistent aberrant pain perception. PURPOSE This review discusses the potential mechanisms underlying aberrant neuroimmune interactions in peripheral sensitization of sensory nerves. A better understanding of the cells, mediators, and molecular mechanisms triggering persistent aberrant neuroimmune interactions brings new insights into their contribution to the physiology and pathophysiology of visceral pain perception and provides novel opportunities for more efficient therapeutic treatments for these disorders.
Collapse
Affiliation(s)
- G E Boeckxstaens
- Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven University, Leuven, Belgium
| | - M M Wouters
- Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven University, Leuven, Belgium
| |
Collapse
|
32
|
Farzaei MH, Bahramsoltani R, Abdollahi M, Rahimi R. The Role of Visceral Hypersensitivity in Irritable Bowel Syndrome: Pharmacological Targets and Novel Treatments. J Neurogastroenterol Motil 2016; 22:558-574. [PMID: 27431236 PMCID: PMC5056566 DOI: 10.5056/jnm16001] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/26/2016] [Accepted: 04/17/2016] [Indexed: 12/13/2022] Open
Abstract
Irritable bowel syndrome (IBS) is the most common disorder referred to gastroenterologists and is characterized by altered bowel habits, abdominal pain, and bloating. Visceral hypersensitivity (VH) is a multifactorial process that may occur within the peripheral or central nervous systems and plays a principal role in the etiology of IBS symptoms. The pharmacological studies on selective drugs based on targeting specific ligands can provide novel therapies for modulation of persistent visceral hyperalgesia. The current paper reviews the cellular and molecular mechanisms underlying therapeutic targeting for providing future drugs to protect or treat visceroperception and pain sensitization in IBS patients. There are a wide range of mediators and receptors participating in visceral pain perception amongst which substances targeting afferent receptors are attractive sources of novel drugs. Novel therapeutic targets for the management of VH include compounds which alter gut-brain pathways and local neuroimmune pathways. Molecular mediators and receptors participating in pain perception and visceroperception include histamine-1 receptors, serotonin (5-hydrodytryptamine) receptors, transient receptor potential vanilloid type I, tachykinins ligands, opioid receptors, voltage-gated channels, tyrosine receptor kinase receptors, protease-activated receptors, adrenergic system ligands, cannabinoid receptors, sex hormones, and glutamate receptors which are discussed in the current review. Moreover, several plant-derived natural compounds with potential to alleviate VH in IBS have been highlighted. VH has an important role in the pathology and severity of complications in IBS. Therefore, managing VH can remarkably modulate the symptoms of IBS. More preclinical and clinical investigations are needed to provide efficacious and targeted medicines for the management of VH.
Collapse
Affiliation(s)
- Mohammad H Farzaei
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah,
Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah,
Iran
| | - Roodabeh Bahramsoltani
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran,
Iran
| | - Mohammad Abdollahi
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran,
Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran,
Iran
| | - Roja Rahimi
- Department of Traditional Pharmacy, School of Traditional Medicine, Tehran University of Medical Sciences, Tehran,
Iran
| |
Collapse
|
33
|
Mondelaers SU, Theofanous SA, Florens MV, Perna E, Aguilera-Lizarraga J, Boeckxstaens GE, Wouters MM. Effect of genetic background and postinfectious stress on visceral sensitivity in Citrobacter rodentium-infected mice. Neurogastroenterol Motil 2016; 28:647-58. [PMID: 26728091 DOI: 10.1111/nmo.12759] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/24/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Infectious gastroenteritis is a major risk factor to develop postinfectious irritable bowel syndrome (PI-IBS). It remains unknown why only a subgroup of infected individuals develops PI-IBS. We hypothesize that immunogenetic predisposition is an important risk factor. Hence, we studied the effect of Citrobacter rodentium infection on visceral sensitivity in Th1-predominant C57BL/6 and Th2-predominant Balb/c mice. METHODS Eight-week-old mice were gavaged with C. rodentium, followed by 1 h of water avoidance stress (WAS) at 5 weeks PI. At 10, 14 days, and 5 weeks PI, samples were assessed for histology and inflammatory gene expression by RT-qPCR. Visceral sensitivity was evaluated by visceromotor response recordings (VMR) to colorectal distension. KEY RESULTS Citrobacter rodentium evoked a comparable colonic inflammatory response at 14 days PI characterized by increased crypt length and upregulation of Th1/Th17 cytokine mRNA levels (puncorrected < 0.05) in both C57BL/6 and Balb/c mice. At 5 weeks PI, inflammatory gene mRNA levels returned to baseline in both strains. The VMR was maximal at 14 days PI in C57BL/6 (150 ± 47%; p = 0.02) and Balb/c mice (243 ± 52%; p = 0.03). At 3 weeks PI, the VMR remained increased in Balb/c (176 ± 23%; p = 0.02), but returned to baseline in C57BL/6 mice. At 5 weeks PI, WAS could not re-introduce visceral hypersensitivity (VHS). CONCLUSIONS & INFERENCES Citrobacter rodentium infection induces transient VHS in C57BL/6 and Balb/c mice, which persisted 1 week longer in Balb/c mice. Although other strain-related differences may contribute, a Th2 background may represent a risk factor for prolonged PI-VHS. As PI-VHS is transient, other factors are crucial for persistent VHS development as observed in PI-IBS.
Collapse
Affiliation(s)
- S U Mondelaers
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - S A Theofanous
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - M V Florens
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - E Perna
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - J Aguilera-Lizarraga
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - G E Boeckxstaens
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - M M Wouters
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| |
Collapse
|
34
|
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
|
35
|
Botschuijver S, Yu Z, Welting O, Cailotto C, Kalsbeek A, van den Wijngaard R. Absence of diurnal variation in visceromotor response to colorectal distention in normal Long Evans rats. F1000Res 2016; 5:98. [PMID: 26925229 PMCID: PMC4748828 DOI: 10.12688/f1000research.7238.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2016] [Indexed: 12/29/2022] Open
Abstract
Background: Enhanced colorectal sensitivity (i.e. visceral hypersensitivity) is thought to be a pathophysiological mechanism in irritable bowel syndrome (IBS). In healthy men a circadian variation in rectal perception to colonic distention was described. Disturbed day and night rhythms, which occur in shift work and trans meridian flights, are associated with the prevalence of IBS. This raises the question whether disruptions of circadian control are responsible for the observed pathology in IBS. Prior to investigating altered rhythmicity in relation to visceral hypersensitivity in a rat model for IBS, it is relevant to establish whether normal rats display circadian variation similar to healthy men. Methodology and findings: In rodents colorectal distension leads to reproducible contractions of abdominal musculature. We used quantification of this so called visceromotor response (VMR) by electromyography (EMG) to assess visceral sensitivity in rats. We assessed the VMR in normal male Long Evans rats at different time points of the light/dark cycle. Although a control experiment with male maternal separated rats confirmed that intentionally inflicted (i.e. stress induced) changes in VMR can be detected, normal male Long Evans rats showed no variation in VMR along the light/dark cycle in response to colorectal distension. Conclusions: In the absence of a daily rhythm of colorectal sensitivity in normal control rats it is not possible to investigate possible aberrancies in our rat model for IBS.
Collapse
Affiliation(s)
- Sara Botschuijver
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal research, Academic Medical Center, Amsterdam, Amsterdam, Netherlands
| | - Zhumei Yu
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal research, Academic Medical Center, Amsterdam, Amsterdam, Netherlands.,Department of Neurobiology, Tongji Medical College, HUST, Wuhan, China
| | - Olaf Welting
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal research, Academic Medical Center, Amsterdam, Amsterdam, Netherlands
| | - Cathy Cailotto
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal research, Academic Medical Center, Amsterdam, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Rene van den Wijngaard
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal research, Academic Medical Center, Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
36
|
Greenwood-Van Meerveld B, Prusator DK, Johnson AC. Animal models of gastrointestinal and liver diseases. Animal models of visceral pain: pathophysiology, translational relevance, and challenges. Am J Physiol Gastrointest Liver Physiol 2015; 308:G885-903. [PMID: 25767262 DOI: 10.1152/ajpgi.00463.2014] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/11/2015] [Indexed: 02/08/2023]
Abstract
Visceral pain describes pain emanating from the thoracic, pelvic, or abdominal organs. In contrast to somatic pain, visceral pain is generally vague, poorly localized, and characterized by hypersensitivity to a stimulus such as organ distension. Animal models have played a pivotal role in our understanding of the mechanisms underlying the pathophysiology of visceral pain. This review focuses on animal models of visceral pain and their translational relevance. In addition, the challenges of using animal models to develop novel therapeutic approaches to treat visceral pain will be discussed.
Collapse
Affiliation(s)
- Beverley Greenwood-Van Meerveld
- Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Dawn K Prusator
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Anthony C Johnson
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
37
|
van Diest SA, van den Elsen LWJ, Klok AJ, Welting O, Hilbers FW, van de Heijning BJ, Gaemers IC, Boeckxstaens GE, Werner MF, Willemsen LEM, de Jonge WJ, van den Wijngaard RM. Dietary Marine n-3 PUFAs Do Not Affect Stress-Induced Visceral Hypersensitivity in a Rat Maternal Separation Model. J Nutr 2015; 145:915-22. [PMID: 25716554 DOI: 10.3945/jn.114.201731] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 02/04/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Although never evaluated for efficacy, n-3 (ω-3) long-chain polyunsaturated fatty acids (LCPUFAs) are commercially offered as treatment for irritable bowel syndrome (IBS). OBJECTIVE This study was designed to investigate, in a mast cell-dependent model for visceral hypersensitivity, whether this pathophysiologic mechanism can be reversed by dietary LCPUFA treatment via peroxisome proliferator-activated receptor γ (PPARG) activation. METHODS Maternally separated rats were subjected to hypersensitivity-inducing acute stress at adult age. Reversal was attempted by protocols with tuna oil-supplemented diets [4% soy oil (SO) and 3% tuna oil (SO-T3) or 3% SO and 7% tuna oil (SO-T7)] and compared with control SO diets (7% or 10% SO) 4 wk after stress. The PPARG agonist rosiglitazone was evaluated in a 1 wk preventive protocol (30 mg · kg⁻¹ · d⁻¹). Erythrocytes were assessed to confirm LCPUFA uptake and tissue expression of lipoprotein lipase and glycerol kinase as indicators of PPARG activation. Colonic mast cell degranulation was evaluated by toluidine blue staining. In vitro, human mast cell line 1 (HMC-1) cells were pretreated with rosiglitazone, eicosapentaenoic acid, or docosahexaenoic acid, stimulated with phorbol 12-myristate 13-acetate (PMA) and calcium ionophore or compound 48/80 and evaluated for tumor necrosis factor α (TNF-α) and β-hexosaminidase release. RESULTS Stress led to visceral hypersensitivity in all groups. Hypersensitivity was not reversed by SO-T3 or control treatment [prestress vs. 24 h poststress vs. posttreatment area under the curve; 76 ± 4 vs. 128 ± 12 (P < 0.05) vs. 115 ± 14 and 82 ± 5 vs. 127 ± 16 (P < 0.01) vs. 113 ± 19, respectively]. Comparison of SO-T7 with its control showed similar results [74 ± 6 vs. 103 ± 13 (P < 0.05) vs. 115 ± 17 and 66 ± 3 vs. 103 ± 10 (P < 0.05) vs. 117 ± 11, respectively]. Erythrocytes showed significant LCPUFA uptake in the absence of colonic PPARG activation. Rosiglitazone induced increased PPARG target gene expression, but did not prevent hypersensitivity. Mast cell degranulation never differed between groups. Rosiglitazone and LCPUFAs significantly reduced PMA/calcium ionophore-induced TNF-α release but not degranulation of HMC-1 cells. CONCLUSION Dietary LCPUFAs did not reverse stress-induced visceral hypersensitivity in maternally separated rats. Although further research is needed, claims concerning LCPUFAs as a treatment option in IBS cannot be confirmed at this point and should be regarded with caution.
Collapse
Affiliation(s)
- Sophie A van Diest
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Lieke W J van den Elsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Allison J Klok
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Olaf Welting
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Francisca W Hilbers
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Ingrid C Gaemers
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Guy E Boeckxstaens
- Division of Gastroenterology, University Hospital Gasthuisberg, University of Leuven, Leuven, Belgium; and
| | - Maria F Werner
- Department of Pharmacology, Biological Sciences Section, Federal University of Paraná, Curitiba, Brazil
| | - Linette E M Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - René M van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands;
| |
Collapse
|
38
|
van den Wijngaard RM, Stanisor OI, van Diest SA, Welting O, Wouters MM, Cailotto C, de Jonge WJ, Boeckxstaens GE. Susceptibility to stress induced visceral hypersensitivity in maternally separated rats is transferred across generations. Neurogastroenterol Motil 2013; 25:e780-90. [PMID: 23965154 DOI: 10.1111/nmo.12202] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/16/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND In irritable bowel syndrome (IBS), familial clustering and transfer across generations may largely depend on environmental factors but this is difficult to establish in the human setting. Therefore, we aimed to set up a relevant animal model. We investigated whether susceptibility to stress induced visceral hypersensitivity in maternally separated (MS) Long Evans rats can be transferred across generations without further separation protocols and, if so, whether this depends on maternal care. METHODS At adult age, we evaluated pre- vs post water avoidance (WA) changes in visceromotor response to distension in non-handled second filial generation offspring (NH-F2) of previously separated MS-F1 dams. Furthermore, the role of maternal care was evaluated by cross-fostering F2 offspring of NH-F1 and MS-F1 dams and subsequent sensitivity measurements at adult age. Involvement of mast cells in post stress hypersensitivity of NH-F2 rats was evaluated by mast cell stabilization. KEY RESULTS In adult NH-F2 offspring of MS-F1 dams, post-WA hypersensitivity to colorectal distension was observed in 80% of rats compared with 19% in offspring of NH-F1 dams. Cross-fostered pups adapted to the phenotype of the foster mother: pups of NH-F1 dams nursed by MS-F1 dams showed post-WA hypersensitivity to distension at adult age and vice versa (100% and 20% respectively). In NH-F2 rats, post-WA hypersensitivity was reversed by mast cell stabilizer doxantrazole. CONCLUSIONS & INFERENCES Maternal separated-induced susceptibility to stress-triggered visceral hypersensitivity is transferred across generations and this transfer depends on maternal care. Thus, MS is a suitable model to evaluate environmental triggers relevant to IBS clustering in families.
Collapse
Affiliation(s)
- R M van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|