1
|
Ren W, Hua M, Cao F, Zeng W. The Sympathetic-Immune Milieu in Metabolic Health and Diseases: Insights from Pancreas, Liver, Intestine, and Adipose Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306128. [PMID: 38039489 PMCID: PMC10885671 DOI: 10.1002/advs.202306128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/28/2023] [Indexed: 12/03/2023]
Abstract
Sympathetic innervation plays a crucial role in maintaining energy balance and contributes to metabolic pathophysiology. Recent evidence has begun to uncover the innervation landscape of sympathetic projections and sheds light on their important functions in metabolic activities. Additionally, the immune system has long been studied for its essential roles in metabolic health and diseases. In this review, the aim is to provide an overview of the current research progress on the sympathetic regulation of key metabolic organs, including the pancreas, liver, intestine, and adipose tissues. In particular, efforts are made to highlight the critical roles of the peripheral nervous system and its potential interplay with immune components. Overall, it is hoped to underscore the importance of studying metabolic organs from a comprehensive and interconnected perspective, which will provide valuable insights into the complex mechanisms underlying metabolic regulation and may lead to novel therapeutic strategies for metabolic diseases.
Collapse
Affiliation(s)
- Wenran Ren
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Meng Hua
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Fang Cao
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhou563000China
| | - Wenwen Zeng
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineTaiyuan030001China
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijing100084China
| |
Collapse
|
2
|
Filippi L, Nardini P, Zizi V, Molino M, Fazi C, Calvani M, Carrozzo F, Cavallaro G, Giuseppetti G, Calosi L, Crociani O, Pini A. β3 Adrenoceptor Agonism Prevents Hyperoxia-Induced Colonic Alterations. Biomolecules 2023; 13:1755. [PMID: 38136626 PMCID: PMC10741994 DOI: 10.3390/biom13121755] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Oxygen level is a key regulator of organogenesis and its modification in postnatal life alters the maturation process of organs, including the intestine, which do not completely develop in utero. The β3-adrenoreceptor (β3-AR) is expressed in the colon and has an oxygen-dependent regulatory mechanism. This study shows the effects of the β3-AR agonist BRL37344 in a neonatal model of hyperoxia-driven colonic injury. For the first 14 days after birth, Sprague-Dawley rat pups were exposed to ambient oxygen levels (21%) or hyperoxia (85%) and treated daily with BRL37344 at 1, 3, 6 mg/kg or untreated. At the end of day 14, proximal colon samples were collected for analysis. Hyperoxia deeply influences the proximal colon development by reducing β3-AR-expressing cells (27%), colonic length (26%) and mucin production (47%), and altering the neuronal chemical coding in the myenteric plexus without changes in the neuron number. The administration of BRL37344 at 3 mg/kg, but not at 1 mg/kg, significantly prevented these alterations. Conversely, it was ineffective in preventing hyperoxia-induced body weight loss. BRL37344 at 6 mg/kg was toxic. These findings pave the way for β3-AR pharmacological targeting as a therapeutic option for diseases caused by hyperoxia-impaired development, typical prematurity disorders.
Collapse
Affiliation(s)
- Luca Filippi
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
| | - Patrizia Nardini
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
- Imaging Platform, Department Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
| | - Virginia Zizi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Marta Molino
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Camilla Fazi
- Department of Pediatric, Meyer Children’s University Hospital, 50139 Florence, Italy;
| | - Maura Calvani
- Division of Pediatric Oncology/Hematology, Meyer University Children’s Hospital, 50139 Florence, Italy; (M.C.); (F.C.)
| | - Francesco Carrozzo
- Division of Pediatric Oncology/Hematology, Meyer University Children’s Hospital, 50139 Florence, Italy; (M.C.); (F.C.)
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Giorgia Giuseppetti
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
| | - Laura Calosi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Olivia Crociani
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Alessandro Pini
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
- Imaging Platform, Department Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
| |
Collapse
|
3
|
Leven P, Schneider R, Schneider L, Mallesh S, Vanden Berghe P, Sasse P, Kalff JC, Wehner S. β-adrenergic signaling triggers enteric glial reactivity and acute enteric gliosis during surgery. J Neuroinflammation 2023; 20:255. [PMID: 37941007 PMCID: PMC10631040 DOI: 10.1186/s12974-023-02937-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Enteric glia contribute to the pathophysiology of various intestinal immune-driven diseases, such as postoperative ileus (POI), a motility disorder and common complication after abdominal surgery. Enteric gliosis of the intestinal muscularis externa (ME) has been identified as part of POI development. However, the glia-restricted responses and activation mechanisms are poorly understood. The sympathetic nervous system becomes rapidly activated by abdominal surgery. It modulates intestinal immunity, innervates all intestinal layers, and directly interfaces with enteric glia. We hypothesized that sympathetic innervation controls enteric glia reactivity in response to surgical trauma. METHODS Sox10iCreERT2/Rpl22HA/+ mice were subjected to a mouse model of laparotomy or intestinal manipulation to induce POI. Histological, protein, and transcriptomic analyses were performed to analyze glia-specific responses. Interactions between the sympathetic nervous system and enteric glia were studied in mice chemically depleted of TH+ sympathetic neurons and glial-restricted Sox10iCreERT2/JellyOPfl/+/Rpl22HA/+ mice, allowing optogenetic stimulation of β-adrenergic downstream signaling and glial-specific transcriptome analyses. A laparotomy model was used to study the effect of sympathetic signaling on enteric glia in the absence of intestinal manipulation. Mechanistic studies included adrenergic receptor expression profiling in vivo and in vitro and adrenergic agonism treatments of primary enteric glial cell cultures to elucidate the role of sympathetic signaling in acute enteric gliosis and POI. RESULTS With ~ 4000 differentially expressed genes, the most substantial enteric glia response occurs early after intestinal manipulation. During POI, enteric glia switch into a reactive state and continuously shape their microenvironment by releasing inflammatory and migratory factors. Sympathetic denervation reduced the inflammatory response of enteric glia in the early postoperative phase. Optogenetic and pharmacological stimulation of β-adrenergic downstream signaling triggered enteric glial reactivity. Finally, distinct adrenergic agonists revealed β-1/2 adrenoceptors as the molecular targets of sympathetic-driven enteric glial reactivity. CONCLUSIONS Enteric glia act as early responders during post-traumatic intestinal injury and inflammation. Intact sympathetic innervation and active β-adrenergic receptor signaling in enteric glia is a trigger of the immediate glial postoperative inflammatory response. With immune-activating cues originating from the sympathetic nervous system as early as the initial surgical incision, adrenergic signaling in enteric glia presents a promising target for preventing POI development.
Collapse
Affiliation(s)
- Patrick Leven
- Department of Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Reiner Schneider
- Department of Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Shilpashree Mallesh
- Department of Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Louvain, Belgium
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
4
|
Li Y, Li YR, Jin Y, Li MY, Zhang Q, Cao J, Li F, Zhang H, Chen J, Li YQ. Involvement of enteric glial cells in colonic motility in a rat model of irritable bowel syndrome with predominant diarrhea. J Chem Neuroanat 2023; 128:102235. [PMID: 36669707 DOI: 10.1016/j.jchemneu.2023.102235] [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: 10/14/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
The enteric nervous system (ENS) is one of the important systems that regulate gastrointestinal function. The ENS is made up of enteric glial cells (EGCs) and neurons. For a long time, it was believed that the function of EGCs was only to give structural support to neurons. However, recent evidence indicates EGCs are involved in most gut functions, including the development and plasticity of the ENS, epithelial barrier, and motility. However, it remains unclear whether EGCs have the potential to modify colonic motility following irritable bowel syndrome (IBS) with predominant diarrhea (IBS-D). This study aimed to investigate changes in EGCs during IBS-D and assessed the effects of manipulating EGCs. An IBS-D rat model was constructed using acetic acid and restraint stress, and DL-fluorocitric acid (FC), an inhibitor of EGCs, was administered. The changes in EGCs and colonic motility were studied by employing techniques comprising morphological, molecular biological and functional experiments. The results showed significant activation of EGCs in the myenteric plexus (MP) of the IBS-D-induced rat colon with accelerated colonic motility. FC significantly reduced the activation of EGCs and colonic motility caused by acetic acid and restraint stress. Hypercontraction of the colon caused by IBS-D may be associated with activation of EGCs in the MP of the colon and this was prevented by FC. Therefore, regulating colon hypercontraction through interference with the activation of EGCs has significant prospects for clinical application to alleviate diarrhea in patients with IBS-D.
Collapse
Affiliation(s)
- Yan Li
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, China; Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, China
| | - Yan-Rong Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Yuan Jin
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, China
| | - Meng-Ying Li
- Department of Endocrinology and Metabolism, Xijing Hospital, The Fourth Medical University, Xi'an 710032, China
| | - Qian Zhang
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, China
| | - Jing Cao
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Fei Li
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China
| | - Hua Zhang
- Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, China.
| | - Jing Chen
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China.
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, China; Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, China; Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
5
|
Baidoo N, Sanger GJ, Belai A. Effect of old age on the subpopulations of enteric glial cells in human descending colon. Glia 2023; 71:305-316. [PMID: 36128665 PMCID: PMC10087700 DOI: 10.1002/glia.24272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/06/2022]
Abstract
Old age is associated with a higher incidence of lower bowel conditions such as constipation. Recent evidence suggest that colonic motility may be influenced by enteric glial cells (EGCs). Little is known about the effect of aging on the subpopulation of EGCs in the human colon. We assessed and compared the pattern of distribution of EGCs in adult and elderly human colon. Human descending colon were obtained from 23 cancer patients comprising of adults (23-63 years; 6 male, 7 female) and elderly (66-81 year; 6 male, 4 female). Specimens were serially-sectioned and immunolabeled with anti-Sox-10, anti-S100 and anti-GFAP for morphometric analysis. Standardized procedures were utilized to ensure unbiased counting and densitometric evaluation of EGCs. The number of Sox-10 immunoreactive (IR) EGCs were unaltered with age in both the myenteric plexus (MP) (respectively, in adult and elderly patients, 1939 ± 82 and 1760 ± 44/mm length; p > .05) and submucosal plexus; there were no apparent differences between adult males and females. The density of S100-IR EGCs declined among the elderly in the circular muscle and within the MP per ganglionic area. In the adult colon, there were more S100-IR EGCs distributed in the circular muscle per unit area than the Taenia coli. There was little or no GFAP-IR EGCs in both adult and elderly colon. We concluded that aging of the human descending colon does not result in a loss of Sox-10-IR EGCs in the MP and SMP but reduces S100-IR EGCs density within the musculature. This alteration in myenteric EGCs density with age may contribute to colonic dysfunction.
Collapse
Affiliation(s)
- Nicholas Baidoo
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Gareth J Sanger
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Abi Belai
- School of Life and Health Sciences, University of Roehampton, London, UK
| |
Collapse
|
6
|
Rotavirus Downregulates Tyrosine Hydroxylase in the Noradrenergic Sympathetic Nervous System in Ileum, Early in Infection and Simultaneously with Increased Intestinal Transit and Altered Brain Activities. mBio 2022; 13:e0138722. [PMID: 36094089 PMCID: PMC9600178 DOI: 10.1128/mbio.01387-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
While rotavirus diarrhea has been considered to occur only due to intrinsic intestinal effects within the enteric nervous system, we provide evidence for central nervous system control underlying the clinical symptomology. Our data visualize infection by large-scale three-dimensional (3D) volumetric tissue imaging of a mouse model and demonstrate that rotavirus infection disrupts the homeostasis of the autonomous system by downregulating tyrosine hydroxylase in the noradrenergic sympathetic nervous system in ileum, concomitant with increased intestinal transit. Interestingly, the nervous response was found to occur before the onset of clinical symptoms. In adult infected animals, we found increased pS6 immunoreactivity in the area postrema of the brain stem and decreased phosphorylated STAT5-immunoreactive neurons in the bed nucleus of the stria terminalis, which has been associated with autonomic control, including stress response. Our observations contribute to knowledge of how rotavirus infection induces gut-nerve-brain interaction early in the disease.
Collapse
|
7
|
Immunohistochemical visualisation of the enteric nervous system architecture in the germ-free piglets. J Mol Histol 2022; 53:773-780. [PMID: 35689149 DOI: 10.1007/s10735-022-10079-5] [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: 12/20/2021] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
The enteric nervous system (ENS), considered as separate branch of the autonomic nervous system, is located throughout the length of the gastrointestinal tract as a series of interconnected ganglionic plexuses. Recently, the ENS is getting more in the focus of gastrointestinal research. For years, the main interest and research was aimed to the enteric neurons and their functional properties in normal conditions, less attention has been paid to the germ-free animals. Germ-free (GF) piglets have clear microbiological background and are reared in sterile environment. GF piglets are regarded as clinically relevant models for studying of human diseases, as these piglets' manifest similar clinical symptoms to humans. In this study we briefly summarised the main characteristics in immunohistochemical distribution of ENS elements in the wall of jejunum and colon of germ-free piglets.
Collapse
|
8
|
Dobson GP, Morris JL, Biros E, Davenport LM, Letson HL. Major surgery leads to a proinflammatory phenotype: Differential gene expression following a laparotomy. Ann Med Surg (Lond) 2021; 71:102970. [PMID: 34745602 PMCID: PMC8554464 DOI: 10.1016/j.amsu.2021.102970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/17/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The trauma of surgery is a neglected area of research. Our aim was to examine the differential expression of genes of stress, metabolism and inflammation in the major organs of a rat following a laparotomy. MATERIALS AND METHODS Anaesthetised Sprague-Dawley rats were randomised into baseline, 6-hr and 3-day groups (n = 6 each), catheterised and laparotomy performed. Animals were sacrificed at each timepoint and tissues collected for gene and protein analysis. Blood stress hormones, cytokines, endothelial injury markers and coagulation were measured. RESULTS Stress hormone corticosterone significantly increased and was accompanied by significant increases in inflammatory cytokines, endothelial markers, increased neutrophils (6-hr), higher lactate (3-days), and coagulopathy. In brain, there were significant increases in M1 muscarinic (31-fold) and α-1A-adrenergic (39-fold) receptor expression. Cortical expression of metabolic genes increased ∼3-fold, and IL-1β by 6-fold at 3-days. Cardiac β-1-adrenergic receptor expression increased up to 8.4-fold, and M2 and M1 muscarinic receptors by 2 to 4-fold (6-hr). At 3-days, cardiac mitochondrial gene expression (Tfam, Mtco3) and inflammation (IL-1α, IL-4, IL-6, MIP-1α, MCP-1) were significantly elevated. Haemodynamics remained stable. In liver, there was a dramatic suppression of adrenergic and muscarinic receptor expression (up to 90%) and increased inflammation. Gut also underwent autonomic suppression with 140-fold increase in IL-1β expression (3-days). CONCLUSIONS A single laparotomy led to a surgical-induced proinflammatory phenotype involving neuroendocrine stress, cortical excitability, immune activation, metabolic changes and coagulopathy. The pervasive nature of systemic and tissue inflammation was noteworthy. There is an urgent need for new therapies to prevent hyper-inflammation and restore homeostasis following major surgery.
Collapse
Affiliation(s)
- Geoffrey P. Dobson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Jodie L. Morris
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Erik Biros
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Lisa M. Davenport
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Hayley L. Letson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| |
Collapse
|
9
|
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
|
10
|
Aurora SK, Shrewsbury SB, Ray S, Hindiyeh N, Nguyen L. A link between gastrointestinal disorders and migraine: Insights into the gut-brain connection. Headache 2021; 61:576-589. [PMID: 33793965 PMCID: PMC8251535 DOI: 10.1111/head.14099] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/03/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022]
Abstract
Background Migraine is a complex, multifaceted, and disabling headache disease that is often complicated by gastrointestinal (GI) conditions, such as gastroparesis, functional dyspepsia, and cyclic vomiting syndrome (CVS). Functional dyspepsia and CVS are part of a spectrum of disorders newly classified as disorders of gut–brain interaction (DGBI). Gastroparesis and functional dyspepsia are both associated with delayed gastric emptying, while nausea and vomiting are prominent in CVS, which are also symptoms that commonly occur with migraine attacks. Furthermore, these gastric disorders are comorbidities frequently reported by patients with migraine. While very few studies assessing GI disorders in patients with migraine have been performed, they do demonstrate a physiological link between these conditions. Objective To summarize the available studies supporting a link between GI comorbidities and migraine, including historical and current scientific evidence, as well as provide evidence that symptoms of GI disorders are also observed outside of migraine attacks during the interictal period. Additionally, the importance of route of administration and formulation of migraine therapies for patients with GI symptoms will be discussed. Methods A literature search of PubMed for articles relating to the relationship between the gut and the brain with no restriction on the publication year was performed. Studies providing scientific support for associations of gastroparesis, functional dyspepsia, and CVS with migraine and the impact these associations may have on migraine treatment were the primary focus. This is a narrative review of identified studies. Results Although the association between migraine and GI disorders has received very little attention in the literature, the existing evidence suggests that they may share a common etiology. In particular, the relationship between migraine, gastric motility, and vomiting has important clinical implications in the treatment of migraine, as delayed gastric emptying and vomiting may affect oral dosing compliance, and thus, the absorption and efficacy of oral migraine treatments. Conclusions There is evidence of a link between migraine and GI comorbidities, including those under the DGBI classification. Many patients do not find adequate relief with oral migraine therapies, which further necessitates increased recognition of GI disorders in patients with migraine by the headache community.
Collapse
Affiliation(s)
- Sheena K Aurora
- Medical Affairs, Impel NeuroPharma, Seattle, WA, USA.,Department of Neurology, Stanford University, Stanford, CA, USA
| | | | - Sutapa Ray
- Medical Affairs, Impel NeuroPharma, Seattle, WA, USA
| | - Nada Hindiyeh
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Linda Nguyen
- Department of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| |
Collapse
|
11
|
Smith-Edwards KM, Edwards BS, Wright CM, Schneider S, Meerschaert KA, Ejoh LL, Najjar SA, Howard MJ, Albers KM, Heuckeroth RO, Davis BM. Sympathetic Input to Multiple Cell Types in Mouse and Human Colon Produces Region-Specific Responses. Gastroenterology 2021; 160:1208-1223.e4. [PMID: 32980343 PMCID: PMC7956113 DOI: 10.1053/j.gastro.2020.09.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/18/2020] [Accepted: 09/15/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND & AIMS The colon is innervated by intrinsic and extrinsic neurons that coordinate functions necessary for digestive health. Sympathetic input suppresses colon motility by acting on intrinsic myenteric neurons, but the extent of sympathetic-induced changes on large-scale network activity in myenteric circuits has not been determined. Compounding the complexity of sympathetic function, there is evidence that sympathetic transmitters can regulate activity in non-neuronal cells (such as enteric glia and innate immune cells). METHODS We performed anatomical tracing, immunohistochemistry, optogenetic (GCaMP calcium imaging, channelrhodopsin), and colon motility studies in mice and single-cell RNA sequencing in human colon to investigate how sympathetic postganglionic neurons modulate colon function. RESULTS Individual neurons in each sympathetic prevertebral ganglion innervated the proximal or distal colon, with processes closely opposed to multiple cell types. Calcium imaging in semi-intact mouse colon preparations revealed changes in spontaneous and evoked neural activity, as well as activation of non-neuronal cells, induced by sympathetic nerve stimulation. The overall pattern of response to sympathetic stimulation was unique to the proximal or distal colon. Region-specific changes in cellular activity correlated with motility patterns produced by electrical and optogenetic stimulation of sympathetic pathways. Pharmacology experiments (mouse) and RNA sequencing (human) indicated that appropriate receptors were expressed on different cell types to account for the responses to sympathetic stimulation. Regional differences in expression of α-1 adrenoceptors in human colon emphasize the translational relevance of our mouse findings. CONCLUSIONS Sympathetic neurons differentially regulate activity of neurons and non-neuronal cells in proximal and distal colon to promote distinct changes in motility patterns, likely reflecting the distinct roles played by these 2 regions.
Collapse
Affiliation(s)
- Kristen M. Smith-Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania,For correspondence: Kristen M. Smith-Edwards, University of Pittsburgh, Department of Neurobiology, 200 Lothrop Street, Pittsburgh, PA 15216, , Ph: 412-648-9745
| | - Brian S. Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christina M. Wright
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Sabine Schneider
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Kimberly A. Meerschaert
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lindsay L. Ejoh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sarah A. Najjar
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Kathryn M. Albers
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert O. Heuckeroth
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Brian M. Davis
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Fung C, Vanden Berghe P. Functional circuits and signal processing in the enteric nervous system. Cell Mol Life Sci 2020; 77:4505-4522. [PMID: 32424438 PMCID: PMC7599184 DOI: 10.1007/s00018-020-03543-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/13/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
The enteric nervous system (ENS) is an extensive network comprising millions of neurons and glial cells contained within the wall of the gastrointestinal tract. The major functions of the ENS that have been most studied include the regulation of local gut motility, secretion, and blood flow. Other areas that have been gaining increased attention include its interaction with the immune system, with the gut microbiota and its involvement in the gut-brain axis, and neuro-epithelial interactions. Thus, the enteric circuitry plays a central role in intestinal homeostasis, and this becomes particularly evident when there are faults in its wiring such as in neurodevelopmental or neurodegenerative disorders. In this review, we first focus on the current knowledge on the cellular composition of enteric circuits. We then further discuss how enteric circuits detect and process external information, how these signals may be modulated by physiological and pathophysiological factors, and finally, how outputs are generated for integrated gut function.
Collapse
Affiliation(s)
- Candice Fung
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Drummond PD, Morellini N, Visser E, Finch PM. Expression of Cutaneous Beta-2 Adrenoceptors Is Similar in Patients with Complex Regional Pain Syndrome and Pain-Free Controls. PAIN MEDICINE 2020; 21:1199-1207. [PMID: 31120123 DOI: 10.1093/pm/pnz110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
OBJECTIVE Studies in rodents suggest that cutaneous beta-2 adrenoceptors (β2-ARs) mediate inflammation and pain after tissue injury and that inflammation and peripheral nerve injury trigger increases in neuronal β2-AR expression. Hence, the aim of this study was to investigate the expression of β2-ARs on keratinocytes and dermal nerves in patients with complex regional pain syndrome (CRPS). DESIGN, SETTING, AND SUBJECTS Fifty-eight patients with CRPS were recruited for this study. In addition, skin biopsies were obtained from 13 pain-free women and three pain-free men of similar age and sex distribution as the patients. METHODS Quantitative sensory tests for assessing sensitivity to pressure, pinprick, light touch, heat, and cold were administered, and skin biopsies were obtained from the affected and contralateral limbs. Skin biopsies were also obtained from a similar site on the dorsal hand or foot of pain-free controls. Immunohistochemistry and confocal microscopy were used to identify β2-ARs on keratinocytes, dermal nerves, and blood vessels in the skin samples. RESULTS The distribution of β2-ARs in keratinocytes and nerves was similar in the affected and contralateral limbs of patients and was similar for target cells in patients and controls. However, elevated β2-AR expression in reticular nerve bundles was associated with heightened sensitivity to heat pain. CONCLUSIONS These findings do not support a major role of cutaneous β2-ARs in CRPS. However, activation of neuronal β2-ARs may contribute to thermal hyperalgesia in a subgroup of patients. Whether activation of β2-ARs on keratinocytes mediates inflammation early in the course of CRPS requires further investigation.
Collapse
Affiliation(s)
- Peter D Drummond
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
| | - Natalie Morellini
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia.,School of Medicine, University of Notre Dame, Fremantle, Western Australia, Australia
| | - Eric Visser
- School of Medicine, University of Notre Dame, Fremantle, Western Australia, Australia
| | - Philip M Finch
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
14
|
Louza GSG, Carmo LLGD, Conceição IM. Effect of Tityus serrulatus scorpion venom on isolated jejunum: A very useful tool to study the interaction between neurons in the enteric nervous system. Auton Neurosci 2020; 227:102676. [PMID: 32464449 DOI: 10.1016/j.autneu.2020.102676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/06/2020] [Accepted: 05/08/2020] [Indexed: 12/24/2022]
Abstract
Scorpion envenomation is a public health problem in tropical and subtropical areas. In Brazil, Tityus serrulatus is the biggest cause of accidents with venomous animals. Tityus serrulatus venom causes symptoms related to a great activation of the autonomic system attributed to a massive release of sympathetic and parasympathetic mediators. This effect is attributed to the presence of toxins acting in Na+ and K+ ion channels, leading to an increase in cell excitability. Although gastrointestinal symptoms, like diarrhoea and sialorrhea, is observed in moderate to severe cases, little attention is given in clinical reports. Gastrointestinal motility is controlled by the enteric nervous system which is composed of a wide variety of interconnected neurons that are influenced by the sympathetic and parasympathetic nervous systems. Thus, this work aimed to characterize the effects of Tityus serrulatus venom on sympathetic and parasympathetic neurotransmission of rat jejunum, as well as to investigate possibles effects on other neurons of the enteric nervous system. To this, we verify the effects of Tityus serrulatus venom on the contractility of isolated rat jejunum through organ-bath experiments. We observed that venom can induce both contraction and relaxation. The contraction was partially inhibited by atropine (1 μM) and by suramin (0.1 mM) through tetrodotoxin-resistant and sensitive mechanisms. The relaxation was completely inhibited by 3 μM propranolol and partially inhibited by 1 μM phentolamine. Suramin induced a slowing of relaxation curve. Tetrodotoxin completely inhibits the relaxation induced by Tityus serrulatus venom, but the contraction curves were only partially reduced in their initial portion. The final part of the curve was largely enhanced by Tetrodotoxin. Atropine blocks almost completely the contraction curve in the presence of Tetrodotoxin. These results indicate that Tityus serrulatus venom induces the release of both excitatory (predominantly acetylcholine) and inhibitory (mainly noradrenaline) neurotransmitters. The effects of Tityus serrulatus venom on organ contractility was quite complex and seem to derive from a diffuse and nonspecific release of mediators from autonomic and enteric nervous systems. Further investigation of venom action and its isolated toxins can reveal important aspects to deepen our knowledge about the enteric nervous system transmission and the interaction between excitatory and inhibitory mediators as well as the physiological role of Na+ and K+ ion channels in gut motility.
Collapse
Affiliation(s)
- Gisele S G Louza
- Unit of Mode of Toxin Action (MATx), Laboratory of Pharmacology, Butantan Institute, São Paulo, Brazil; Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | | |
Collapse
|
15
|
Guerra DD, Bok R, Lorca RA, Hurt KJ. Protein kinase A facilitates relaxation of mouse ileum via phosphorylation of neuronal nitric oxide synthase. Br J Pharmacol 2020; 177:2765-2778. [PMID: 31975425 DOI: 10.1111/bph.15001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/03/2020] [Accepted: 01/18/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The enteric neurotransmitter nitric oxide (NO) regulates gastrointestinal motility by relaxing smooth muscle. Pharmacological cAMP induction also relaxes gastrointestinal smooth muscle, but it is uncertain whether cAMP augments or suppresses enteric NO signalling. In other organ systems, cAMP can increase neuronal NO production by stimulating protein kinase A (PKA) to phosphorylate neuronal NOS (nNOS) Serine-1412 (S1412). We hypothesized that cAMP also increases nNOS S1412 phosphorylation by PKA in enteric neurons to augment nitrergic relaxation of mouse ileum. EXPERIMENTAL APPROACH We measured contractile force and nNOS S1412 phosphorylation in ileal rings suspended in an organ bath. We used forskolin to induce cAMP-dependent relaxation of wild type, nNOSS1412A knock-in and nNOSα-null ileal rings in the presence or absence of PKA, protein kinase B (Akt) and NOS inhibitors. KEY RESULTS Forskolin stimulated phosphorylation of nNOS S1412 in mouse ileum. Forskolin relaxed nNOSα-null and nNOSS1412A ileal rings less than wild-type ileal rings. PKA inhibition blocked forskolin-induced nNOS phosphorylation and attenuated relaxation of wild type but not nNOSS1412A ileum. Akt inhibition did not alter nNOS phosphorylation with forskolin but did attenuate relaxation of wild type and nNOSS1412A . NOS inhibition with L-NAME eliminated the effects of PKA and Akt inhibitors on relaxation. CONCLUSION AND IMPLICATIONS PKA phosphorylation of nNOS S1412 augments forskolin-induced nitrergic ileal relaxation. The relationship between cAMP/PKA and NO is therefore synergistic in enteric nitrergic neurons. Because NO regulates gut motility, selective modulation of enteric neuronal cAMP synthesis may be useful for the treatment of gastrointestinal motility disorders.
Collapse
Affiliation(s)
- Damian D Guerra
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Rachael Bok
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - K Joseph Hurt
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
16
|
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
|
17
|
Delvalle NM, Fried DE, Rivera-Lopez G, Gaudette L, Gulbransen BD. Cholinergic activation of enteric glia is a physiological mechanism that contributes to the regulation of gastrointestinal motility. Am J Physiol Gastrointest Liver Physiol 2018; 315:G473-G483. [PMID: 29927320 PMCID: PMC6230698 DOI: 10.1152/ajpgi.00155.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The reflexive activities of the gastrointestinal tract are regulated, in part, by precise interactions between neurons and glia in the enteric nervous system (ENS). Intraganglionic enteric glia are a unique type of peripheral glia that surround enteric neurons and regulate neuronal function, activity, and survival. Enteric glia express numerous neurotransmitter receptors that allow them to sense neuronal activity, but it is not clear if enteric glia monitor acetylcholine (ACh), the primary excitatory neurotransmitter in the ENS. Here, we tested the hypothesis that enteric glia detect ACh and that glial activation by ACh contributes to the physiological regulation of gut functions. Our results show that myenteric enteric glia express both the M3 and M5 subtypes of muscarinic receptors (MRs) and that muscarine drives intracellular calcium (Ca2+) signaling predominantly through M3R activation. To elucidate the functional effects of activation of glial M3Rs, we used GFAP::hM3Dq mice that express a modified human M3R (hM3Dq) exclusively on glial fibrillary acidic protein (GFAP) positive glia to directly activate glial hM3Dqs using clozapine- N-oxide. Using spatiotemporal mapping analysis, we found that the activation of glial hM3Dq receptors enhances motility reflexes ex vivo. Continuous stimulation of hM3Dq receptors in vivo, drove changes in gastrointestinal motility without affecting neuronal survival in the ENS and glial muscarinic receptor activation did not alter neuron survival in vitro. Our results provide the first evidence that GFAP intraganglionic enteric glia express functional muscarinic receptors and suggest that the activation of glial muscarinic receptors contributes to the physiological regulation of functions. NEW & NOTEWORTHY Enteric glia are emerging as novel regulators of enteric reflex circuits, but little is still known regarding the effects of specific transmitter pathways on glia and the resulting consequences on enteric reflexes. Here, we provide the first evidence that enteric glia monitor acetylcholine in the enteric nervous system and that glial activation by acetylcholine is a physiological mechanism that contributes to the functional regulation of intestinal reflexes.
Collapse
Affiliation(s)
| | - David E. Fried
- 2Department of Physiology, Michigan State University, East Lansing, Michigan
| | | | - Luke Gaudette
- 1Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Brian D. Gulbransen
- 1Neuroscience Program, Michigan State University, East Lansing, Michigan,2Department of Physiology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
18
|
Valès S, Touvron M, Van Landeghem L. Enteric glia: Diversity or plasticity? Brain Res 2018; 1693:140-145. [PMID: 29425908 DOI: 10.1016/j.brainres.2018.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/27/2018] [Accepted: 02/01/2018] [Indexed: 01/16/2023]
Abstract
Glial cells of the enteric nervous system correspond to a unique glial lineage distinct from other central and peripheral glia, and form a vast and abundant network spreading throughout all the layers of the gastrointestinal wall. Research over the last two decades has demonstrated that enteric glia regulates all major gastrointestinal functions via multiple bi-directional crosstalk with enteric neurons and other neighboring cell types. Recent studies propose that enteric glia represents a heterogeneous population associated with distinct localization within the gut wall, phenotype and activity. Compelling evidence also indicates that enteric glial cells are capable of plasticity leading to phenotypic changes whose pinnacle so far has been shown to be the generation of enteric neurons. While alterations of the glial network have been heavily incriminated in the development of gastrointestinal pathologies, enteric glial cells have also recently emerged as an active player in gut-brain signaling. Therefore, the development of tools and techniques to better appraise enteric glia heterogeneity and plasticity will undoubtedly unveil critical regulatory mechanisms implicated in gut health and disease, as well as disorders of the gut-brain axis.
Collapse
Affiliation(s)
- Simon Valès
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, 1 rue Gaston Veil, 44035 Nantes Cedex, France.
| | - Melissa Touvron
- Department of Molecular Biomedical Sciences, North Carolina State University, College of Veterinary Medicine, CVM Main Building, Campus Box #8401, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| | - Laurianne Van Landeghem
- Department of Molecular Biomedical Sciences, North Carolina State University, College of Veterinary Medicine, CVM Main Building, Campus Box #8401, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| |
Collapse
|
19
|
Hibberd TJ, Feng J, Luo J, Yang P, Samineni VK, Gereau RW, Kelley N, Hu H, Spencer NJ. Optogenetic Induction of Colonic Motility in Mice. Gastroenterology 2018; 155:514-528.e6. [PMID: 29782847 PMCID: PMC6715392 DOI: 10.1053/j.gastro.2018.05.029] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 04/12/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND & AIMS Strategies are needed to increase gastrointestinal transit without systemic pharmacologic agents. We investigated whether optogenetics, focal application of light to control enteric nervous system excitability, could be used to evoke propagating contractions and increase colonic transit in mice. METHODS We generated transgenic mice with Cre-mediated expression of light-sensitive channelrhodopsin-2 (ChR2) in calretinin neurons (CAL-ChR2 Cre+ mice); Cre- littermates served as controls. Colonic myenteric neurons were analyzed by immunohistochemistry, patch-clamp, and calcium imaging studies. Motility was assessed by mechanical, electrophysiological, and video recording in vitro and by fecal output in vivo. RESULTS In isolated colons, focal light stimulation of calretinin enteric neurons evoked classic polarized motor reflexes (50/58 stimulations), followed by premature anterograde propagating contractions (39/58 stimulations). Light stimulation could evoke motility from sites along the entire colon. These effects were prevented by neural blockade with tetrodotoxin (n = 2), and did not occur in control mice (n = 5). Light stimulation of proximal colon increased the proportion of natural fecal pellets expelled over 15 minutes in vitro (75% ± 17% vs 32% ± 8% for controls) (P < .05). In vivo, activation of wireless light-emitting diodes implanted onto the colon wall significantly increased hourly fecal pellet output in conscious, freely moving mice (4.2 ± 0.4 vs 1.3 ± 0.3 in controls) (P < .001). CONCLUSIONS In studies of mice, we found that focal activation of a subset of enteric neurons can increase motility of the entire colon in vitro, and fecal output in vivo. Optogenetic control of enteric neurons might therefore be used to modify gut motility.
Collapse
Affiliation(s)
- Timothy J Hibberd
- College of Medicine and Public Health & Centre for Neuroscience, Flinders University, Australia
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| | - Jialie Luo
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| | - Pu Yang
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| | - Vijay K Samineni
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Robert W Gereau
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Nigel Kelley
- SA Biomedical Engineering, SA Health, Government of South Australia, Australia
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St Louis, Missouri.
| | - Nick J Spencer
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, Australia.
| |
Collapse
|
20
|
Grubišić V, Verkhratsky A, Zorec R, Parpura V. Enteric glia regulate gut motility in health and disease. Brain Res Bull 2018; 136:109-117. [PMID: 28363846 PMCID: PMC5620110 DOI: 10.1016/j.brainresbull.2017.03.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/20/2017] [Accepted: 03/28/2017] [Indexed: 12/16/2022]
Abstract
The enteric nervous system, often referred to as the second brain, is the largest assembly of neurons and glia outside the central nervous system. The enteric nervous system resides within the wall of the digestive tract and regulates local gut reflexes involved in gastrointestinal motility and fluid transport; these functions can be accomplished in the absence of the extrinsic innervation from the central nervous system. It is neurons and their circuitry within the enteric nervous system that govern the gut reflexes. However, it is becoming clear that enteric glial cells are also actively involved in this process through the bidirectional signaling with neurons and other cells in the gut wall. We synthesize the recently discovered modulatory roles of enteric gliotransmission in gut motility and provide our perspective for future lines of research.
Collapse
Affiliation(s)
- Vladimir Grubišić
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA; Neuroscience Program, Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824, USA
| | - Alexei Verkhratsky
- The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology University of Ljubljana, Ljubljana, Slovenia; Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
21
|
Sánchez M, Suárez L, Andrés MT, Flórez BH, Bordallo J, Riestra S, Cantabrana B. Modulatory effect of intestinal polyamines and trace amines on the spontaneous phasic contractions of the isolated ileum and colon rings of mice. Food Nutr Res 2017; 61:1321948. [PMID: 28659731 PMCID: PMC5475348 DOI: 10.1080/16546628.2017.1321948] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/20/2017] [Indexed: 11/08/2022] Open
Abstract
Background: Gastrointestinal motility modulatory factors include substances of the intestinal content, such as polyamines and trace amines (TAs), the focus of this study. Methods: The amines of food, intestinal content and from faecal bacteria of Swiss mice were determined by HPLC and functionally characterised in isolated distal ileum and medial colon rings. Results: Mouse food and intestinal content contain polyamines (spermidine>putrescine>spermine) and TAs (isoamylamine>cadaverine). Intestinal bacteria mainly produce putrescine and cadaverine. The amines inhibited the spontaneous motility of the ileum (0.1-3 mM) and colon rings (0.01-3 mM, with lower IC50), with: spermine~isoamylamine~spermidine. Spermine inhibition was tetrodotoxin (TTX)-insensitive, while isoamylamine was TTX-sensitive, suggesting neural control. Mainly in the ileum, isoamylamine (3 mM) elicited acute effects modified by TTX, atropine and propranolol, and suppressed by spermine (3 mM), not being localized at the smooth muscle level. The amines assayed (3 mM), except putrescine and cadaverine in the ileum and isoamylamine in the colon, antagonised acetylcholine (ACh, 0.1 mM)-elicited phasic contractions. Isoamylamine and spermine in colon relaxed KCl (100 mM)-elicited tonic contractions, suggesting an effect on smooth muscle, but did not justify the suppression of motility caused by spermine and isoamylamine. Conclusions: Polyamines and TAs of the intestinal content might act on chemosensors and modulate intestinal peristalsis.
Collapse
Affiliation(s)
- Manuel Sánchez
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Lorena Suárez
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - María Teresa Andrés
- Laboratorio de Microbiología Oral, Escuela de Estomatología, Universidad de Oviedo, Oviedo, Spain
| | - Blanca Henar Flórez
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Javier Bordallo
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Sabino Riestra
- Servicio de Aparato Digestivo, Unidad de Enfermedad Inflamatoria Intestinal, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Begoña Cantabrana
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| |
Collapse
|
22
|
Chow AK, Gulbransen BD. Potential roles of enteric glia in bridging neuroimmune communication in the gut. Am J Physiol Gastrointest Liver Physiol 2017; 312:G145-G152. [PMID: 28039160 PMCID: PMC5338608 DOI: 10.1152/ajpgi.00384.2016] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 01/31/2023]
Abstract
The enteric nervous system (ENS) is a network of neurons and glia that controls ongoing gastrointestinal (GI) functions. Damage or injury to the ENS can lead to functional GI disorders. Current data support the conclusion that many functional GI disorders are caused by an imbalance between gut microbes and the immune system, but how the ENS is involved in these interactions is less understood. Because of the proximity of the ENS to bacteria and other foreign antigens in the GI tract, it is important to prevent the passage of these antigens through the GI epithelium. If any foreign compounds manage to pass through the GI epithelium, an immune response is triggered to prevent injury to the ENS and underlying structures. However, careful modulation of the inflammatory response is required to allow for adequate elimination of foreign antigens while avoiding inappropriate overactivation of the immune system as in autoimmune disorders. Enteric neurons and glial cells are capable of performing these immunomodulatory functions to provide adequate protection to the ENS. We review recent studies examining the interactions between the ENS and the immune system, with specific focus on enteric glial cells and their ability to modulate inflammation in the ENS.
Collapse
Affiliation(s)
- Aaron K. Chow
- 1Department of Physiology, Michigan State University, East Lansing, Michigan; and
| | - Brian D. Gulbransen
- 1Department of Physiology, Michigan State University, East Lansing, Michigan; and ,2Neuroscience Program, Michigan State University, East Lansing, Michigan
| |
Collapse
|
23
|
Grubišić V, Gulbransen BD. Enteric glia: the most alimentary of all glia. J Physiol 2017; 595:557-570. [PMID: 27106597 PMCID: PMC5233670 DOI: 10.1113/jp271021] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
Glia (from Greek γλοία meaning 'glue') pertains to non-neuronal cells in the central (CNS) and peripheral nervous system (PNS) that nourish neurons and maintain homeostasis. In addition, glia are now increasingly appreciated as active regulators of numerous physiological processes initially considered exclusively under neuronal regulation. For instance, enteric glia, a collection of glial cells residing within the walls of the intestinal tract, regulate intestinal motility, a well-characterized reflex controlled by enteric neurons. Enteric glia also interact with various non-neuronal cell types in the gut wall such as enterocytes, enteroendocrine and immune cells and are therefore emerging as important local regulators of diverse gut functions. The intricate molecular mechanisms that govern glia-mediated regulation are beginning to be discovered, but much remains unknown about the functions of enteric glia in health and disease. Here we present a current view of the enteric glia and their regulatory roles in gastrointestinal (GI) (patho)physiology; from GI motility and epithelial barrier function to enteric neuroinflammation.
Collapse
Affiliation(s)
- Vladimir Grubišić
- Neuroscience Program, Department of PhysiologyMichigan State University567 Wilson RoadEast LansingMI48824USA
| | - Brian D. Gulbransen
- Neuroscience Program, Department of PhysiologyMichigan State University567 Wilson RoadEast LansingMI48824USA
| |
Collapse
|
24
|
da Cunha Franceschi R, Nardin P, Machado CV, Tortorelli LS, Martinez-Pereira MA, Zanotto C, Gonçalves CA, Zancan DM. Enteric glial reactivity to systemic LPS administration: Changes in GFAP and S100B protein. Neurosci Res 2017; 119:15-23. [PMID: 28063977 DOI: 10.1016/j.neures.2016.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/29/2016] [Accepted: 12/27/2016] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharide (LPS) is used to induce inflammation and promotes nervous system activation. Different regions of the brain present heterogeneous glial responses; thus, in order to verify whether systemic LPS-induced inflammation affects the enteric glia differently across the intestinal segments, we evaluated the expressions of two glial activity markers, GFAP and S100B protein, in different intestine segments, at 1h, 24h and 7days after acute systemic LPS administration (0.25 or 2.5mgkg-1) in rats. Histological inflammatory analysis indicated that the cecum was most affected when compared to the duodenum and proximal colon at the highest doses of LPS. LPS induced an increased S100B content after 24h in all three regions, which decreased at 7days after the highest dose in all regions. Moreover, at 24h, this dose of LPS increased ex-vivo S100B secretion only in the cecum. The highest dose of LPS also increased GFAP in all regions at 24h, but earlier in the cecum, where LPS-induced enteric S100B and GFAP alterations were dependent on dose, time and intestine region. No associated changes in serum S100B were observed. Our results indicate heterogeneous enteric glial responses to inflammatory insult, as observed in distinct brain areas.
Collapse
Affiliation(s)
- Raphaela da Cunha Franceschi
- Laboratory of Comparative Neurobiology, Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Graduate Program in Neuroscience, ICBS, UFRGS, Brazil
| | - Patrícia Nardin
- Laboratory of Calcium-Binding Proteins, Department of Biochemistry, ICBS, UFRGS, Brazil
| | - Clivia Valle Machado
- Laboratory of Comparative Neurobiology, Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Graduate Program in Neuroscience, ICBS, UFRGS, Brazil
| | | | | | - Caroline Zanotto
- Laboratory of Calcium-Binding Proteins, Department of Biochemistry, ICBS, UFRGS, Brazil
| | - Carlos-Alberto Gonçalves
- Graduate Program in Neuroscience, ICBS, UFRGS, Brazil; Laboratory of Calcium-Binding Proteins, Department of Biochemistry, ICBS, UFRGS, Brazil.
| | - Denise Maria Zancan
- Laboratory of Comparative Neurobiology, Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Graduate Program in Neuroscience, ICBS, UFRGS, Brazil
| |
Collapse
|
25
|
Reed DE, Zhang Y, Beyak MJ, Lourenssen S, Blennerhassett MG, Paterson WG, Vanner SJ. Stress increases descending inhibition in mouse and human colon. Neurogastroenterol Motil 2016; 28:569-80. [PMID: 26744175 DOI: 10.1111/nmo.12755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND A relationship between stress and the symptoms of irritable bowel syndrome (IBS) has been well established but the cellular mechanisms are poorly understood. Therefore, we investigated effects of stress and stress hormones on colonic descending inhibition and transit in mouse models and human tissues. METHODS Stress was applied using water avoidance stress (WAS) in the animal model or mimicked using stress hormones, adrenaline (5 nM), and corticosterone (1 μM). Intracellular recordings were obtained from colonic circular smooth muscle cells in isolated smooth muscle/myenteric plexus preparations and the inhibitory junction potential (IJP) was elicited by nerve stimulation or balloon distension oral to the site of recording. KEY RESULTS Water avoidance stress increased the number of fecal pellets compared to control (p < 0.05). WAS also caused a significant increase in IJP amplitude following balloon distension. Stress hormones also increased the IJP amplitude following nerve stimulation and balloon distension (p < 0.05) in control mice but had no effect in colons from stressed mice. No differences were observed with application of ATP between stress and control tissues, suggesting the actions of stress hormones were presynaptic. Stress hormones had a large effect in the nerve stimulated IJP in human colon (increased >50%). Immunohistochemical studies identified alpha and beta adrenergic receptor immunoreactivity on myenteric neurons in human colon. CONCLUSIONS & INFERENCES These studies suggest that WAS and stress hormones can signal via myenteric neurons to increase inhibitory neuromuscular transmission. This could lead to greater descending relaxation, decreased transit time, and subsequent diarrhea.
Collapse
Affiliation(s)
- D E Reed
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - Y Zhang
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - M J Beyak
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - S Lourenssen
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | | | - W G Paterson
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - S J Vanner
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| |
Collapse
|
26
|
Increased Noradrenergic Neurotransmission to a Pain Facilitatory Area of the Brain Is Implicated in Facilitation of Chronic Pain. Anesthesiology 2015; 123:642-53. [DOI: 10.1097/aln.0000000000000749] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Abstract
Background:
Noradrenaline reuptake inhibitors are known to produce analgesia through a spinal action but they also act in the brain. However, the action of noradrenaline on supraspinal pain control regions is understudied. The authors addressed the noradrenergic modulation of the dorsal reticular nucleus (DRt), a medullary pronociceptive area, in the spared nerve injury (SNI) model of neuropathic pain.
Methods:
The expression of the phosphorylated cAMP response element-binding protein (pCREB), a marker of neuronal activation, was evaluated in the locus coeruleus and A5 noradrenergic neurons (n = 6 rats/group). pCREB was studied in noradrenergic DRt-projecting neurons retrogradely labeled in SNI animals (n = 3). In vivo microdialysis was used to measure noradrenaline release in the DRt on nociceptive stimulation or after DRt infusion of clonidine (n = 5 to 6 per group). Pharmacology, immunohistochemistry, and western blot were used to study α-adrenoreceptors in the DRt (n = 4 to 6 per group).
Results:
pCREB expression significantly increased in the locus coeruleus and A5 of SNI animals, and most noradrenergic DRt-projecting neurons expressed pCREB. In SNI animals, noradrenaline levels significantly increased on pinprick (mean ± SD, 126 ± 14%; P = 0.025 vs. baseline) and acetone stimulation (mean ± SD, 151 ± 12%; P < 0.001 vs. baseline), and clonidine infusion showed decreased α2-mediated inhibitory function. α1-adrenoreceptor blockade decreased nociceptive behavioral responses in SNI animals. α2-adrenoreceptor expression was not altered.
Conclusions:
Chronic pain induces brainstem noradrenergic activation that enhances descending facilitation from the DRt. This suggests that antidepressants inhibiting noradrenaline reuptake may enhance pain facilitation from the brain, counteracting their analgesic effects at the spinal cord.
Collapse
|
27
|
Rao M, Nelms BD, Dong L, Salinas-Rios V, Rutlin M, Gershon MD, Corfas G. Enteric glia express proteolipid protein 1 and are a transcriptionally unique population of glia in the mammalian nervous system. Glia 2015; 63:2040-2057. [PMID: 26119414 DOI: 10.1002/glia.22876] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/26/2015] [Accepted: 06/02/2015] [Indexed: 12/27/2022]
Abstract
In the enteric nervous system (ENS), glia outnumber neurons by 4-fold and form an extensive network throughout the gastrointestinal tract. Growing evidence for the essential role of enteric glia in bowel function makes it imperative to understand better their molecular marker expression and how they relate to glia in the rest of the nervous system. We analyzed expression of markers of astrocytes and oligodendrocytes in the ENS and found, unexpectedly, that proteolipid protein 1 (PLP1) is specifically expressed by glia in adult mouse intestine. PLP1 and S100β are the markers most widely expressed by enteric glia, while glial fibrillary acidic protein expression is more restricted. Marker expression in addition to cellular location and morphology distinguishes a specific subpopulation of intramuscular enteric glia, suggesting that a combinatorial code of molecular markers can be used to identify distinct subtypes. To assess the similarity between enteric and extraenteric glia, we performed RNA sequencing analysis on PLP1-expressing cells in the mouse intestine and compared their gene expression pattern to that of other types of glia. This analysis shows that enteric glia are transcriptionally unique and distinct from other cell types in the nervous system. Enteric glia express many genes characteristic of the myelinating glia, Schwann cells and oligodendrocytes, although there is no evidence of myelination in the murine ENS. GLIA 2015;63:2040-2057.
Collapse
Affiliation(s)
- Meenakshi Rao
- F.M. Kirby Neurobiology Program, Boston Children's Hospital, Boston, Massachusetts.,Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Columbia University, New York
| | - Bradlee D Nelms
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts
| | - Lauren Dong
- Department of Pediatrics, Columbia University, New York
| | - Viviana Salinas-Rios
- F.M. Kirby Neurobiology Program, Boston Children's Hospital, Boston, Massachusetts
| | - Michael Rutlin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York
| | | | - Gabriel Corfas
- F.M. Kirby Neurobiology Program, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Harvard Medical School, Boston, Massachusetts.,Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
28
|
Wu X, Conlin VS, Morampudi V, Ryz NR, Nasser Y, Bhinder G, Bergstrom KS, Yu HB, Waterhouse CCM, Buchan AMJ, Popescu OE, Gibson WT, Waschek JA, Vallance BA, Jacobson K. Vasoactive intestinal polypeptide promotes intestinal barrier homeostasis and protection against colitis in mice. PLoS One 2015; 10:e0125225. [PMID: 25932952 PMCID: PMC4416880 DOI: 10.1371/journal.pone.0125225] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/22/2015] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease is a chronic gastrointestinal inflammatory disorder associated with changes in neuropeptide expression and function, including vasoactive intestinal peptide (VIP). VIP regulates intestinal vasomotor and secretomotor function and motility; however, VIP's role in development and maintenance of colonic epithelial barrier homeostasis is unclear. Using VIP deficient (VIPKO) mice, we investigated VIP's role in epithelial barrier homeostasis, and susceptibility to colitis. Colonic crypt morphology and epithelial barrier homeostasis were assessed in wildtype (WT) and VIPKO mice, at baseline. Colitic responses were evaluated following dinitrobenzene sulfonic acid (DNBS) or dextran-sodium sulfate (DSS) exposure. Mice were also treated with exogenous VIP. At baseline, VIPKO mice exhibited distorted colonic crypts, defects in epithelial cell proliferation and migration, increased apoptosis, and altered permeability. VIPKO mice also displayed reduced goblet cell numbers, and reduced expression of secreted goblet cell factors mucin 2 and trefoil factor 3. These changes were associated with reduced expression of caudal type homeobox 2 (Cdx2), a master regulator of intestinal function and homeostasis. DNBS and DSS-induced colitis were more severe in VIPKO than WT mice. VIP treatment rescued the phenotype, protecting VIPKO mice against DSS colitis, with results comparable to WT mice. In conclusion, VIP plays a crucial role in the development and maintenance of colonic epithelial barrier integrity under physiological conditions and promotes epithelial repair and homeostasis during colitis.
Collapse
Affiliation(s)
- Xiujuan Wu
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Victoria S. Conlin
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Vijay Morampudi
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Natasha R. Ryz
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Yasmin Nasser
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Ganive Bhinder
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Kirk S. Bergstrom
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, United States of America
| | - Hong B. Yu
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Oana E. Popescu
- Department of Pathology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - William T. Gibson
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - James A. Waschek
- Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, United States of America
| | - Bruce A. Vallance
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevan Jacobson
- Department of Pediatrics, Division of Gastroenterology, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
29
|
Abstract
Fat is a vital macronutrient, and its intake is closely monitored by an array of molecular sensors distributed throughout the alimentary canal. In the mouth, dietary fat constituents such as mono- and diunsaturated fatty acids give rise to taste signals that stimulate food intake, in part by enhancing the production of lipid-derived endocannabinoid messengers in the gut. As fat-containing chyme enters the small intestine, it causes the formation of anorexic lipid mediators, such as oleoylethanolamide, which promote satiety. These anatomically and functionally distinct responses may contribute to the homeostatic control and, possibly, the pathological dysregulation of food intake.
Collapse
Affiliation(s)
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology
- Department of Pharmacology, and
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA
- Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
30
|
Hetz S, Acikgoez A, Moll C, Jahnke HG, Robitzki AA, Metzger R, Metzger M. Age-related gene expression analysis in enteric ganglia of human colon after laser microdissection. Front Aging Neurosci 2014; 6:276. [PMID: 25360110 PMCID: PMC4197768 DOI: 10.3389/fnagi.2014.00276] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 09/24/2014] [Indexed: 01/16/2023] Open
Abstract
The enteric nervous system (ENS) poses the intrinsic innervation of the gastrointestinal tract and plays a critical role for all stages of postnatal life. There is increasing scientific and clinical interest in acquired or age-related gastrointestinal dysfunctions that can be manifested in diseases such as gut constipation or fecal incontinence. In this study, we sought to analyze age-dependent changes in the gene expression profile of the human ENS, particularly in the myenteric plexus. Therefore, we used the laser microdissection technique which has been proven as a feasible tool to analyze distinct cell populations within heterogeneously composed tissues. Full biopsy gut samples were prepared from children (4-12 months), middle aged (48-58 years) and aged donors (70-95 years). Cryosections were histologically stained with H&E, the ganglia of the myenteric plexus identified and RNA isolated using laser microdissection technique. Quantitative PCR was performed for selected neural genes, neurotransmitters and receptors. Data were confirmed on protein level using NADPH-diaphorase staining and immunohistochemistry. As result, we demonstrate age-associated alterations in site-specific gene expression pattern of the ENS. Thus, in the adult and aged distal parts of the colon a marked decrease in relative gene expression of neural key genes like NGFR, RET, NOS1 and a concurrent increase of CHAT were observed. Further, we detected notable regional differences of RET, CHAT, TH, and S100B comparing gene expression in aged proximal and distal colon. Interestingly, markers indicating cellular senescence or oxidative stress (SNCA, CASP3, CAT, SOD2, and TERT) were largely unchanged within the ENS. For the first time, our study also describes the age-dependent expression pattern of all major sodium channels within the ENS. Our results are in line with previous studies showing spatio-temporal differences within the mammalian ENS.
Collapse
Affiliation(s)
- Susan Hetz
- CELLT Research Group, Translational Centre for Regenerative Medicine, University of Leipzig Leipzig, Germany
| | - Ali Acikgoez
- Department of General and Visceral Surgery, St. George's Hospital Leipzig, Germany
| | - Corinna Moll
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg Wuerzburg, Germany
| | - Heinz-Georg Jahnke
- Division of Molecular biological-biochemical Processing Technology, Center for Biotechnology and Biomedicine (BBZ), University of Leipzig Leipzig, Germany
| | - Andrea A Robitzki
- Division of Molecular biological-biochemical Processing Technology, Center for Biotechnology and Biomedicine (BBZ), University of Leipzig Leipzig, Germany
| | - Roman Metzger
- Department of Pediatric Surgery, University of Leipzig Leipzig, Germany
| | - Marco Metzger
- CELLT Research Group, Translational Centre for Regenerative Medicine, University of Leipzig Leipzig, Germany ; Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg Wuerzburg, Germany
| |
Collapse
|
31
|
Boesmans W, Lasrado R, Vanden Berghe P, Pachnis V. Heterogeneity and phenotypic plasticity of glial cells in the mammalian enteric nervous system. Glia 2014; 63:229-41. [PMID: 25161129 DOI: 10.1002/glia.22746] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/05/2014] [Indexed: 12/24/2022]
Abstract
Enteric glial cells are vital for the autonomic control of gastrointestinal homeostasis by the enteric nervous system. Several different functions have been assigned to enteric glial cells but whether these are performed by specialized subtypes with a distinctive phenotype and function remains elusive. We used Mosaic Analysis with Double Markers and inducible lineage tracing to characterize the morphology and dynamic molecular marker expression of enteric GLIA in the myenteric plexus. Functional analysis in individually identified enteric glia was performed by Ca(2+) imaging. Our experiments have identified four morphologically distinct subpopulations of enteric glia in the gastrointestinal tract of adult mice. Marker expression analysis showed that the majority of glia in the myenteric plexus co-express glial fibrillary acidic protein (GFAP), S100β, and Sox10. However, a considerable fraction (up to 80%) of glia outside the myenteric ganglia, did not label for these markers. Lineage tracing experiments suggest that these alternative combinations of markers reflect dynamic gene regulation rather than lineage restrictions. At the functional level, the three myenteric glia subtypes can be distinguished by their differential response to adenosine triphosphate. Together, our studies reveal extensive heterogeneity and phenotypic plasticity of enteric glial cells and set a framework for further investigations aimed at deciphering their role in digestive function and disease.
Collapse
Affiliation(s)
- Werend Boesmans
- Laboratory for Enteric NeuroScience (LENS), TARGID, Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
32
|
Clairembault T, Leclair-Visonneau L, Neunlist M, Derkinderen P. Enteric glial cells: new players in Parkinson's disease? Mov Disord 2014; 30:494-8. [PMID: 25100667 DOI: 10.1002/mds.25979] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/14/2014] [Accepted: 07/17/2014] [Indexed: 12/14/2022] Open
Abstract
Lewy pathology has been described in neurons of the enteric nervous system in nearly all Parkinson's disease (PD) patients at autopsy. The enteric nervous system not only contains a variety of functionally distinct enteric neurons but also harbors a prominent component of glial cells, the so-called enteric glial cells, which, like astrocytes of the central nervous system, contribute to support, protect, and maintain the neural network. A growing body of evidence supports a role for enteric glial cells in the pathophysiology of gastrointestinal disorders such as inflammatory bowel disease and chronic constipation. We have recently shown that enteric glial cell dysfunction occurs in PD. In the present review, we discuss the possible implications of enteric glia in PD-related gut dysfunction as well as in disease initiation and development.
Collapse
Affiliation(s)
- Thomas Clairembault
- Inserm, U913, Nantes, F-44093, France; University Nantes, Nantes, F-44093, France; CHU Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, F-44093, France
| | | | | | | |
Collapse
|
33
|
Song J, Zheng L, Zhang X, Feng X, Fan R, Sun L, Hong F, Zhang Y, Zhu J. Upregulation of β1-adrenoceptors is involved in the formation of gastric dysmotility in the 6-hydroxydopamine rat model of Parkinson's disease. Transl Res 2014; 164:22-31. [PMID: 24467967 DOI: 10.1016/j.trsl.2014.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 01/01/2014] [Accepted: 01/03/2014] [Indexed: 11/16/2022]
Abstract
Gastrointestinal dysmotility is one of the nonmotor symptoms of Parkinson's disease (PD). Gastroparesis and upregulated β-adrenoceptors (β-ARs) have been reported in rats with bilateral microinjection of 6-hydroxydopamine (6-OHDA) in the substantia nigra, but the underlying mechanism is unclear. The aim of the current study is to investigate the role of β-ARs in gastroparesis in 6-OHDA rats. Gastric motility was studied through strain gauge measurement. Immunofluorescence, real-time reverse transcription-polymerase chain reaction and Western blotting were performed to examine the expression of β-ARs. Norepinephrine (NE) inhibited gastric motility in a dose-dependent fashion in both control and 6-OHDA rats, but much stronger adrenergic reactivity was observed in the 6-OHDA rats. The inhibition of gastric motility by NE in both control and 6-OHDA rats was not affected by tetrodotoxin, a neural sodium channel blocker. Blocking β1-AR or β2-AR did not affect the inhibition of strip contraction by NE in control rats, but β1-AR blockage obviously enhanced the half maximal inhibitory concentration value of NE in 6-OHDA rats. Selective inhibition of β3-AR blocked the effect of NE significantly in both control and 6-OHDA rats. The protein expression of β1-AR, but not β2-AR and β3-AR in gastric muscularis externa was increased significantly in 6-OHDA rats. In conclusion, β3-AR involves the regulation of gastric motility in control rats, whereas the upregulation of β1-AR is responsible for enhanced NE reactivity in 6-OHDA rats and therefore is involved in the formation of gastroparesis. The effect of both β1-AR and β3-AR on gastric motility is independent of the enteric nervous system.
Collapse
Affiliation(s)
- Jin Song
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lifei Zheng
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaoli Zhang
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaoyan Feng
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ruifang Fan
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lu Sun
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Feng Hong
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yue Zhang
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jinxia Zhu
- Gastrointestinal Research Group, Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
34
|
Campos-Rodríguez R, Godínez-Victoria M, Abarca-Rojano E, Pacheco-Yépez J, Reyna-Garfias H, Barbosa-Cabrera RE, Drago-Serrano ME. Stress modulates intestinal secretory immunoglobulin A. Front Integr Neurosci 2013; 7:86. [PMID: 24348350 PMCID: PMC3845795 DOI: 10.3389/fnint.2013.00086] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 11/12/2013] [Indexed: 01/08/2023] Open
Abstract
Stress is a response of the central nervous system to environmental stimuli perceived as a threat to homeostasis. The stress response triggers the generation of neurotransmitters and hormones from the hypothalamus pituitary adrenal axis, sympathetic axis and brain gut axis, and in this way modulates the intestinal immune system. The effects of psychological stress on intestinal immunity have been investigated mostly with the restraint/immobilization rodent model, resulting in an up or down modulation of SIgA levels depending on the intensity and time of exposure to stress. SIgA is a protein complex formed by dimeric (dIgA) or polymeric IgA (pIgA) and the secretory component (SC), a peptide derived from the polymeric immunoglobulin receptor (pIgR). The latter receptor is a transmembrane protein expressed on the basolateral side of gut epithelial cells, where it uptakes dIgA or pIgA released by plasma cells in the lamina propria. As a result, the IgA-pIgR complex is formed and transported by vesicles to the apical side of epithelial cells. pIgR is then cleaved to release SIgA into the luminal secretions of gut. Down modulation of SIgA associated with stress can have negative repercussions on intestinal function and integrity. This can take the form of increased adhesion of pathogenic agents to the intestinal epithelium and/or an altered balance of inflammation leading to greater intestinal permeability. Most studies on the molecular and biochemical mechanisms involved in the stress response have focused on systemic immunity. The present review analyzes the impact of stress (mostly by restraint/immobilization, but also with mention of other models) on the generation of SIgA, pIgR and other humoral and cellular components involved in the intestinal immune response. Insights into these mechanisms could lead to better therapies for protecting against pathogenic agents and avoiding epithelial tissue damage by modulating intestinal inflammation.
Collapse
Affiliation(s)
- Rafael Campos-Rodríguez
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico NacionalDistrito Federal, México
| | - Marycarmen Godínez-Victoria
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico NacionalDistrito Federal, México
| | - Edgar Abarca-Rojano
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico NacionalDistrito Federal, México
| | - Judith Pacheco-Yépez
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico NacionalDistrito Federal, México
| | - Humberto Reyna-Garfias
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico NacionalDistrito Federal, México
| | | | - Maria Elisa Drago-Serrano
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico NacionalDistrito Federal, México
| |
Collapse
|
35
|
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
|
36
|
Goetz B, Benhaqi P, Müller MH, Kreis ME, Kasparek MS. Changes in beta-adrenergic neurotransmission during postoperative ileus in rat circular jejunal muscle. Neurogastroenterol Motil 2013; 25:154-e84. [PMID: 23009554 DOI: 10.1111/nmo.12020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND To explore postoperative changes in β-adrenergic neurotransmission that participate in pathophysiology of postoperative ileus. METHODS Contractile activity of circular jejunal muscle strips was studied. Groups (n = 6/group) were: naïve controls, sham controls 1 and 7 days after laparotomy, and rats 12 h, 1, 3, and 7 days after laparotomy with standardized small bowel manipulation (postoperative ileus). Dose-responses to the β-agonist isoprenaline (3 × 10(-10) - 10(-7) mol L(-1)) were studied in presence/absence of tetrodotoxin (global neural blockade; 10(-6) mol L(-1) ), N6-(1-iminoethyl)-l-lysine (inhibition of inducible nitric oxide synthesis; 10(-4) mol L(-1)), nimesulide (cyclooxygenase-2 inhibition; 10(-5) mol L(-1)), or propranolol (β-blockade; 5 × 10(-6) mol L(-1)). Histochemistry for inflammatory cells and intestinal transit were studied. KEY RESULTS Intramural inflammation and delayed transit (postoperative ileus) occurred only in ileus groups. The inhibitory potential of isoprenaline decreased in all postoperative groups including sham (P < 0.05). Tetrodotoxin enhanced isoprenaline-induced inhibition in ileus and sham groups (P < 0.05). N6-(1-iminoethyl)-l-lysine and nimesulide decreased isoprenaline-induced inhibition in ileus groups 12 h, 1, and 7 days, and in sham controls 7 days postoperatively (P < 0.05). Propranolol prevented isoprenaline effects in all groups (P < 0.05). CONCLUSIONS & INFERENCES Inhibitory effects of isoprenaline on contractile activity were decreased for 7 days postoperatively. Changes in β-adrenergic neurotransmission do not induce postoperative ileus and appear to be caused by anesthesia and laparotomy rather than bowel manipulation.
Collapse
Affiliation(s)
- B Goetz
- Walter Brendel Center of Experimental Medicine, Munich, Germany
| | | | | | | | | |
Collapse
|
37
|
Boesmans W, Cirillo C, Van den Abbeel V, Van den Haute C, Depoortere I, Tack J, Vanden Berghe P. Neurotransmitters involved in fast excitatory neurotransmission directly activate enteric glial cells. Neurogastroenterol Motil 2013; 25:e151-60. [PMID: 23279281 DOI: 10.1111/nmo.12065] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND The intimate association between glial cells and neurons within the enteric nervous system has confounded careful examination of the direct responsiveness of enteric glia to different neuroligands. Therefore, we aimed to investigate whether neurotransmitters known to elicit fast excitatory potentials in enteric nerves also activate enteric glia directly. METHODS We studied the effect of acetylcholine (ACh), serotonin (5-HT), and adenosine triphosphate (ATP) on intracellular Ca(2+) signaling using aequorin-expressing and Fluo-4 AM-loaded CRL-2690 rat and human enteric glial cell cultures devoid of neurons. The influence of these neurotransmitters on the proliferation of glia was measured and their effect on the expression of c-Fos as well as glial fibrillary acidic protein (GFAP), Sox10, and S100 was examined by immunohistochemistry and quantitative RT-PCR. KEY RESULTS Apart from ATP, also ACh and 5-HT induced a dose-dependent increase in intracellular Ca(2+) concentration in CRL-2690 cells. Similarly, these neurotransmitters also evoked Ca(2+) transients in human primary enteric glial cells obtained from mucosal biopsies. In contrast with ATP, stimulation with ACh and 5-HT induced early gene expression in CRL-2690 cells. The proliferation of enteric glia and their expression of GFAP, Sox10, and S100 were not affected following stimulation with these neurotransmitters. CONCLUSIONS & INFERENCES We provide evidence that enteric glial cells respond to fast excitatory neurotransmitters by changes in intracellular Ca(2+). On the basis of our experimental in vitro setting, we show that enteric glia are not only directly responsive to purinergic but also to serotonergic and cholinergic signaling mechanisms.
Collapse
Affiliation(s)
- W Boesmans
- Laboratory of Enteric NeuroScience, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
38
|
Gulbransen BD, Sharkey KA. Novel functional roles for enteric glia in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2012; 9:625-32. [PMID: 22890111 DOI: 10.1038/nrgastro.2012.138] [Citation(s) in RCA: 271] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Enteric glia are a unique class of peripheral glial cells within the gastrointestinal tract. Major populations of enteric glia are found in enteric ganglia in the myenteric and submucosal plexuses of the enteric nervous system (ENS); these cells are also found outside of the ENS, within the circular muscle and in the lamina propria of the mucosa. These different populations of cells probably represent unique classes of glial cells with differing functions. In the past few years, enteric glia have been found to be involved in almost every gut function including motility, mucosal secretion and host defence. Subepithelial glia seem to have a trophic and supporting relationship with intestinal epithelial cells, but the necessity of these roles in the maintenance of normal epithelial functions remains to be shown. Likewise, glia within enteric ganglia are activated by synaptic stimulation, suggesting an active role in synaptic transmission, but the precise role of glial activation in normal enteric network activity is unclear. Excitingly, enteric glia can also give rise to new neurons, but seemingly only under limited circumstances. In this Review, we discuss the current body of evidence supporting functional roles of enteric glia and identify key gaps in our understanding of the physiology of these unique cells.
Collapse
Affiliation(s)
- Brian D Gulbransen
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive North West Calgary, AB T2N 4N1, Canada
| | | |
Collapse
|
39
|
FGF2 deficit during development leads to specific neuronal cell loss in the enteric nervous system. Histochem Cell Biol 2012; 139:47-57. [PMID: 22955838 DOI: 10.1007/s00418-012-1023-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2012] [Indexed: 10/27/2022]
Abstract
The largest part of the peripheral nervous system is the enteric nervous system (ENS). It consists of an intricate network of several enteric neuronal subclasses with distinct phenotypes and functions within the gut wall. The generation of these enteric phenotypes is dependent upon appropriate neurotrophic support during development. Glial cell line-derived neurotrophic factor (GDNF) and fibroblast growth factor-2 (FGF2) play an important role in the differentiation and function of the ENS. A lack of GDNF or its receptor (Ret) causes intestinal aganglionosis in mice, while fibroblast growth factor receptor signaling antagonist is identified as regulating proteins in the GDNF/Ret signaling in the developing ENS. Primary myenteric plexus cultures and wholemount preparations of wild type (WT) and FGF2-knockout mice were used to analyze distinct enteric subpopulations. Fractal dimension (D) as a measure of self-similarity is an excellent tool to analyze complex geometric shape and was applied to classify the subclasses of enteric neurons concerning their individual morphology. As a consequence of a detailed analysis of subpopulation variations, wholemount preparations were stained for the calcium binding proteins calbindin and calretinin. The fractal analysis showed a reliable consistence of subgroups with different fractal dimensions (D) in each culture investigated. Seven different neuronal subtypes could be differentiated according to a rising D. Within the same D, the neurite length revealed significant differences between wild type and FGF2-knockout cultures, while the subclass distribution was also altered. Depending on the morphological characteristics, the reduced subgroup was supposed to be a secretomotor neuronal type, which could be confirmed by calbindin and calretinin staining of the wholemount preparations. These revealed a reduction up to 40 % of calbindin-positive neurons in the FGF2-knockout mouse. We therefore consider FGF2 playing a more important role in the fine-tuning of the ENS during development as previously assumed.
Collapse
|
40
|
Serres F, Rodriguez M, Rivet JM, Galizzi JP, Lockhart B, Sharp T, Millan MJ. Blockade of α2-adrenoceptors induces Arc gene expression in rat brain in a glutamate receptor-dependent manner: a combined qPCR, in situ hybridisation and immunocytochemistry study. Neuropharmacology 2012; 63:992-1001. [PMID: 22828637 DOI: 10.1016/j.neuropharm.2012.06.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 06/27/2012] [Accepted: 06/30/2012] [Indexed: 10/28/2022]
Abstract
Studies of 5-HT-glutamate interactions suggest that activation of brain 5-HT(2A) receptors leads to an AMPA receptor-mediated induction of the immediate early (activity-dependent) gene, Arc (Arg3.1). In this respect, noradrenaline-glutamate interactions are poorly characterised. Here we investigated the influence on regional brain Arc gene expression of selective blockade of α(2)-adrenoceptors in rats. Several complementary techniques were used: qPCR (mRNA, discrete tissue punches), in situ hybridisation (mRNA, sections) and immunocytochemistry. The α(2)-adrenoceptor antagonist, RX 821002, dose-dependently and time-dependently (maximal effect 2 h) increased Arc mRNA levels as demonstrated both by qPCR and in situ hybridisation. The α(2)-adrenoceptor antagonist, atipamezole, also increased Arc mRNA in in situ hybridisation studies. Changes in Arc mRNA after RX 821002 were of similar magnitude in punches and intact tissue sections and region-specific, with effects being most pronounced in parietal cortex and caudate putamen, less robust in frontal cortex, and not detectable in hippocampal sub-regions. Both qPCR and in situ hybridisation studies demonstrated that RX 821002-induced Arc mRNA was blocked by the AMPA antagonist, GYKI 52466. Pretreatment with the NMDA antagonist MK 801 also prevented RX 821002-induced Arc mRNA, as did the mGluR5 antagonist MPEP, whilst the mGluR2/3 antagonist, LY341495, had no effect. Finally, immunocytochemical studies showed that RX 821002 increased Arc-immunoreactivity in cells in close apposition to α(2)-adrenoceptor-positive processes. Thus, employing three complementary techniques, these observations demonstrate that blockade of α(2)-adrenoceptors triggers brain expression of the immediate early gene, Arc, and that this effect involves the recruitment of AMPA, NMDA and mGluR5 but not mGluR2/3 glutamatergic receptors.
Collapse
Affiliation(s)
- Florence Serres
- University Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | | | | | | | | | | | | |
Collapse
|
41
|
Sympathetic activity controls fat-induced oleoylethanolamide signaling in small intestine. J Neurosci 2011; 31:5730-6. [PMID: 21490214 DOI: 10.1523/jneurosci.5668-10.2011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ingestion of dietary fat stimulates production of the small-intestinal satiety factors oleoylethanolamide (OEA) and N-palmitoyl-phosphatidylethanolamine (NPPE), which reduce food intake through a combination of local (OEA) and systemic (NPPE) actions. Previous studies have shown that sympathetic innervation of the gut is necessary for duodenal infusions of fat to induce satiety, suggesting that sympathetic activity may engage small-intestinal satiety signals such as OEA and NPPE. In the present study, we show that surgical resection of the sympathetic celiac-superior mesenteric ganglion complex, which sends projections to the upper gut, abolishes feeding-induced OEA production in rat small-intestinal cells. These effects are accounted for by suppression of OEA biosynthesis, and are mimicked by administration of the selective β2-adrenergic receptor antagonist ICI-118,551. We further show that sympathetic ganglionectomy or pharmacological blockade of β2-adrenergic receptors prevents NPPE release into the circulation. In addition, sympathetic ganglionectomy increases meal frequency and lowers satiety ratio, and these effects are corrected by pharmacological administration of OEA. The results suggest that sympathetic activity controls fat-induced satiety by enabling the coordinated production of local (OEA) and systemic (NPPE) satiety signals in the small intestine.
Collapse
|
42
|
The role of α – and β – adrenergic receptors in the spasmolytic effects on rat ileum of Petroselinum crispum Latifolum (parsley). ASIAN PAC J TROP MED 2010. [DOI: 10.1016/s1995-7645(10)60208-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
43
|
Antisecretory effects of neuropeptide Y in the mouse colon are region-specific and are lost in DSS-induced colitis. ACTA ACUST UNITED AC 2010; 165:138-45. [PMID: 20561896 DOI: 10.1016/j.regpep.2010.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/20/2010] [Accepted: 05/30/2010] [Indexed: 12/13/2022]
Abstract
Regulation of water movement in the gut is an important homeostatic event that is critical to normal intestinal function. We assessed the effect of neuropeptide Y (NPY) on epithelial ion transport in the normal and inflamed mouse colons. Colitis was induced by dextran sodium sulfate (DSS, 4% wt./vol.) administered in the drinking water for 5 days followed by 3 days of regular water. Segments of proximal and distal colons were excised and short-circuit current (I(SC)) was measured in Ussing chambers to assess net electrogenic active ion transport. NPY Y(1) receptor (Y(1)R) expression was measured by quantitative real-time PCR and immunohistochemistry. Challenge of distal colon from normal mice with NPY (10(-7)M) evoked a drop in I(SC) (51.4±9.1 μA/cm(2)), which was dependent on Cl(-) flux, was insensitive to neural blockade with tetrodotoxin and was mediated primarily through the Y(1)R. In contrast, the proximal colon was largely unresponsive to NPY, expressing ~ten-fold less Y(1)R mRNA compared to the distal colon. These findings confirm that specific regional regulation of ion transport occurs in the colon. Segments of proximal and distal colons from mice with DDS-induced colitis were virtually unresponsive to NPY, expressed less Y(1)R mRNA than tissues from control mice and displayed loss of Y(1)R protein expression in the colonic epithelium. This hypo-responsiveness to an antisecretory stimulus adds to the well-documented loss of responsiveness to pro-secretory agents during inflammation, attesting to a profound loss of control of active ion transport during enteric inflammatory disease.
Collapse
|
44
|
Ampatzis K, Dermon CR. Regional distribution and cellular localization of beta2-adrenoceptors in the adult zebrafish brain (Danio rerio). J Comp Neurol 2010; 518:1418-41. [PMID: 20187137 DOI: 10.1002/cne.22278] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The beta(2)-adrenergic receptors (ARs) are G-protein-coupled receptors that mediate the physiological responses to adrenaline and noradrenaline. The present study aimed to determine the regional distribution of beta(2)-ARs in the adult zebrafish (Danio rerio) brain by means of in vitro autoradiographic and immunohistochemical methods. The immunohistochemical localization of beta(2)-ARs, in agreement with the quantitative beta-adrenoceptor autoradiography, showed a wide distribution of beta(2)-ARs in the adult zebrafish brain. The cerebellum and the dorsal zone of periventricular hypothalamus exhibited the highest density of [(3)H]CGP-12177 binding sites and beta(2)-AR immunoreactivity. Neuronal cells strongly stained for beta(2)-ARs were found in the periventricular ventral telencephalic area, magnocellular and parvocellular superficial pretectal nuclei (PSm, PSp), occulomotor nucleus (NIII), locus coeruleus (LC), medial octavolateral nucleus (MON), magnocellular octaval nucleus (MaON) reticular formation (SRF, IMRF, IRF), and ganglionic cell layer of cerebellum. Interestingly, in most cases (NIII, LC, MON, MaON, SRF, IMRF, ganglionic cerebellar layer) beta(2)-ARs were colocalized with alpha(2A)-ARs in the same neuron, suggesting their interaction for mediating the physiological functions of nor/adrenaline. Moderate to low labeling of beta(2)-ARs was found in neurons in dorsal telencephalic area, optic tectum (TeO), torus semicircularis (TS), and periventricular gray zone of optic tectum (PGZ). In addition to neuronal, glial expression of beta(2)-ARs was found in astrocytic fibers located in the central gray and dorsal rhombencephalic midline, in close relation to the ventricle. The autoradiographic and immunohistochemical distribution pattern of beta(2)-ARs in the adult zebrafish brain further support the conserved profile of adrenergic/noradrenergic system through vertebrate brain evolution.
Collapse
|
45
|
Leptin-sensitive neurons in mouse preoptic area express alpha 1A- and alpha 2A-adrenergic receptor isoforms. Neurosci Lett 2010; 471:83-8. [PMID: 20080149 DOI: 10.1016/j.neulet.2010.01.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 12/22/2009] [Accepted: 01/08/2010] [Indexed: 01/08/2023]
Abstract
Leptin binding to its functional receptor stimulates the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling pathway, finally resulting in nuclear translocation of the phosphorylated STAT3 (P-STAT3). Systemic treatment with leptin (3mg/kg; intraperitoneal injection) induced the appearance of P-STAT3-immunoreactive cells in adult mouse preoptic area (POA). Here we show that the vast majority of leptin-responsive cells were located in medial POA (mPOA), followed by the median preoptic nucleus. Rare, scattered and weakly stained cells were found in ventromedial preoptic nucleus and lateral preoptic area. Co-localization studies disclosed that mPOA leptin-responsive cells were neurons, and that a large proportion expressed the alpha(1A)- and/or alpha(2A)-adrenergic receptor (AR) isoforms. Although understanding the functional relevance of leptin-responsive POA neurons requires further investigation, the finding that they bear alpha-ARs suggests that they may be targeted by the ascending noradrenergic system, which densely innervates the mPOA, and thus be involved in thermoregulation, arousal and/or the sleep-wake cycle.
Collapse
|
46
|
Casanova E, Guetg N, Vigot R, Seddik R, Julio-Pieper M, Hyland NP, Cryan JF, Gassmann M, Bettler B. A mouse model for visualization of GABA(B) receptors. Genesis 2010; 47:595-602. [PMID: 19603512 DOI: 10.1002/dvg.20535] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
GABA(B) receptors are the G-protein-coupled receptors for the neurotransmitter gamma-aminobutyric acid (GABA). Receptor subtypes are based on the subunit isoforms GABA(B1a) and GABA(B1b), which combine with GABA(B2) subunits to form heteromeric receptors. Here, we used a modified bacterial artificial chromosome (BAC) containing the GABA(B1) gene to generate transgenic mice expressing GABA(B1a) and GABA(B1b) subunits fused to the enhanced green fluorescence protein (eGFP). We demonstrate that the GABA(B1)-eGFP fusion proteins reproduce the cellular expression patterns of endogenous GABA(B1) proteins in the brain and in peripheral tissue. Crossing the GABA(B1)-eGFP BAC transgene into the GABA(B1) (-/-) background restores pre and postsynaptic GABA(B) functions, showing that the GABA(B1)-eGFP fusion proteins substitute for the lack of endogenous GABA(B1) proteins. Finally, we demonstrate that the GABA(B1)-eGFP fusion proteins replicate the temporal expression patterns of native GABA(B) receptors in cultured neurons. These transgenic mice therefore provide a validated tool for direct visualization of native GABA(B) receptors.
Collapse
Affiliation(s)
- Emilio Casanova
- Department of Biomedicine, Institute of Physiology, University of Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lomax AE, Sharkey KA, Furness JB. The participation of the sympathetic innervation of the gastrointestinal tract in disease states. Neurogastroenterol Motil 2010; 22:7-18. [PMID: 19686308 DOI: 10.1111/j.1365-2982.2009.01381.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Knowledge of neural circuits, neurotransmitters and receptors involved in the sympathetic regulation of gastrointestinal (GI) function is well established. However, it is only recently that the interaction of sympathetic neurons, and of sympathetic transmitters, with the GI immune system and with gut flora has begun to be explored. Changes in the behaviour of sympathetic nerves when gut function is compromised, for example in ileus and in inflammation, have been observed, but the roles of the sympathetic innervation in these and other pathologies are not adequately understood. In this article, we first review the principal roles of the sympathetic innervation of the GI tract in controlling motility, fluid exchange and gut blood flow in healthy individuals. We then discuss the evidence that there are important interactions of sympathetic transmitters with the gut immune system and enteric glia, and evidence that inflammation has substantial effects on sympathetic neurons. These reciprocal interactions contribute to pathological changes in ways that are not yet clarified. Finally, we focus on inflammation, diabetes and postoperative ileus as conditions in which there is sympathetic involvement in compromised gut function.
Collapse
Affiliation(s)
- A E Lomax
- Gastrointestinal Diseases Research Unit, Department of Physiology, Queen's University, Kingston, ON, Canada.
| | | | | |
Collapse
|
48
|
Zhang J, Halm ST, Halm DR. Adrenergic activation of electrogenic K+ secretion in guinea pig distal colonic epithelium: involvement of beta1- and beta2-adrenergic receptors. Am J Physiol Gastrointest Liver Physiol 2009; 297:G269-77. [PMID: 19460844 PMCID: PMC2724081 DOI: 10.1152/ajpgi.00076.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Adrenergic stimulation of electrogenic K+ secretion in isolated mucosa from guinea pig distal colon required activation of two beta-adrenergic receptor subtypes (beta-AdrR). Addition of epinephrine (epi) or norepinephrine (norepi) to the bathing solution of mucosae in Ussing chambers increased short-circuit current (Isc) and transepithelial conductance (Gt), consistent with this cation secretion. A beta-adrenergic classification was supported by propranolol antagonism of this secretory response and the lack of effect by the alpha-AdrR antagonists BE2254 (alpha1-AdrR) and yohimbine (alpha2-AdrR). Subtype-selective antagonists CGP20712A (beta1-AdrR), ICI-118551 (beta2-AdrR), and SR59320A (beta3-AdrR) were relatively ineffective at inhibiting the epi-stimulated Isc response. In combination, CGP20712A and ICI-118551 inhibited the response, which supported a synergistic action by beta1-AdrR and beta2-AdrR. Expression of mRNA for both beta1-AdrR and beta2-AdrR was indicated by RT-PCR of RNA from colonic epithelial cells. Protein expression was indicated by immunoblot showing bands at molecular weights consistent with monomers and oligomers. Immunoreactivity (ir) for beta1-AdrR and beta2-AdrR was prominent in basolateral membranes of columnar epithelial cells in the crypts of Lieberkühn as well as intercrypt surface epithelium. Cells in the pericryptal sheath also had beta1-AdrR(ir) but did not have discernable beta2-AdrR(ir). The adrenergic sensitivity of K+ secretion measured by Isc and Gt was relatively low as indicated by EC(50)s of 41 +/- 7 nM for epi and 50 +/- 14 nM for norepi. Adrenergic activation of electrogenic K+ secretion required the involvement of both beta1-AdrR and beta2-AdrR, occurring with an agonist sensitivity reduced compared with reported values for either receptor subtype.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Susan T. Halm
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Dan R. Halm
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| |
Collapse
|
49
|
Gomes P, Chevalier J, Boesmans W, Roosen L, van den Abbeel V, Neunlist M, Tack J, Vanden Berghe P. ATP-dependent paracrine communication between enteric neurons and glia in a primary cell culture derived from embryonic mice. Neurogastroenterol Motil 2009; 21:870-e62. [PMID: 19368656 DOI: 10.1111/j.1365-2982.2009.01302.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The importance of dynamic interactions between glia and neurons is increasingly recognized, both in the central and enteric nervous system. However, apart from their protective role, little is known about enteric neuro-glia interaction. The aim was to investigate neuro-glia intercellular communication in a mouse culture model using optical techniques. Complete embryonic (E13) guts were enzymatically dissociated, seeded on coverslips and studied with immunohistochemistry and Ca(2+)-imaging. Putative progenitor-like cells (expressing both PGP9.5 and S-100) differentiated over approximately 5 days into glia or neurons expressing typical cell-specific markers. The glia-neuron ratio could be manipulated by specific supplements (N2, G5). Neurons and glia were functionally identified both by their Ca(2+)-response to either depolarization (high K(+)) or lysophosphatidic acid and by the expression of typical markers. Neurons responded to ACh, DMPP, 5-HT, ATP and electrical stimulation, while glia responded to ATP and ADPbetas. Inhibition of glial responses by MRS2179 suggests involvement of P2Y1 receptors. Neuronal stimulation also caused delayed glial responses, which were reduced by suramin and by exogenous apyrases that catalyse nucleotide breakdown. Conversely, glial responses were enhanced by ARL-67156, an ecto-ATPase inhibitor. In this mouse enteric co-culture, functional glia and neurons can be easily monitored using optical techniques. Glial cells can be activated directly by ATP or ADPbetas. Activation of neuronal cells (DMPP, K(+)) causes secondary responses in glial cells, which can be modulated by tuning ATP and ADP breakdown. This strongly supports the involvement of paracrine purinergic communication between enteric neurons and glia.
Collapse
Affiliation(s)
- P Gomes
- Center for Gastroenterological Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Gulbransen BD, Sharkey KA. Purinergic neuron-to-glia signaling in the enteric nervous system. Gastroenterology 2009; 136:1349-58. [PMID: 19250649 DOI: 10.1053/j.gastro.2008.12.058] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 12/03/2008] [Accepted: 12/29/2008] [Indexed: 01/28/2023]
Abstract
BACKGROUND & AIMS Enteric glia are intimately associated with neurons in the enteric nervous system (ENS) and display morphologic and molecular similarities to central nervous system (CNS) astrocytes. Enteric glia express neurotransmitter receptors, suggesting that, like astrocytes, they are active participants in neuronal communication. In the ENS, the purine adenosine triphosphate (ATP) is co-released with the neurotransmitters noradrenaline and acetylcholine. Enteric glia express purinergic receptors and respond to ATP in vitro, suggesting that enteric glia participate in functional gastrointestinal responses to nerve signaling. We investigated whether enteric glia are activated by ATP released from enteric neurons. METHODS Synaptic activity was elicited in enteric neurons by electrically stimulating interganglionic connectives in the myenteric plexus of the guinea pig colon. Activity in enteric glial cells was detected by imaging intracellular calcium in situ. RESULTS Neuronal stimulation elicited increases in intracellular calcium in enteric glial cells that were blocked by tetrodotoxin, the nonselective purinergic receptor antagonist pyridoxal phosphate-6-azo(benzene-2,4-disulfonic acid) tetrasodium salt hydrate (PPADS), and the phospholipase C inhibitor U73122. Furthermore, enteric glia responded robustly to exogenously applied ATP in situ, and the ATP response was blocked by PPADS and U73122. Data from pharmacologic profiling and immunohistochemical analyses support the hypothesis that P2Y4 is the major functional receptor underlying the ATP response in enteric glia. CONCLUSIONS Our results provide direct evidence for functional purinergic neuron-glia communication in the enteric nervous system, raising the possibility that ATP released with neurotransmitters during enteric synaptic transmission functions to signal to enteric glia.
Collapse
Affiliation(s)
- Brian D Gulbransen
- Hotchkiss Brain Institute, Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta, Canada.
| | | |
Collapse
|