1
|
Davis KL, Claudio-Etienne E, Frischmeyer-Guerrerio PA. Atopic dermatitis and food allergy: More than sensitization. Mucosal Immunol 2024; 17:1128-1140. [PMID: 38906220 PMCID: PMC11471387 DOI: 10.1016/j.mucimm.2024.06.005] [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: 03/27/2024] [Revised: 06/01/2024] [Accepted: 06/13/2024] [Indexed: 06/23/2024]
Abstract
The increased risk of food allergy in infants with atopic dermatitis (AD) has long been recognized; an epidemiologic phenomenon termed "the atopic march." Current literature supports the hypothesis that food antigen exposure through the disrupted skin barrier in AD leads to food antigen-specific immunoglobulin E production and food sensitization. However, there is growing evidence that inflammation in the skin drives intestinal remodeling via circulating inflammatory signals, microbiome alterations, metabolites, and the nervous system. We explore how this skin-gut axis helps to explain the link between AD and food allergy beyond sensitization.
Collapse
Affiliation(s)
- Katelin L Davis
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Comparative Biomedical Scientist Training Program, The Molecular Pathology Unit, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, The National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Comparative Pathobiology Department, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Estefania Claudio-Etienne
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela A Frischmeyer-Guerrerio
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Theoharides TC, Twahir A, Kempuraj D. Mast cells in the autonomic nervous system and potential role in disorders with dysautonomia and neuroinflammation. Ann Allergy Asthma Immunol 2024; 132:440-454. [PMID: 37951572 DOI: 10.1016/j.anai.2023.10.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/16/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023]
Abstract
Mast cells (MC) are ubiquitous in the body, and they are critical for not only in allergic diseases but also in immunity and inflammation, including having potential involvement in the pathophysiology of dysautonomias and neuroinflammatory disorders. MC are located perivascularly close to nerve endings and sites such as the carotid bodies, heart, hypothalamus, the pineal gland, and the adrenal gland that would allow them not only to regulate but also to be affected by the autonomic nervous system (ANS). MC are stimulated not only by allergens but also many other triggers including some from the ANS that can affect MC release of neurosensitizing, proinflammatory, and vasoactive mediators. Hence, MC may be able to regulate homeostatic functions that seem to be dysfunctional in many conditions, such as postural orthostatic tachycardia syndrome, autism spectrum disorder, myalgic encephalomyelitis/chronic fatigue syndrome, and Long-COVID syndrome. The evidence indicates that there is a possible association between these conditions and diseases associated with MC activation. There is no effective treatment for any form of these conditions other than minimizing symptoms. Given the many ways MC could be activated and the numerous mediators released, it would be important to develop ways to inhibit stimulation of MC and the release of ANS-relevant mediators.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida; Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts.
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| |
Collapse
|
3
|
Wen X, Dong H, Zou W. The role of gut microorganisms and metabolites in intracerebral hemorrhagic stroke: a comprehensive review. Front Neurosci 2024; 18:1346184. [PMID: 38449739 PMCID: PMC10915040 DOI: 10.3389/fnins.2024.1346184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
Intracerebral hemorrhagic stroke, characterized by acute hemorrhage in the brain, has a significant clinical prevalence and poses a substantial threat to individuals' well-being and productivity. Recent research has elucidated the role of gut microorganisms and their metabolites in influencing brain function through the microbiota-gut-brain axis (MGBA). This article provides a comprehensive review of the current literature on the common metabolites, short-chain fatty acids (SCFAs) and trimethylamine-N-oxide (TMAO), produced by gut microbiota. These metabolites have demonstrated the potential to traverse the blood-brain barrier (BBB) and directly impact brain tissue. Additionally, these compounds have the potential to modulate the parasympathetic nervous system, thereby facilitating the release of pertinent substances, impeding the buildup of inflammatory agents within the brain, and manifesting anti-inflammatory properties. Furthermore, this scholarly analysis delves into the existing dearth of investigations concerning the influence of gut microorganisms and their metabolites on cerebral functions, while also highlighting prospective avenues for future research.
Collapse
Affiliation(s)
- Xin Wen
- The First Clinical Medical College, Heilongjiang University Of Chinese Medicine, Harbin, China
| | - Hao Dong
- The First Clinical Medical College, Heilongjiang University Of Chinese Medicine, Harbin, China
| | - Wei Zou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Qiu P, Li D, Xiao C, Xu F, Chen X, Chang Y, Liu L, Zhang L, Zhao Q, Chen Y. The Eph/ephrin system symphony of gut inflammation. Pharmacol Res 2023; 197:106976. [PMID: 38032293 DOI: 10.1016/j.phrs.2023.106976] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
The extent of gut inflammation depends largely on the gut barrier's integrity and enteric neuroimmune interactions. However, the factors and molecular mechanisms that regulate inflammation-related changes in the enteric nervous system (ENS) remain largely unexplored. Eph/ephrin signaling is critical for inflammatory response, neuronal activation, and synaptic plasticity in the brain, but its presence and function in the ENS have been largely unknown to date. This review discusses the critical role of Eph/ephrin in regulating gut homeostasis, inflammation, neuroimmune interactions, and pain pathways. Targeting the Eph/ephrin system offers innovative treatments for gut inflammation disorders, offering hope for enhanced patient prognosis, pain management, and overall quality of life.
Collapse
Affiliation(s)
- Peishan Qiu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Daojiang Li
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Cong Xiao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Fei Xu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Xiaoyu Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China.
| | - Yuhua Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan 430071, China.
| |
Collapse
|
5
|
Brandlhuber M, Benhaqi P, Brandlhuber B, Koliogiannis V, Kasparek MS, Mueller MH, Kreis ME. The role of vagal innervation on the early development of postoperative ileus in mice. Neurogastroenterol Motil 2022; 34:e14308. [PMID: 34962331 DOI: 10.1111/nmo.14308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/30/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Postoperative ileus (POI) involves an intestinal inflammatory response that is modulated by afferent and efferent vagal activation. We aimed to identify the potential influence of the vagus nerve on POI by tracking central vagal activation and its role for peripheral inflammatory changes during the early hours after surgery. METHODS C57BL6 mice were vagotomized (V) 3-4 days prior to experiments, while control animals received sham vagotomy (SV). Subgroups underwent either laparotomy (sham operation; S-POI) or laparotomy followed by standardized small bowel manipulation to induce postoperative ileus (POI). Three hours and 9 h later, respectively, a jejunal segment was harvested and infiltration of inflammatory cells in intestinal muscularis was evaluated by fluorescein isothiocyanate (FITC) avidin and myeloperoxidase (MPO) staining. Moreover, the brain stem was harvested, and central nervous activation was investigated by Fos immunochemistry in both the nucleus of the solitary tract (NTS) and the area postrema (AP). Data are presented as mean ± SEM, and a p < 0.05 was considered statistically significant. KEY RESULTS Three hour experiments revealed no significant differences between all experimental groups, except MPO staining: 3 h after abdominal surgery, there were significantly more MPO-positive cells in vagotomized S-POI animals compared to sham-vagotomized S-POI animals (26.7 ± 7.1 vs. 5.1 ± 2.4, p < 0.01). Nine hour postoperatively intramuscular mast cells (IMMC) were significantly decreased in the intestinal muscularis of V/POI animals compared to SV/POI animals (1.5 ± 0.3 vs. 5.9 ± 0.2, p < 0.05), while MPO-positive cells were increased in V/POI animals compared to SV/POI animals (713.2 ± 99.4 vs. 46.9 ± 5.8, p < 0.05). There were less Fos-positive cells in the NTS of V/POI animals compared to SV/POI animals (64.7 ± 7.8 vs. 132.8 ± 23.9, p < 0.05) and more Fos-positive cells in the AP of V/POI animals compared to SV/POI animals 9 h postoperatively (38.0 ± 2.0 vs. 13.7 ± 0.9, p < 0.001). CONCLUSIONS AND INTERFERENCES Afferent nerve signaling to the central nervous system during the development of early POI seems to be mediated mainly via the vagus nerve and to a lesser degree via systemic circulation. During the early hours of POI, the intestinal immune response may be attenuated by vagal modulation, suggesting interactions between the central nervous system and the intestine.
Collapse
Affiliation(s)
- Martina Brandlhuber
- Department of Radiology, Grosshadern Clinic, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Petra Benhaqi
- Center for Human Genetics and Laboratory Diagnostics, Medical Labs Martinsried, Martinsried, Germany
| | | | - Vanessa Koliogiannis
- Department of Radiology, Grosshadern Clinic, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Mario H Mueller
- Department of Surgery and Minimal-Invasive Surgery, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Martin E Kreis
- Department of General and Visceral Surgery, Charité University Medicine Berlin, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
6
|
Duan H, Cai X, Luan Y, Yang S, Yang J, Dong H, Zeng H, Shao L. Regulation of the Autonomic Nervous System on Intestine. Front Physiol 2021; 12:700129. [PMID: 34335306 PMCID: PMC8317205 DOI: 10.3389/fphys.2021.700129] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Intestine is composed of various types of cells including absorptive epithelial cells, goblet cells, endocrine cells, Paneth cells, immunological cells, and so on, which play digestion, absorption, neuroendocrine, immunological function. Intestine is innervated with extrinsic autonomic nerves and intrinsic enteric nerves. The neurotransmitters and counterpart receptors are widely distributed in the different intestinal cells. Intestinal autonomic nerve system includes sympathetic and parasympathetic nervous systems, which regulate cellular proliferation and function in intestine under physiological and pathophysiological conditions. Presently, distribution and functional characteristics of autonomic nervous system in intestine were reviewed. How autonomic nervous system regulates intestinal cell proliferation was discussed. Function of autonomic nervous system on intestinal diseases was extensively reviewed. It might be helpful to properly manipulate autonomic nervous system during treating different intestinal diseases.
Collapse
Affiliation(s)
- Hongyi Duan
- Medical College of Nanchang University, Nanchang, China
| | - Xueqin Cai
- Medical College of Nanchang University, Nanchang, China
| | - Yingying Luan
- Medical College of Nanchang University, Nanchang, China
| | - Shuo Yang
- Medical College of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, China
| | - Juan Yang
- Medical College of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, China
| | - Hui Dong
- Medical College of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, China
| | - Huihong Zeng
- Medical College of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, China
| | - Lijian Shao
- Medical College of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Cordner ZA, Li Q, Liu L, Tamashiro KL, Bhargava A, Moran TH, Pasricha PJ. Vagal gut-brain signaling mediates amygdaloid plasticity, affect, and pain in a functional dyspepsia model. JCI Insight 2021; 6:144046. [PMID: 33591956 PMCID: PMC8026195 DOI: 10.1172/jci.insight.144046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Functional dyspepsia (FD) is associated with chronic gastrointestinal distress and with anxiety and depression. Here, we hypothesized that aberrant gastric signals, transmitted by the vagus nerve, may alter key brain regions modulating affective and pain behavior. Using a previously validated rat model of FD characterized by gastric hypersensitivity, depression-like behavior, and anxiety-like behavior, we found that vagal activity - in response to gastric distention - was increased in FD rats. The FD phenotype was associated with gastric mast cell hyperplasia and increased expression of corticotrophin-releasing factor (Crh) and decreased brain-derived neurotrophic factor genes in the central amygdala. Subdiaphragmatic vagotomy reversed these changes and restored affective behavior to that of controls. Vagotomy partially attenuated pain responses to gastric distention, which may be mediated by central reflexes in the periaqueductal gray, as determined by local injection of lidocaine. Ketotifen, a mast cell stabilizer, reduced vagal hypersensitivity, normalized affective behavior, and attenuated gastric hyperalgesia. In conclusion, vagal activity, partially driven by gastric mast cells, induces long-lasting changes in Crh signaling in the amygdala that may be responsible for enhanced pain and enhanced anxiety- and depression-like behaviors. Together, these results support a "bottom-up" pathway involving the gut-brain axis in the pathogenesis of both gastric pain and psychiatric comorbidity in FD.
Collapse
Affiliation(s)
| | - Qian Li
- Center for Neurogastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liansheng Liu
- Center for Neurogastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Aditi Bhargava
- Department of Obstetrics and Gynecology and The Center for Reproductive Sciences, UCSF, San Francisco, California, USA
| | | | - Pankaj Jay Pasricha
- Center for Neurogastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Sun DL, Qi YX, Yang T, Lin YY, Li SM, Li YJ, Xu QW, Sun YB, Li WM, Chen XZ, Xu PY. Early oral nutrition improves postoperative ileus through the TRPA1/CCK1-R-mediated mast cell-nerve axis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:179. [PMID: 32309326 PMCID: PMC7154392 DOI: 10.21037/atm.2020.01.95] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background The mechanism of early oral nutrition that regulates the mast cell-nerve axis to improve postoperative ileus (POI) remains unclear. This study aims to investigate whether early oral nutrition can improve POI through Transient receptor potential ankyrin-1 (TRPA1)/cholecystokinin 1 receptor (CCK1-R) in the mast cell-nerve axis. Methods Experiment 1: Male Sprague-Dawley (SD) rats were randomly divided into the TRPA1 inhibitor + oral nutrition group (TI + ON + POI), oral nutrition group (ON + POI), POI group (POI) and sham surgery group (Sham). Nine rats in each group were treated. Experiment 2: Primary cultures of mast cells and dorsal root ganglion cells were created, and a non-contact co-culture system was established. The cells were divided into the dorsal root ganglion (DRG) group, mast cell group, DRG + mast cell group, TRPA1 inhibitor or enhancer group, mast cell stabilizer or enhancer group, CCK1-R inhibitor or enhancer group. The results of expression of TRPA1, CCK1-R and histamine in colon tissue, portal vein blood, supernatant or dorsal root ganglia, intestinal transport test and mast cell morphology were analysed. Results In experiment 1, Early oral nutrition could alleviate the degranulation and activation of mast cells and alleviate the inflammatory reaction of intestinal wall muscles (P<0.05). Early oral nutrition improved POI by stabilizing mast cells with TRPA1. TRPA1 inhibitor decreased CCK1-R concentrations in portal vein blood and CCK1-R expression in colonic smooth muscle (P<0.05). In experiment 2, the change in mast cell function regulated the secretion of CCK1-R by neurons, CCK1-R negatively regulated the degranulation and activation of mast cells (P<0.05), and mast cells positively regulated the expression of TRPA1 protein in DRG (P<0.05). Conclusions Early enteral nutrition can improve POI through the TRPA1/CCK1-R-mediated mast cell-nerve axis. TRPA1 positively regulates CCK1-R to stabilize mast cells, but TRPA1 is not the target of the downstream CCK1-R pathway.
Collapse
Affiliation(s)
- Da-Li Sun
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Yu-Xing Qi
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Ting Yang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Yue-Ying Lin
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Shu-Min Li
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Yi-Jun Li
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Qing-Wen Xu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Yan-Bo Sun
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Wei-Ming Li
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Xiong-Zhi Chen
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| | - Peng-Yuan Xu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China.,Yunnan Research Center for Surgical Clinical Nutrition, Kunming 650101, China
| |
Collapse
|
9
|
Misto A, Provensi G, Vozella V, Passani MB, Piomelli D. Mast Cell-Derived Histamine Regulates Liver Ketogenesis via Oleoylethanolamide Signaling. Cell Metab 2019; 29:91-102.e5. [PMID: 30318340 DOI: 10.1016/j.cmet.2018.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 06/11/2018] [Accepted: 09/12/2018] [Indexed: 01/30/2023]
Abstract
The conversion of lipolysis-derived fatty acids into ketone bodies (ketogenesis) is a crucial metabolic adaptation to prolonged periods of food scarcity. The process occurs primarily in liver mitochondria and is initiated by fatty-acid-mediated stimulation of the ligand-operated transcription factor, peroxisome proliferator-activated receptor-α (PPAR-α). Here, we present evidence that mast cells contribute to the control of fasting-induced ketogenesis via a paracrine mechanism that involves secretion of histamine into the hepatic portal circulation, stimulation of liver H1 receptors, and local biosynthesis of the high-affinity PPAR-α agonist, oleoylethanolamide (OEA). Genetic or pharmacological interventions that disable any one of these events, including mast cell elimination, deletion of histamine- or OEA-synthesizing enzymes, and H1 blockade, blunt ketogenesis without affecting lipolysis. The results reveal an unexpected role for mast cells in the regulation of systemic fatty-acid homeostasis, and suggest that OEA may act in concert with lipolysis-derived fatty acids to activate liver PPAR-α and promote ketogenesis.
Collapse
Affiliation(s)
- Alessandra Misto
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genoa 16163, Italy; School of Advanced Studies Sant'Anna, Pisa 56127, Italy
| | - Gustavo Provensi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy
| | - Valentina Vozella
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | | | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, Biological Chemistry and Pharmacology, School of Medicine, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
10
|
Fornai M, van den Wijngaard RM, Antonioli L, Pellegrini C, Blandizzi C, de Jonge WJ. Neuronal regulation of intestinal immune functions in health and disease. Neurogastroenterol Motil 2018; 30:e13406. [PMID: 30058092 DOI: 10.1111/nmo.13406] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Nerve-mucosa interactions control various elements of gastrointestinal functions, including mucosal host defense, gut barrier function, and epithelial cell growth and differentiation. In both intestinal and extra-intestinal diseases, alterations of autonomic nerve activity have been observed to be concurrent with the disease course, such as in inflammatory and functional bowel diseases, and neurodegenerative diseases. This is relevant as the extrinsic autonomic nervous system is increasingly recognized to modulate gut inflammatory responses. The molecular and cellular mechanisms through which the extrinsic and intrinsic nerve pathways may regulate digestive mucosal functions have been investigated in several pre-clinical and clinical studies. PURPOSE The present review focuses on the involvement of neural pathways in gastrointestinal disease, and addresses the current strategies to intervene with neuronal pathway as a means of treatment.
Collapse
Affiliation(s)
- M Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - R M van den Wijngaard
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - L Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - C Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - C Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - W J de Jonge
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Cartolano MC, Amador MHB, Tzaneva V, Milsom WK, McDonald MD. Extrinsic nerves are not involved in branchial 5-HT dynamics or pulsatile urea excretion in Gulf toadfish, Opsanus beta. Comp Biochem Physiol A Mol Integr Physiol 2017; 214:58-65. [PMID: 28887162 DOI: 10.1016/j.cbpa.2017.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 10/18/2022]
Abstract
Gulf toadfish (Opsanus beta) can switch from continuously excreting ammonia as their primary nitrogenous waste to excreting predominantly urea in distinct pulses. Previous studies have shown that the neurotransmitter serotonin (5-HT) is involved in controlling this process, but it is unknown if 5-HT availability is under central nervous control or if the 5-HT signal originates from a peripheral source. Following up on a previous study, cranial nerves IX (glossopharyngeal) and X (vagus) were sectioned to further characterize their role in controlling pulsatile urea excretion and 5-HT release within the gill. In contrast to an earlier study, nerve sectioning did not result in a change in urea pulse frequency. Total urea excretion, average pulse size, total nitrogen excretion, and percent ureotely were reduced the first day post-surgery in nerve-sectioned fish but recovered by 72h post-surgery. Nerve sectioning also had no effect on toadfish urea transporter (tUT), 5-HT transporter (SERT), or 5-HT2A receptor mRNA expression or 5-HT and 5-hydroxyindoleacetic acid (5-HIAA) abundance in the gill, all of which were found consistently across the three gill arches except 5-HIAA, which was undetectable in the first gill arch. Our findings indicate that the central nervous system does not directly control pulsatile urea excretion or local changes in gill 5-HT and 5-HIAA abundance.
Collapse
Affiliation(s)
- Maria C Cartolano
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, USA.
| | - Molly H B Amador
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, USA
| | - Velislava Tzaneva
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - William K Milsom
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - M Danielle McDonald
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, USA
| |
Collapse
|
12
|
Mackenzie AE, Milligan G. The emerging pharmacology and function of GPR35 in the nervous system. Neuropharmacology 2017; 113:661-671. [DOI: 10.1016/j.neuropharm.2015.07.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/20/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
|
13
|
Rizzi A, Crivellato E, Benagiano V, Ribatti D. Mast cells in human digestive tube in normal and pathological conditions. Immunol Lett 2016; 177:16-21. [DOI: 10.1016/j.imlet.2016.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/20/2016] [Accepted: 07/04/2016] [Indexed: 01/15/2023]
|
14
|
de Jonge WJ. Neuronal Regulation of Mucosal Immune Responses. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00046-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Abstract
Often considered as the archetype of neuroimmune communication, much of our understanding of the bidirectional relationship between the nervous and immune systems has come from the study of mast cell-nerve interaction. Mast cells play a role in resistance to infection and are extensively involved in inflammation and subsequent tissue repair. Thus, the relationship between mast cells and neurons enables the involvement of peripheral and central nervous systems in the regulation of host defense mechanisms and inflammation. Recently, with the identification of the cholinergic anti-inflammatory pathway, there has been increased interest in the role of the parasympathetic nervous system in regulating immune responses. Classical neurotransmitters and neuropeptides released from cholinergic and inhibitory NANC neurons can modulate mast cell activity, and there is good evidence for the existence of parasympathetic nerve-mast cell functional units in the skin, lung, and intestine that have the potential to regulate a range of physiological processes.
Collapse
Affiliation(s)
- Paul Forsythe
- Department of Medicine, The Brain-Body Institute, St. Joseph's Healthcare, McMaster University, 50 Charlton Avenue East, T3302, Hamilton, ON, Canada, L8N 4A6,
| |
Collapse
|
16
|
Bell A, Althaus M, Diener M. Communication between mast cells and rat submucosal neurons. Pflugers Arch 2014; 467:1809-23. [DOI: 10.1007/s00424-014-1609-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 11/29/2022]
|
17
|
Yan XJ, Feng CC, Liu Q, Zhang LY, Dong X, Liu ZL, Cao ZJ, Mo JZ, Li Y, Fang JY, Chen SL. Vagal Afferents Mediate Antinociception of Estrogen in a Rat Model of Visceral Pain: The Involvement of Intestinal Mucosal Mast Cells and 5-Hydroxytryptamine 3 Signaling. THE JOURNAL OF PAIN 2014; 15:204-17. [DOI: 10.1016/j.jpain.2013.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 09/14/2013] [Accepted: 10/31/2013] [Indexed: 12/19/2022]
|
18
|
de Haan JJ, Hadfoune M, Lubbers T, Hodin C, Lenaerts K, Ito A, Verbaeys I, Skynner MJ, Cailotto C, van der Vliet J, de Jonge WJ, Greve JWM, Buurman WA. Lipid-rich enteral nutrition regulates mucosal mast cell activation via the vagal anti-inflammatory reflex. Am J Physiol Gastrointest Liver Physiol 2013; 305:G383-91. [PMID: 23812038 DOI: 10.1152/ajpgi.00333.2012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nutritional stimulation of the cholecystokinin-1 receptor (CCK-1R) and nicotinic acetylcholine receptor (nAChR)-mediated vagal reflex was shown to reduce inflammation and preserve intestinal integrity. Mast cells are important early effectors of the innate immune response; therefore modulation of mucosal mast cells is a potential therapeutic target to control the acute inflammatory response in the intestine. The present study investigates intestinal mast cell responsiveness upon nutritional activation of the vagal anti-inflammatory reflex during acute inflammation. Mucosal mast cell degranulation was induced in C57/Bl6 mice by administration of Salmonella enterica LPS. Lipid-rich enteral feeding prior to LPS significantly decreased circulatory levels of mouse mast cell protease at 30 min post-LPS compared with isocaloric low-lipid nutrition or fasting. CCK-1R blockage reversed the inhibitory effects of lipid-rich feeding, whereas stimulation of the peripheral CCK-1R mimicked nutritional mast cell inhibition. The effects of lipid-rich nutrition were negated by nAChR blockers chlorisondamine and α-bungarotoxin and vagal intestinal denervation. Accordingly, release of β-hexosaminidase by MC/9 mast cells following LPS or IgE-ovalbumin complexes was dose dependently inhibited by acetylcholine and nicotine. Application of GSK1345038A, a specific agonist of the nAChR α7, in bone marrow-derived mast cells from nAChR β2-/- and wild types indicated that cholinergic inhibition of mast cells is mediated by the nAChR α7 and is independent of the nAChR β2. Together, the present study reveals mucosal mast cells as a previously unknown target of the nutritional anti-inflammatory vagal reflex.
Collapse
Affiliation(s)
- Jacco J de Haan
- Dept. of Surgery at Maastricht Univ. Medical Centre+, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bombardi C, Grandis A, Gardini A, Sorteni C, Clavenzani P, Chiocchetti R. Expression of β2 adrenoceptors within enteric neurons of the horse ileum. Res Vet Sci 2013; 95:837-45. [PMID: 23941962 DOI: 10.1016/j.rvsc.2013.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 05/10/2013] [Accepted: 07/08/2013] [Indexed: 12/31/2022]
Abstract
The activity of the gastrointestinal tract is regulated through the activation of adrenergic receptors (ARs). Since data concerning the distribution of ARs in the horse intestine is virtually absent, we investigated the distribution of β2-AR in the horse ileum using double-immunofluorescence. The β2-AR-immunoreactivity (IR) was observed in most (95%) neurons located in submucosal plexus (SMP) and in few (8%) neurons of the myenteric plexus (MP). Tyrosine hydroxylase (TH)-IR fibers were observed close to neurons expressing β2-AR-IR. Since β2-AR is virtually expressed in most neurons located in the horse SMP and in a lower percentage of neurons in the MP, it is reasonable to retain that this adrenergic receptor could regulate the activity of both secretomotor neurons and motor neurons innervating muscle layers and blood vessels. The high density of TH-IR fibers near β2-AR-IR enteric neurons indicates that the excitability of these cells could be directly modulated by the sympathetic system.
Collapse
Affiliation(s)
- Cristiano Bombardi
- Department of Veterinary Medical Science, University of Bologna, 40064 Ozzano dell'Emilia, Bologna, Italy.
| | | | | | | | | | | |
Collapse
|
20
|
The Gut's Little Brain in Control of Intestinal Immunity. ISRN GASTROENTEROLOGY 2013; 2013:630159. [PMID: 23691339 PMCID: PMC3649343 DOI: 10.1155/2013/630159] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 01/12/2013] [Indexed: 12/13/2022]
Abstract
The gut immune system shares many mediators and receptors with the autonomic nervous system. Good examples thereof are the parasympathetic (vagal) and sympathetic neurotransmitters, for which many immune cell types in a gut context express receptors or enzymes required for their synthesis. For some of these the relevance for immune regulation has been recently defined. Earlier and more recent studies in neuroscience and immunology have indicated the anatomical and cellular basis for bidirectional interactions between the nervous and immune systems. Sympathetic immune modulation is well described earlier, and in the last decade the parasympathetic vagal nerve has been put forward as an integral part of an immune regulation network via its release of Ach, a system coined "the cholinergic anti-inflammatory reflex." A prototypical example is the inflammatory reflex, comprised of an afferent arm that senses inflammation and an efferent arm: the cholinergic anti-inflammatory pathway, that inhibits innate immune responses. In this paper, the current understanding of how innate mucosal immunity can be influenced by the neuronal system is summarized, and cell types and receptors involved in this interaction will be highlighted. Focus will be given on the direct neuronal regulatory mechanisms, as well as current advances regarding the role of microbes in modulating communication in the gut-brain axis.
Collapse
|
21
|
Furuya S, Furuya K. Roles of substance P and ATP in the subepithelial fibroblasts of rat intestinal villi. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 304:133-89. [PMID: 23809436 DOI: 10.1016/b978-0-12-407696-9.00003-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ingestion of food and water induces chemical and mechanical signals that trigger peristaltic reflexes and also villous movement in the gut. In the intestinal villi, subepithelial fibroblasts under the epithelium form contractile cellular networks and closely contact to the varicosities of substance P and nonsubstance P afferent neurons. Subepithelial fibroblasts of the duodenal villi possess purinergic receptor P2Y1 and tachykinin receptor NK1. ATP and substance P induce increase in intracellular Ca(2+) and cell contraction in subepithelial fibroblasts. They are highly mechanosensitive and release ATP by mechanical stimuli. Released ATP spreads to form an ATP "cloud" with nearly 1μM concentration and activates the surroundings via P2Y1 and afferent neurons via P2X receptors. These findings suggest that villous subepithelial fibroblasts and afferent neurons interact via ATP and substance P. This mutual interaction may play important roles in the signal transduction of mechano reflex pathways including a coordinate villous movement and also in the maturation of the structure and function of the intestinal villi.
Collapse
Affiliation(s)
- Sonoko Furuya
- Section of Brain Structure Information, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan.
| | | |
Collapse
|
22
|
Vagal nerve stimulation modulates gut injury and lung permeability in trauma-hemorrhagic shock. J Trauma Acute Care Surg 2012; 73:338-42; discussion 342. [PMID: 22846937 DOI: 10.1097/ta.0b013e31825debd3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Hemorrhagic shock is known to disrupt the gut barrier leading to end-organ dysfunction. The vagus nerve can inhibit detrimental immune responses that contribute to organ damage in hemorrhagic shock. Therefore, we explored whether stimulation of the vagus nerve can protect the gut and recover lung permeability in trauma-hemorrhagic shock (THS). METHODS Male Sprague-Dawley rats were subjected to left cervical vagus nerve stimulation at 5 V for 10 minutes. The right internal jugular and femoral artery were cannulated for blood withdrawal and blood pressure monitoring, respectively. Animals were then subjected to hemorrhagic shock to a mean arterial pressure between 30 mm Hg and 35 mm Hg for 90 minutes then reperfused with their own whole blood. After observation for 3 hours, gut permeability was assessed with fluorescein dextran 4 in vivo injections in a ligated portion of distal ileum followed by Evans blue dye injection to assess lung permeability. Pulmonary myeloperoxidase levels were measured and compared. RESULTS Vagal nerve stimulation abrogated THS-induced lung injury (mean [SD], 8.46 [0.36] vs. 4.87 [0.78]; p < 0.05) and neutrophil sequestration (19.39 [1.01] vs. 12.83 [1.16]; p < 0.05). Likewise, THS gut permeability was reduced to sham levels. CONCLUSION Neuromodulation decreases injury in the THS model as evidenced by decreased gut permeability as well as decreased lung permeability and pulmonary neutrophil sequestration in a rat model.
Collapse
|
23
|
Duncker SC, Philippe D, Martin-Paschoud C, Moser M, Mercenier A, Nutten S. Nigella sativa (black cumin) seed extract alleviates symptoms of allergic diarrhea in mice, involving opioid receptors. PLoS One 2012; 7:e39841. [PMID: 22768141 PMCID: PMC3387213 DOI: 10.1371/journal.pone.0039841] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 05/27/2012] [Indexed: 12/12/2022] Open
Abstract
The incidence of food hypersensitivity and food allergies is on the rise and new treatment approaches are needed. We investigated whether N. sativa, one of its components, thymoquinone, or synthetic opioid receptor (OR)-agonists can alleviate food allergy. Hence, ovalbumin (OVA)-sensitized BALB/c-mice were pre-treated either with a hexanic N. sativa seed extract, thymoquinone, kappa-(U50'4889) or mu-OR-agonists (DAMGO) and subsequently challenged intra-gastrically with OVA. All 4 treatments significantly decreased clinical scores of OVA-induced diarrhea. N. sativa seed extract, thymoquinone, and U50'488 also decreased intestinal mast cell numbers and plasma mouse mast cell protease-1 (MMCP-1). DAMGO, in contrast, had no effect on mast cell parameters but decreased IFNγ, IL-4, IL-5, and IL-10 concentration after ex vivo re-stimulation of mesenteric lymphocytes. The effects on allergy symptoms were reversible by OR-antagonist pre-treatment, whereas most of the effects on immunological parameter were not. We demonstrate that N. sativa seed extract significantly improves symptoms and immune parameters in murine OVA-induced allergic diarrhea; this effect is at least partially mediated by thymoquinone. ORs may also be involved and could be a new target for intestinal allergy symptom alleviation. N. sativa seed extract seems to be a promising candidate for nutritional interventions in humans with food allergy.
Collapse
MESH Headings
- Animals
- Benzoquinones/pharmacology
- Benzoquinones/therapeutic use
- Biomarkers/metabolism
- Chymases/metabolism
- Diarrhea/complications
- Diarrhea/drug therapy
- Diarrhea/immunology
- Food Hypersensitivity/complications
- Food Hypersensitivity/drug therapy
- Food Hypersensitivity/immunology
- Ligands
- Male
- Mice
- Mice, Inbred BALB C
- Nigella sativa/chemistry
- Ovalbumin/immunology
- Phytotherapy
- Plant Extracts/pharmacology
- Plant Extracts/therapeutic use
- Receptors, Opioid/agonists
- Receptors, Opioid/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Seeds/chemistry
Collapse
Affiliation(s)
- Swantje C Duncker
- Nestlé Research Center, Nestec Ltd., Vers-chez-les-Blancs, Lausanne, Switzerland.
| | | | | | | | | | | |
Collapse
|
24
|
McDonald MD, Gilmour KM, Walsh PJ. New insights into the mechanisms controlling urea excretion in fish gills. Respir Physiol Neurobiol 2012; 184:241-8. [PMID: 22684040 DOI: 10.1016/j.resp.2012.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/31/2012] [Accepted: 06/01/2012] [Indexed: 12/28/2022]
Abstract
Not long ago, urea was believed to freely diffuse across plasma membranes. The discovery of specialized proteins to facilitate the movement of urea across the fish gill, similar to those found in mammalian kidney, was exciting, and at the same time, perplexing; especially considering the fact that, aside from elasmobranchs, most fish do not produce urea as their primary nitrogenous waste. Increasingly, it has become apparent that many fish do indeed produce at least a small amount of urea through various processes and continued work on branchial urea transporters in teleost and elasmobranch fishes has led to recent advances in the regulation of these mechanisms. The following review outlines the substantial progress that has been made towards understanding environmental and developmental impacts on fish gill urea transport. This review also outlines the work that has been done regarding endocrine and neural control of urea excretion, most of which has been collected from only a handful of teleost fish. It is evident that more research is needed to establish the endocrine and neural control of urea excretion in fish, including fish representative of more ancient lineages (hagfish and lamprey), and elasmobranch fish.
Collapse
Affiliation(s)
- M Danielle McDonald
- Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL 33149, USA.
| | | | | |
Collapse
|
25
|
Dhawan S, Cailotto C, Harthoorn LF, de Jonge WJ. Cholinergic signalling in gut immunity. Life Sci 2012; 91:1038-42. [PMID: 22580288 DOI: 10.1016/j.lfs.2012.04.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/17/2012] [Accepted: 04/24/2012] [Indexed: 12/20/2022]
Abstract
The gut immune system shares many signalling molecules and receptors with the autonomic nervous system. A good example is the vagal neurotransmitter acetylcholine (ACh), for which many immune cell types express cholinergic receptors (AChR). In the last decade the vagal nerve has emerged as an integral part of an immune regulation network via its release of ACh; a system coined "the cholinergic anti-inflammatory reflex". The perspective of cholinergic immune regulation in the gut mucosa has been widened by the recent discovery of populations of ACh producing immune cells in the spleen and other organs. As such, ACh, classically referred to as neurotransmitter, may serve a much broader function as bi-directional signalling molecule between neurons and non-neuronal cell types of the immune system.
Collapse
Affiliation(s)
- Shobhit Dhawan
- Tytgat Institute for Liver and GI research, AMC Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
26
|
Mast cell–nerve axis with a focus on the human gut. Biochim Biophys Acta Mol Basis Dis 2012; 1822:85-92. [DOI: 10.1016/j.bbadis.2011.06.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 06/07/2011] [Accepted: 06/10/2011] [Indexed: 02/07/2023]
|
27
|
Vits S, Cesko E, Enck P, Hillen U, Schadendorf D, Schedlowski M. Behavioural conditioning as the mediator of placebo responses in the immune system. Philos Trans R Soc Lond B Biol Sci 2011; 366:1799-807. [PMID: 21576137 DOI: 10.1098/rstb.2010.0392] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current placebo research postulates that conditioning processes are one of the major mechanisms of the placebo response. Behaviourally conditioned changes in peripheral immune functions have been demonstrated in experimental animals, healthy subjects and patients. The physiological mechanisms responsible for this 'learned immune response' are not yet fully understood, but some relevant afferent and efferent pathways in the communication between the brain and the peripheral immune system have been identified. In addition, possible benefits and applicability in clinical settings have been demonstrated where behaviourally conditioned immunosuppression attenuated the exacerbation of autoimmune diseases, prolonged allograft survival and affected allergic responses. Here, we summarize data describing the mechanisms and the potential clinical benefit of behaviourally conditioned immune functions, with particular focus on learned placebo effects on allergic reactions.
Collapse
Affiliation(s)
- Sabine Vits
- Institute of Medical Psychology and Behavioral Immunobiology, University Clinic Essen, Essen, Germany.
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW To review our current understanding of the relationship between absorption of nutrients and intestinal inflammatory response. RECENT FINDINGS There is increasing evidence linking gut local inflammatory events with the intake of nutrients. Our recent studies, using the conscious lymph fistula rat model, demonstrate that fat absorption activates the intestinal mucosal mast cells. This is accompanied by a dramatic increase in the lymphatic release of mast cell mediators including histamine, rat mucosal mast cell protease II (RMCPII), as well as the lipid mediator prostaglandin D2 (PGD2). Clinical studies suggest that increased consumption of animal fat may play a role in the pathogenesis of inflammatory bowel disease. This impact of dietary fat may not be restricted to the gut but may extend to the whole body. There is evidence linking a high-fat diet-induced metabolic syndrome, with a low-grade chronic inflammatory state. In this review, we hope to convince the readers that fat absorption can have far reaching physiological and pathophysiological consequences. SUMMARY Understanding the relationship between nutrient absorption and intestinal inflammation is important. We need a better understanding of the interaction between enterocytes and the intestinal immune cells in nutrient absorption and the gut inflammatory responses.
Collapse
Affiliation(s)
- Yong Ji
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio 45237, USA
| | | | | |
Collapse
|
29
|
Katiraei P, Bultron G. Need for a comprehensive medical approach to the neuro-immuno-gastroenterology of irritable bowel syndrome. World J Gastroenterol 2011; 17:2791-800. [PMID: 21734786 PMCID: PMC3120938 DOI: 10.3748/wjg.v17.i23.2791] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 03/08/2011] [Accepted: 03/15/2011] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is defined by the Rome III criteria as symptoms of recurrent abdominal pain or discomfort with the onset of a marked change in bowel habits with no evidence of an inflammatory, anatomic, metabolic, or neoplastic process. As such, many clinicians regard IBS as a central nervous system problem of altered pain perception. Here, we review the recent literature and discuss the evidence that supports an organic based model, which views IBS as a complex, heterogeneous, inter-dependent, and multi-variable inflammatory process along the neuronal-gut axis. We delineate the organic pathophysiology of IBS, demonstrate the role of inflammation in IBS, review the possible differences between adult and pediatric IBS, discuss the merits of a comprehensive treatment model as taught by the Institute of Functional Medicine, and describe the potential for future research for this syndrome.
Collapse
|
30
|
De Winter BY, van den Wijngaard RM, de Jonge WJ. Intestinal mast cells in gut inflammation and motility disturbances. Biochim Biophys Acta Mol Basis Dis 2011; 1822:66-73. [PMID: 21497195 DOI: 10.1016/j.bbadis.2011.03.016] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 03/20/2011] [Accepted: 03/25/2011] [Indexed: 12/12/2022]
Abstract
Mast cells may be regarded as prototypes of innate immune cells that can be controlled by neuronal mediators. Their activation has been implicated in many types of neuro-inflammatory responses, and related disturbances of gut motility, via direct or indirect mechanisms that involve several mechanisms relevant to disease pathogenesis such as changes in epithelial barrier function or activation of adaptive or innate immune responses. Here we review the evidence for the involvement of mast cells in the inflammation of the bowel wall caused by bowel manipulation that leads to motility disturbances such as postoperative gastroparesis and ileus. Also in IBD there is substantial evidence for the involvement of mast cells and a mast cell-mediated neuroimmune interaction showing an increased number and an increased degranulation of mast cells. We discuss the potential of mast cell inhibition as a bona fide drug target to relief postoperative ileus. Further research on mast cell-related therapy either by stabilizing the mast cells or by blocking specific mast cell mediators as adjunctive therapy in IBD is encouraged, bearing in mind that several drugs currently used in the treatment of IBD possess properties affecting mast cell activities. This article is part of a Special Issue entitled: Mast cells in inflammation.
Collapse
Affiliation(s)
- Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Department of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | | | | |
Collapse
|
31
|
van Diest SA, Stanisor OI, Boeckxstaens GE, de Jonge WJ, van den Wijngaard RM. Relevance of mast cell-nerve interactions in intestinal nociception. Biochim Biophys Acta Mol Basis Dis 2011; 1822:74-84. [PMID: 21496484 DOI: 10.1016/j.bbadis.2011.03.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 03/11/2011] [Accepted: 03/30/2011] [Indexed: 01/06/2023]
Abstract
Cross-talk between the immune- and nervous-system is considered an important biological process in health and disease. Because mast cells are often strategically placed between nerves and surrounding (immune)-cells they may function as important intermediate cells. This review summarizes the current knowledge on bidirectional interaction between mast cells and nerves and its possible relevance in (inflammation-induced) increased nociception. Our main focus is on mast cell mediators involved in sensitization of TRP channels, thereby contributing to nociception, as well as neuron-released neuropeptides and their effects on mast cell activation. Furthermore we discuss mechanisms involved in physical mast cell-nerve interactions. This article is part of a Special Issue entitled: Mast cells in inflammation.
Collapse
Affiliation(s)
- Sophie A van Diest
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
32
|
Rychter JW, Van Nassauw L, Timmermans JP, Akkermans LMA, Westerink RHS, Kroese ABA. CGRP1 receptor activation induces piecemeal release of protease-1 from mouse bone marrow-derived mucosal mast cells. Neurogastroenterol Motil 2011; 23:e57-68. [PMID: 20964790 DOI: 10.1111/j.1365-2982.2010.01617.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The parasitized or inflamed gastrointestinal mucosa shows an increase in the number of mucosal mast cells (MMC) and the density of extrinsic primary afferent nerve fibers containing the neuropeptide, calcitonin gene-related peptide (CGRP). Currently, the mode of action of CGRP on MMC is unknown. METHODS The effects of CGRP on mouse bone marrow-derived mucosal mast cells (BMMC) were investigated by measurements of intracellular Ca(2+)[Ca(2+)](i) and release of mMCP-1. KEY RESULTS Bone marrow-derived mucosal mast cells responded to the application of CGRP with a single transient rise in [Ca(2+)](i). The proportion of responding cells increased concentration-dependently to a maximum of 19 ± 4% at 10(-5)mol L(-1) (mean ±SEM; C48/80 100%; EC(50)10(-8) mol L(-1) ). Preincubation with the CGRP receptor antagonist BIBN4096BS (10(-5) mol L(-1)) completely inhibited BMMC activation by CGRP [range 10(-5) to 10(-11) mol L(-1); analysis of variance (ANOVA) P < 0.001], while preincubation with LaCl(3) to block Ca(2+) entry did not affect the response (P = 0.18). The presence of the CGRP1 receptor on BMMC was confirmed by simultaneous immunofluorescent detection of RAMP1 or CRLR, the two components of the CGRP1 receptor, and mMCP-1. Application of CGRP for 1 h evoked a concentration-dependent release of mMCP-1 (at EC(50) 10% of content) but not of β-hexosaminidase and alterations in granular density indicative of piecemeal release. CONCLUSIONS & INFERENCES We demonstrate that BMMC express functional CGRP1 receptors and that their activation causes mobilization of Ca(2+) from intracellular stores and piecemeal release of mMCP-1. These findings support the hypothesis that the CGRP signaling from afferent nerves to MMC in the gastrointestinal wall is receptor-mediated.
Collapse
Affiliation(s)
- J W Rychter
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
33
|
Protection against early intestinal compromise by lipid-rich enteral nutrition through cholecystokinin receptors. Crit Care Med 2010; 38:1592-7. [PMID: 20453642 DOI: 10.1097/ccm.0b013e3181e2cd4d] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Early gut wall integrity loss and local intestinal inflammation are associated with the development of inflammatory complications in surgical and trauma patients. Prevention of these intestinal events is a potential target for therapies aimed to control systemic inflammation. Previously, we demonstrated in a rodent shock model that lipid-rich enteral nutrition attenuated systemic inflammation and prevented organ damage through a cholecystokinin receptor-dependent vagal pathway. The influence of lipid-rich nutrition on very early intestinal compromise as seen after shock is investigated. Next, the involvement of cholecystokinin receptors on the nutritional modulation of immediate gut integrity loss and intestinal inflammation is studied. DESIGN Randomized controlled in vivo study. SETTING University research unit. SUBJECTS Male Sprague-Dawley rats. INTERVENTIONS Liquid lipid-rich nutrition or control low-lipid feeding was administered per gavage before hemorrhagic shock. Cholecystokinin receptor antagonists were used to investigate involvement of the vagal antiinflammatory pathway. MEASUREMENTS AND MAIN RESULTS Gut permeability to horseradish peroxidase increased as soon as 30 mins postshock and was prevented by lipid-rich nutrition compared with low-lipid (p<.01) and fasted controls (p<.001). Furthermore, lipid-rich nutrition reduced plasma levels of enterocyte damage marker ileal lipid binding protein at 60 mins (p<.05). Early gut barrier dysfunction correlated with rat mast cell protease plasma concentrations at 30 mins (rs=0.67; p<.001) and intestinal myeloperoxidase levels at 60 mins (rs=0.58; p<.05). Lipid-rich nutrition significantly reduced plasma rat mast cell protease (p<.01) and myeloperoxidase (p<.05) before systemic inflammation was detectable. Protective effects of lipid-rich nutrition were abrogated by cholecystokinin receptor antagonists (horseradish peroxidase; p<.05 and rat mast cell protease; p<.05). CONCLUSIONS Lipid-rich enteral nutrition prevents early gut barrier loss, enterocyte damage, and local intestinal inflammation before systemic inflammation develops in a cholecystokinin receptor-dependent manner. This study identifies activation of the vagal antiinflammatory pathway with lipid-rich nutrition as a potential therapy in patients prone to develop a compromised gut.
Collapse
|
34
|
Estofolete CF, Botelho-Machado C, Taboga SR, Zucoloto S, Polli-Lopes AC, Gil CD. Effects of myenteric denervation on extracellular matrix fibers and mast cell distribution in normal stomach and gastric lesions. Cancer Cell Int 2010; 10:18. [PMID: 20569458 PMCID: PMC2908609 DOI: 10.1186/1475-2867-10-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Accepted: 06/22/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In this study the effect of myenteric denervation induced by benzalconium chloride (BAC) on distribution of fibrillar components of extracellular matrix (ECM) and inflammatory cells was investigated in gastric carcinogenesis induced by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Rats were divided in four experimental groups: non-denervated (I) and denervated stomach (II) without MNNG treatment; non-denervated (III) and denervated stomachs (IV) treated with MNNG. For histopathological, histochemical and stereological analysis, sections of gastric fragments were stained with Hematoxylin-Eosin, Picrosirius-Hematoxylin, Gomori reticulin, Weigert's Resorcin-Fuchsin, Toluidine Blue and Alcian-Blue/Safranin (AB-SAF). RESULTS BAC denervation causes an increase in the frequency of reticular and elastic fibers in the denervated (group II) compared to the non-denervated stomachs (group I). The treatment of the animals with MNNG induced the development of adenocarcinomas in non-denervated and denervated stomachs (groups III and IV, respectively) with a notable increase in the relative volume of the stroma, the frequency of reticular fibers and the inflammatory infiltrate that was more intense in group IV. An increase in the frequency of elastic fibers was observed in adenocarcinomas of denervated (group IV) compared to the non-denervated stomachs (group III) that showed degradation of these fibers. The development of lesions (groups III and IV) was also associated with an increase in the mast cell population, especially AB and AB-SAF positives, the latter mainly in the denervated group IV. CONCLUSIONS The results show a strong association in the morphological alteration of the ECM fibrillar components, the increased density of mast cells and the development of tumors induced by MNNG in the non-denervated rat stomach or denervated by BAC. This suggests that the study of extracellular and intracellular components of tumor microenvironment contributes to understanding of tumor biology by action of myenteric denervation.
Collapse
Affiliation(s)
- Cássia F Estofolete
- Department of Anatomy, School of Medicine - FAMERP, Avenida Brigadeiro Faria Lima 5416, CEP 15090-000, São José do Rio Preto, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
35
|
Traver E, Torres R, de Mora F, Vergara P. Mucosal mast cells mediate motor response induced by chronic oral exposure to ovalbumin in the rat gastrointestinal tract. Neurogastroenterol Motil 2010; 22:e34-43. [PMID: 19682267 DOI: 10.1111/j.1365-2982.2009.01377.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We previously demonstrated that oral chronic exposure to ovalbumin (OVA) causes intestinal hypermotility in Sprague-Dawley rats. In this study, the objective was to determine the mechanism of action of OVA and the role of mucosal mast cells in the regulation of motor activity in this model. Rats were orally exposed to OVA during 6 weeks. Intestinal mucosal mast cells (IMMCs) were counted and rat mast cell protease II (RMCPII) measured in duodenum, jejunum, ileum and colon. Anti-OVA IgE, IgG, and IL-4 were measured in serum. Eosinophils and IgE(+) cells were counted in jejunum. In an additional study rats were treated with the mast cell stabilizer ketotifen and mast cell number, RMCPII concentration and motor activity in vitro were evaluated. OVA exposed rats showed an increase in mucosal mast cell number and in RMCPII content in small intestine and colon. However, variables of a Th(2) type response were not affected by exposure to OVA: (i) neither OVA specific IgE nor IgG were found; (ii) IL-4 did not increase and, (iii) the number of eosinophils and IgE(+) cells was identical in the exposed and unexposed groups. These results brought us to hypothesize a possible non-Ig-mediated action of OVA on mast cells. Ketotifen significantly diminished the response to OVA: Ketotifen reduced the number of mast cells and the RMCPII content and blocked increased intestinal contractility. In addition ketotifen modified motor response in both OVA exposed and unexposed animals giving evidence of the importance of mast cells in intestine motor activity driving.
Collapse
Affiliation(s)
- E Traver
- Department of Cell Biology, Immunology and Physiology, Universitat Autònoma de Barcelona, Spain
| | | | | | | |
Collapse
|
36
|
Yuan XJ, Huang LB, Qiao HL, Deng ZP, Fa JJ. Uterine autonomic nerve innervation plays a crucial role in regulating rat uterine mast cell functions during embryo implantation. Prostaglandins Other Lipid Mediat 2009; 90:94-7. [DOI: 10.1016/j.prostaglandins.2009.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2009] [Revised: 08/19/2009] [Accepted: 09/09/2009] [Indexed: 11/16/2022]
|
37
|
Lipid-rich enteral nutrition reduces postoperative ileus in rats via activation of cholecystokinin-receptors. Ann Surg 2009; 249:481-7. [PMID: 19247038 DOI: 10.1097/sla.0b013e318194d187] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study investigates the effect of lipid-rich nutrition on the local inflammatory response and gastrointestinal hypomotility in a rat model of postoperative ileus. BACKGROUND Postoperative ileus is a major clinical problem, in which inflammation of the intestinal muscularis plays a key pathogenic event. Previously, administration of lipid-rich nutrition has been shown to reduce inflammation by activation of the autonomic nervous system via cholecystokinin-receptors. METHODS Postoperative ileus was induced by manipulation of the small intestine in rats. Peritoneal lavage fluid, plasma, and jejunal segments were collected at several time points to determine inflammatory mediators in fasted rats and rats fed a lipid-rich or control nutrition. Gastrointestinal transit was measured 24 hours after surgery. RESULTS Administration of lipid-rich nutrition markedly reduced the manipulation-induced local inflammatory response compared to rats treated with control nutrition. The intervention with lipid-rich nutrition significantly reduced plasma levels of rat mast cell protease-II (P < 0.05) and peritoneal levels of tumor necrosis factor-alpha (P < 0.01) and interleukin-6 (P < 0.05). Furthermore, the influx of neutrophils, expressed as tissue level myeloperoxidase was significantly prevented by lipid-rich nutrition (P < 0.05). Above all administration of lipid-rich enteral nutrition resulted in a significant improvement of gastrointestinal transit compared to control nutrition (P < 0.05). Blocking of cholecystokinin-receptors prevented the anti-inflammatory and motility promoting effect of lipid-rich feeding. CONCLUSION Our data demonstrate that nutritional stimulation of the autonomic nervous system with enteral lipids reduces postoperative ileus by inhibition of inflammation. Clinically, lipid-rich enteral nutrition may be a new therapeutic option in the treatment of postoperative ileus.
Collapse
|
38
|
Walker MM, Talley NJ. Functional gastrointestinal disorders and the potential role of eosinophils. Gastroenterol Clin North Am 2008; 37:383-95, vi. [PMID: 18499026 DOI: 10.1016/j.gtc.2008.02.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The eosinophil-mast cell-neural pathway may be important in the pathophysiology of functional gastrointestinal disorders characterized by unexplained abdominal pain, disordered defecation, or meal-related discomfort. There is evidence that duodenal eosinophils are increased in functional dyspepsia, whereas mast cells are increased in the lower gut in irritable bowel syndrome, directly supporting a role for a hypersensitivity-type reaction in these disorders. The trigger may be a pathogen, food, or other allergen in the gut mucosa. This trigger may evoke eosinophils, mast cells, and other components to cascade to up-regulate serotonin release, with modulation of the enteric and central nervous systems, creating a vicious cycle. If correct, this theory suggests treatment should specifically target the eosinophil-mast cell pathway.
Collapse
Affiliation(s)
- Marjorie M Walker
- Department of Histopathology, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom.
| | | |
Collapse
|
39
|
Mazzone A, Farrugia G. Evolving concepts in the cellular control of gastrointestinal motility: neurogastroenterology and enteric sciences. Gastroenterol Clin North Am 2007; 36:499-513, vii. [PMID: 17950435 DOI: 10.1016/j.gtc.2007.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The enteric nervous system is an independent nervous system with a complexity comparable with the central nervous system. This complex system is integrated into several other complex systems, such as interstitial cells of Cajal networks and immune cells. The result of these interactions is effective coordination of motility, secretion, and blood flow in the gastrointestinal tract. Loss of subsets of enteric nerves, of interstitial cells of Cajal, malfunction of smooth muscle, and alteration in immune cells have been identified as the basis of many motility disorders. The initial factors triggering these changes and how to intervene to prevent, halt, and reverse them needs to be understood.
Collapse
Affiliation(s)
- Amelia Mazzone
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | |
Collapse
|
40
|
Trimble N, Johnson AC, Foster A, Greenwood-van Meerveld B. Corticotropin-releasing factor receptor 1-deficient mice show decreased anxiety and colonic sensitivity. Neurogastroenterol Motil 2007; 19:754-60. [PMID: 17539891 DOI: 10.1111/j.1365-2982.2007.00951.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Corticotropin releasing factor (CRF) is an important mediator in the stress response. Previous studies in rodent models demonstrated that stress-induced colonic hypersensitivity was inhibited by CRF1 receptor antagonism. As CRF(1)R-deficient mice have (+/+), CRF(1)R (+/-) and CRF(1)R (-/-) mice colonic sensitivity was assessed via a visceromotor behavioural response (VMR) induced by colorectal distension (CRD, 0-60 mmHg). In the CRF(1)R (+/+) mice there was a pressure-dependent increase in the VMR to CRD that was moderately attenuated in the CRF1R (+/-) mice. However in the CRF(1)R (-/-) mice a VMR to CRD was only observed at the highest distension pressure (60 mmHg). A CRF(1)R antagonist, NBI 30775 (30 mg kg(-1) i.p.) significantly decreased the VMR to CRD in CRF(1)R +/+ mice. An identical inhibitory effect of NBI 30775 was observed in 43% of the CRF(1)R +/- mice. This study provides pharmacological and genetic evidence for the importance of CRF(1)R in colonic sensitivity and suggests a link between stress and visceral perception.
Collapse
Affiliation(s)
- N Trimble
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, OK, USA
| | | | | | | |
Collapse
|
41
|
de Jonge WJ, Ulloa L. The alpha7 nicotinic acetylcholine receptor as a pharmacological target for inflammation. Br J Pharmacol 2007; 151:915-29. [PMID: 17502850 PMCID: PMC2042938 DOI: 10.1038/sj.bjp.0707264] [Citation(s) in RCA: 467] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The physiological regulation of the immune system encompasses comprehensive anti-inflammatory mechanisms that can be harnessed for the treatment of infectious and inflammatory disorders. Recent studies indicate that the vagal nerve, involved in control of heart rate, hormone secretion and gastrointestinal motility, is also an immunomodulator. In experimental models of inflammatory diseases, vagal nerve stimulation attenuates the production of proinflammatory cytokines and inhibits the inflammatory process. Acetylcholine, the principal neurotransmitter of the vagal nerve, controls immune cell functions via the alpha7 nicotinic acetylcholine receptor (alpha7nAChR). From a pharmacological perspective, nicotinic agonists are more efficient than acetylcholine at inhibiting the inflammatory signaling and the production of proinflammatory cytokines. This 'nicotinic anti-inflammatory pathway' may have clinical implications as treatment with nicotinic agonists can modulate the production of proinflammatory cytokines from immune cells. Nicotine has been tested in clinical trials as a treatment for inflammatory diseases such as ulcerative colitis, but the therapeutic potential of this mechanism is limited by the collateral toxicity of nicotine. Here, we review the recent advances that support the design of more specific receptor-selective nicotinic agonists that have anti-inflammatory effects while eluding its collateral toxicity.
Collapse
Affiliation(s)
- W J de Jonge
- Department of Gastroenterology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | |
Collapse
|
42
|
McDonald MD, Gilmour KM, Barimo JF, Frezza PE, Walsh PJ, Perry SF. Is urea pulsing in toadfish related to environmental O2 or CO2 levels? Comp Biochem Physiol A Mol Integr Physiol 2007; 146:366-74. [PMID: 17196858 DOI: 10.1016/j.cbpa.2006.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Revised: 11/12/2006] [Accepted: 11/14/2006] [Indexed: 11/17/2022]
Abstract
The neurochemical, serotonin (5-hydroxytryptamine; 5-HT) is involved in the regulation of toadfish pulsatile urea excretion as well as the teleost hypoxia response. Thus, the goal of this study was to determine whether environmental conditions that activate branchial chemoreceptors also trigger pulsatile urea excretion in toadfish, since environmental dissolved oxygen levels in a typical toadfish habitat show significant diel fluctuations, often reaching hypoxic conditions at dawn. Toadfish were fitted with arterial, venous and/or buccal catheters and were exposed to various environmental conditions, and/or injected with the O(2) chemoreceptor agonist NaCN or the 5-HT(2) receptor agonist alpha-methyl-5HT. Arterial PO(2), as well as ammonia and urea excretion were monitored. Natural fluctuations in arterial PO(2) levels in toadfish did not correlate with the occurrence of a urea pulse. Chronic exposure (24 h) of toadfish to hyperoxia was without effect on nitrogen excretion, however, exposure to hypoxia caused a significant reduction in the frequency of urea pulses, and exposure to hypercapnia resulted in a reduction in the percentage of nitrogen waste excreted as urea. Of toadfish exposed acutely to hypoxia, 20% pulsed within 1 h, whereas none pulsed after normoxic or hypercapnic treatments. Furthermore, 20% of fish injected intravenously with NaCN pulsed within 1 h of injection, but no fish pulsed after injection of NaCN into the buccal cavity. To test whether environmental conditions affected 5-HT(2) receptors, toadfish were injected with alpha-methyl-5HT, which elicits urea pulses in toadfish. No significant differences in pulse size occurred among the various environmental treatments. Our findings suggest that neither the environmental conditions of hypoxia, hyperoxia or hypercapnia, nor direct branchial chemoreceptor activation by NaCN play a major role in the regulation of pulsatile urea excretion in toadfish.
Collapse
Affiliation(s)
- M Danielle McDonald
- Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, Florida, 33149-1098, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Van Nassauw L, Adriaensen D, Timmermans JP. The bidirectional communication between neurons and mast cells within the gastrointestinal tract. Auton Neurosci 2006; 133:91-103. [PMID: 17169619 DOI: 10.1016/j.autneu.2006.10.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 09/26/2006] [Accepted: 10/10/2006] [Indexed: 12/12/2022]
Abstract
Normal or disordered behaviour of the gastrointestinal tract is determined by a complex interplay between the epithelial barrier, immune cells, blood vessels, smooth muscle and intramurally located nerve elements. Mucosal mast cells (MMCs), which are able to detect noxious and antigenic threats and to generate or amplify signals to the other cells, are assigned a rather central position in this complex network. Signal input from MMCs to intrinsic enteric neurons is particularly crucial, because the enteric nervous system fulfils a pivotal role in the control of gastrointestinal functions. Activated enteric neurons are able to generate an alarm program involving alterations in motility and secretion. MMC signalling to extrinsic nerve fibres takes part in pathways generating visceral pain or extrinsic reflexes contributing to the disturbed motor and secretory function. Morphological and functional studies, especially studies concerning physiological stress, have provided evidence that, apart from the interaction between the enteric nervous system and MMCs, there is also a functional communication between the central nervous system and these mast cells. Psychological factors trigger neuronal pathways, which directly or indirectly affect MMCs. Further basic and clinical research will be needed to clarify in more detail whether basic patterns of this type of interactions are conserved between species including humans.
Collapse
Affiliation(s)
- Luc Van Nassauw
- Research Group Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Belgium
| | | | | |
Collapse
|