Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 744
Filter
1.
Am J Physiol Regul Integr Comp Physiol ; 327(2): R173-R187, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38860288

ABSTRACT

Vagal afferents to the gastrointestinal tract are crucial for the regulation of food intake, signaling negative feedback that contributes to satiation and positive feedback that produces appetition and reward. Vagal afferents to the small intestinal mucosa contribute to this regulation by sensing luminal stimuli and reporting this information to the brain. These afferents respond to mechanical, chemical, thermal, pH, and osmolar stimuli, as well as to bacterial products and immunogens. Surprisingly, little is known about how these stimuli are transduced by vagal mucosal afferents or how their transduction is organized among these afferents' terminals. Furthermore, the effects of stimulus concentration ranges or physiological stimuli on vagal activity have not been examined for some of these stimuli. Also, detection of luminal stimuli has rarely been examined in rodents, which are most frequently used for studying small intestinal innervation. Here we review what is known about stimulus detection by vagal mucosal afferents and illustrate the complexity of this detection using nutrients as an exemplar. The accepted model proposes that nutrients bind to taste receptors on enteroendocrine cells (EECs), which excite them, causing the release of hormones that stimulate vagal mucosal afferents. However, evidence reviewed here suggests that although this model accounts for many aspects of vagal signaling about nutrients, it cannot account for all aspects. A major goal of this review is therefore to evaluate what is known about nutrient absorption and detection and, based on this evaluation, identify candidate mucosal cells and structures that could cooperate with EECs and vagal mucosal afferents in stimulus detection.


Subject(s)
Intestinal Mucosa , Intestine, Small , Vagus Nerve , Animals , Vagus Nerve/physiology , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Humans , Intestine, Small/innervation , Intestine, Small/metabolism , Afferent Pathways/physiology , Taste/physiology , Neurons, Afferent/physiology
2.
Cell Stem Cell ; 30(9): 1166-1178.e8, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37597516

ABSTRACT

The intestinal epithelium has high intrinsic turnover rate, and the precise renewal of the epithelium is dependent on the microenvironment. The intestine is innervated by a dense network of peripheral nerves that controls various aspects of intestinal physiology. However, the role of neurons in regulating epithelial cell regeneration remains largely unknown. Here, we investigated the effects of gut-innervating adrenergic nerves on epithelial cell repair following irradiation (IR)-induced injury. We observed that adrenergic nerve density in the small intestine increased post IR, while chemical adrenergic denervation impaired epithelial regeneration. Single-cell RNA sequencing experiments revealed a decrease in IL-22 signaling post IR in denervated animals. Combining pharmacologic and genetic tools, we demonstrate that ß-adrenergic receptor signaling drives IL-22 production from type 3 innate lymphoid cells (ILC3s) post IR, which in turn promotes epithelial regeneration. These results define an adrenergic-ILC3 axis important for intestinal regeneration.


Subject(s)
Adrenergic Neurons , Immunity, Innate , Intestinal Mucosa , Lymphocytes , Regeneration , Animals , Signal Transduction , Adrenergic Neurons/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Intestinal Mucosa/physiology , Mice , Interleukin-22
3.
Front Immunol ; 12: 718220, 2021.
Article in English | MEDLINE | ID: mdl-34621267

ABSTRACT

A large and expending body of evidence indicates that the gut-brain axis likely plays a crucial role in neurological diseases, including multiple sclerosis (MS). As a whole, the gut-brain axis can be considered as a bi-directional multi-crosstalk pathway that governs the interaction between the gut microbiota and the organism. Perturbation in the commensal microbial population, referred to as dysbiosis, is frequently associated with an increased intestinal permeability, or "leaky gut", which allows the entrance of exogeneous molecules, in particular bacterial products and metabolites, that can disrupt tissue homeostasis and induce inflammation, promoting both local and systemic immune responses. An altered gut microbiota could therefore have significant repercussions not only on immune responses in the gut but also in distal effector immune sites such as the CNS. Indeed, the dysregulation of this bi-directional communication as a consequence of dysbiosis has been implicated as playing a possible role in the pathogenesis of neurological diseases. In multiple sclerosis (MS), the gut-brain axis is increasingly being considered as playing a crucial role in its pathogenesis, with a major focus on specific gut microbiota alterations associated with the disease. In both MS and its purported murine model, experimental autoimmune encephalomyelitis (EAE), gastrointestinal symptoms and/or an altered gut microbiota have been reported together with increased intestinal permeability. In both EAE and MS, specific components of the microbiota have been shown to modulate both effector and regulatory T-cell responses and therefore disease progression, and EAE experiments with germ-free and specific pathogen-free mice transferred with microbiota associated or not with disease have clearly demonstrated the possible role of the microbiota in disease pathogenesis and/or progression. Here, we review the evidence that can point to two possible consequences of the gut-brain axis dysfunction in MS and EAE: 1. A pro-inflammatory intestinal environment and "leaky" gut induced by dysbiosis could lead to an altered communication with the CNS through the cholinergic afferent fibers, thereby contributing to CNS inflammation and disease pathogenesis; and 2. Neuroinflammation affecting efferent cholinergic transmission could result in intestinal inflammation as disease progresses.


Subject(s)
Brain-Gut Axis , Brain/metabolism , Disease Susceptibility , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Animals , Biomarkers , Brain/immunology , Brain/pathology , Brain-Gut Axis/immunology , Cell Communication , Combined Modality Therapy/methods , Disease Management , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gastrointestinal Microbiome/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy
4.
Toxicol Appl Pharmacol ; 432: 115737, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34662668

ABSTRACT

Cadmium (Cd) is one of the most harmful environmental pollutants and has been found to have adverse effects on the gut. However, the toxic effects and potential mechanism of Cd on intestinal epithelial cells (IECs) are poorly understood. This study evaluated the effects of Cd exposure (0, 0.25, 0.5, 1, 2, and 4 µM) on IEC-6 cells in terms of cell viability and apoptosis, as well as apoptosis-associated gene expression. The results indicated that low doses (0.25- 1 µM) of Cd exhibited hormetic effects, while high doses of Cd (2 and 4 µM) reduced cell viability. The apoptotic effect increased in a dose-dependent pattern. Moreover, the mRNA levels of the Bcl-2, Bax and Caspase 3 genes were altered, which was in agreement with their protein expression. Based on sequencing analysis, the expression pattern of the microRNAs (miRNAs) changed significantly in the 2 µM Cd-treated group. QRT-PCR verified that 7 miRNAs, including miR-124-3p and miR-370-3p, were all upregulated with dose-effect relationship. Besides, transfection of miR-124-3p and miR-370-3p mimics /inhibitor and Bcl-2 siRNA into IEC-6 cells verified that these two miRNAs could regulate Cd-induced apoptosis by targeting Bcl-2. Finally, the direct targeting relationship between miR-370-3p and Bcl-2 gene was confirmed by luciferase reporter assay. Overall, the results demonstrated that Cd exposure could induce apoptosis in IEC-6 cells. The potential mechanism may be interference with the regulation of Bcl-2 gene expression by miR-370-3p and miR-124-3p.


Subject(s)
Apoptosis/drug effects , Cadmium Compounds/toxicity , Intestinal Mucosa/drug effects , MicroRNAs/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cell Line , Down-Regulation , Intestinal Mucosa/innervation , Intestinal Mucosa/pathology , MicroRNAs/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Signal Transduction
5.
Cells ; 10(9)2021 08 30.
Article in English | MEDLINE | ID: mdl-34571902

ABSTRACT

Inflammatory bowel diseases (IBD) are characterized by chronic dysregulation of immune homeostasis, epithelial demise, immune cell activation, and microbial translocation. Each of these processes leads to proinflammatory changes via the release of cytokines, damage-associated molecular patterns (DAMPs), and pathogen-associated molecular patterns (PAMPs), respectively. The impact of these noxious agents on the survival and function of the enteric nervous system (ENS) is poorly understood. Here, we show that in contrast to an expected decrease, experimental as well as clinical colitis causes an increase in the transcript levels of enteric neuronal and glial genes. Immunostaining revealed an elevated neuronal innervation of the inflamed regions of the gut mucosa. The increase was seen in models with overt damage to epithelial cells and models of T cell-induced colitis. Transcriptomic data from treatment naïve pediatric IBD patients also confirmed the increase in the neuroglial genes and were replicated on an independent adult IBD dataset. This induction in the neuroglial genes was transient as levels returned to normal upon the induction of remission in both mouse models as well as colitis patients. Our data highlight the dynamic and robust nature of the enteric nervous system in colitis and open novel questions on its regulation.


Subject(s)
Colitis/pathology , Enteric Nervous System/pathology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/innervation , Neurons/pathology , Transcriptome , Animals , Colitis/etiology , Colitis/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Mice , Neurons/immunology , Neurons/metabolism
6.
Eur J Pharmacol ; 906: 174220, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34081905

ABSTRACT

Schisandrin C (Sch C) is one of the main components of Schisandra chinensis (Schisandra). Since the olden times, Schisandra has been used as a traditional herbal medicine in Asia. Recent studies have shown that Schisandra is effective against irritable bowel syndrome (IBS) in an animal model and affects IBS through the 5-HT3A pathway in the IBS rat model. However, there lacks fundamental research on the interaction of specific components of Schisandra with the 5-HT3A receptor for the treatment of IBS. We hypothesized that a component of Schisandra binds to the 5-HT3A receptor and identified Sch C via a screening work using two electrode-voltage clamps (TEVC). Thus, we aimed to elucidate the neuropharmacological actions between Sch C and the 5-HT3A receptor at molecular and cellular levels. Co-treatment of Sch C with 5-HT inhibited I5-HT in a reversible, concentrate-dependent, like-competition, and voltage-independent manner, and IC50 values of Sch C. Besides, the main binding positions of Sch C were identified through 3D modeling and point mutation were V225A and V288Y on 5-HT3A receptor. Thus, we suggest the potential of Sch C in treating IBS in a manner that suppresses excessive neuronal serotonin signaling in the synapse of sensory neurons and enterochromaffin (EC) cells. In conclusion, the results demonstrate the mechanism of interaction between Sch C and 5-HT3A receptor and reveal Sch C as a novel antagonist.


Subject(s)
Lignans/pharmacology , Polycyclic Compounds/pharmacology , Receptors, Serotonin, 5-HT3/metabolism , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Animals , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Enterochromaffin Cells/drug effects , Enterochromaffin Cells/metabolism , Humans , Inhibitory Concentration 50 , Intestinal Mucosa/drug effects , Intestinal Mucosa/innervation , Intestinal Mucosa/pathology , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/pathology , Lignans/therapeutic use , Molecular Docking Simulation , Oocytes , Patch-Clamp Techniques , Polycyclic Compounds/therapeutic use , Receptors, Serotonin, 5-HT3/genetics , Receptors, Serotonin, 5-HT3/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Serotonin 5-HT3 Receptor Antagonists/therapeutic use , Xenopus laevis
7.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946994

ABSTRACT

The gastrointestinal lumen is a rich source of eukaryotic and prokaryotic viruses which, together with bacteria, fungi and other microorganisms comprise the gut microbiota. Pathogenic viruses inhabiting this niche have the potential to induce local as well as systemic complications; among them, the viral ability to disrupt the mucosal barrier is one mechanism associated with the promotion of diarrhea and tissue invasion. This review gathers recent evidence showing the contributing effects of diet, gut microbiota and the enteric nervous system to either support or impair the mucosal barrier in the context of viral attack.


Subject(s)
Bacteriophages/physiology , Diet , Enteric Nervous System/physiology , Gastric Mucosa/virology , Gastrointestinal Microbiome , Host Microbial Interactions/physiology , Intestinal Mucosa/virology , Viruses , Defensins/physiology , Digestion , Disease Susceptibility , Enteric Nervous System/virology , Food/virology , Gastric Mucosa/immunology , Gastric Mucosa/innervation , Gastric Mucosa/metabolism , Gastroenteritis/virology , Host Microbial Interactions/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Malnutrition/virology , Mucus/metabolism , Mucus/virology , Neurons/virology , Opportunistic Infections/virology , Plant Viruses , Virus Diseases/microbiology , Virus Diseases/physiopathology
8.
Neurogastroenterol Motil ; 33(9): e14140, 2021 09.
Article in English | MEDLINE | ID: mdl-33939232

ABSTRACT

BACKGROUND: The type 3 adenylyl cyclase (AC3) enzyme is involved in the synthesis of cyclic adenosine monophosphate (cAMP). It is primarily expressed in the central nervous system (CNS) and plays a crucial role in neurogenesis and neural dendritic arborization. However, the AC3's functional role in the gastrointestinal tract remains ambiguous. METHODS: AC3 expression in enteric tissue of AC3+/+ mice was investigated using immunohistochemistry and RT-PCR. AC3 knock-out mice (AC3-/- ) were used to examine the effect of AC3 on the enteric nervous system (ENS) function and the number of cilia and apoptotic cells. Additionally, total gastrointestinal transit time and colonic motility were compared between the AC3-/- and AC3+/+ groups of mice. KEY RESULTS: AC3 was predominately expressed in the myenteric plexus of the large intestine. Colonic-bead expulsion analysis showed accelerated propulsion in the large intestine of the AC3-/- mice. The AC3-/- mice demonstrated reduced nerve fibers and enteric glial cells count in colonic mucosa compared to the AC3+/+ mice. Furthermore, AC3-/- mice exhibited increased cellular apoptosis and reduced ARL13B+ cilium cells in the colonic lamina propria compared to the AC3+/+ mice. CONCLUSIONS: In AC3-/- mice, innervation of the lamina propria in the colonic mucosa was reduced and colonic propulsion was accelerated. AC3 is crucial for the development and function of the adult neural network of ENS. AC3 deficiency caused atrophy in the colonic mucosal neural network of mice.


Subject(s)
Adenylyl Cyclases/metabolism , Enteric Nervous System/enzymology , Intestinal Mucosa/innervation , Animals , Gastrointestinal Motility/physiology , Mice , Mice, Knockout
9.
Cell Mol Gastroenterol Hepatol ; 12(2): 507-545, 2021.
Article in English | MEDLINE | ID: mdl-33741501

ABSTRACT

BACKGROUND & AIMS: Hirschsprung's disease (HSCR) is a congenital intestinal motility disorder defined by the absence of enteric neuronal cells (ganglia) in the distal gut. The development of HSCR-associated enterocolitis remains a life-threatening complication. Absence of enteric ganglia implicates innervation of acetylcholine-secreting (cholinergic) nerve fibers. Cholinergic signals have been reported to control excessive inflammation, but the impact on HSCR-associated enterocolitis is unknown. METHODS: We enrolled 44 HSCR patients in a prospective multicenter study and grouped them according to their degree of colonic mucosal acetylcholinesterase-positive innervation into low-fiber and high-fiber patient groups. The fiber phenotype was correlated with the tissue cytokine profile as well as immune cell frequencies using Luminex analysis and fluorescence-activated cell sorting analysis of colonic tissue and immune cells. Using confocal immunofluorescence microscopy, macrophages were identified in close proximity to nerve fibers and characterized by RNA-seq analysis. Microbial dysbiosis was analyzed in colonic tissue using 16S-rDNA gene sequencing. Finally, the fiber phenotype was correlated with postoperative enterocolitis manifestation. RESULTS: The presence of mucosal nerve fiber innervation correlated with reduced T-helper 17 cytokines and cell frequencies. In high-fiber tissue, macrophages co-localized with nerve fibers and expressed significantly less interleukin 23 than macrophages from low-fiber tissue. HSCR patients lacking mucosal nerve fibers showed microbial dysbiosis and had a higher incidence of postoperative enterocolitis. CONCLUSIONS: The mucosal fiber phenotype might serve as a prognostic marker for enterocolitis development in HSCR patients and may offer an approach to personalized patient care and new therapeutic options.


Subject(s)
Cholinergic Neurons/pathology , Enterocolitis/etiology , Hirschsprung Disease/complications , Intestinal Mucosa/innervation , Intestinal Mucosa/pathology , Acetylcholinesterase/metabolism , Child , Child, Preschool , Cohort Studies , Cytokines/metabolism , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Hirschsprung Disease/pathology , Hirschsprung Disease/surgery , Humans , Infant , Infant, Newborn , Inflammation/immunology , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Male , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors
10.
Clin Transl Gastroenterol ; 12(2): e00313, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33617189

ABSTRACT

INTRODUCTION: Despite heterogeneity, an increased prevalence of psychological comorbidity and an altered pronociceptive gut microenvironment have repeatedly emerged as causative pathophysiology in patients with irritable bowel syndrome (IBS). Our aim was to study these phenomena by comparing gut-related symptoms, psychological scores, and biopsy samples generated from a detailed diarrhea-predominant IBS patient (IBS-D) cohort before their entry into a previously reported clinical trial. METHODS: Data were generated from 42 patients with IBS-D who completed a daily 2-week bowel symptom diary, the Hospital Anxiety and Depression score, and the Patient Health Questionnaire-12 Somatic Symptom score and underwent unprepared flexible sigmoidoscopy. Sigmoid mucosal biopsies were separately evaluated using immunohistochemistry and culture supernatants to determine cellularity, mediator levels, and ability to stimulate colonic afferent activity. RESULTS: Pain severity scores significantly correlated with the daily duration of pain (r = 0.67, P < 0.00001), urgency (r = 0.57, P < 0.0005), and bloating (r = 0.39, P < 0.05), but not with psychological symptom scores for anxiety, depression, or somatization. Furthermore, pain severity scores from individual patients with IBS-D were significantly correlated (r = 0.40, P < 0.008) with stimulation of colonic afferent activation mediated by their biopsy supernatant, but not with biopsy cell counts nor measured mediator levels. DISCUSSION: Peripheral pronociceptive changes in the bowel seem more important than psychological factors in determining pain severity within a tightly phenotyped cohort of patients with IBS-D. No individual mediator was identified as the cause of this pronociceptive change, suggesting that nerve targeting therapeutic approaches may be more successful than mediator-driven approaches for the treatment of pain in IBS-D.


Subject(s)
Abdominal Pain/etiology , Afferent Pathways/physiopathology , Colon, Sigmoid/innervation , Irritable Bowel Syndrome/physiopathology , Adult , Animals , Anxiety , Biopsy , Depression , Diarrhea/etiology , Female , Gain of Function Mutation , Humans , Immunohistochemistry , Intestinal Mucosa/innervation , Irritable Bowel Syndrome/genetics , Irritable Bowel Syndrome/psychology , Male , Mice , NAV1.9 Voltage-Gated Sodium Channel/genetics , Severity of Illness Index , Sigmoidoscopy
11.
Nature ; 592(7852): 99-104, 2021 04.
Article in English | MEDLINE | ID: mdl-33627870

ABSTRACT

The small intestine is the main organ for nutrient absorption, and its extensive resection leads to malabsorption and wasting conditions referred to as short bowel syndrome (SBS). Organoid technology enables an efficient expansion of intestinal epithelium tissue in vitro1, but reconstruction of the whole small intestine, including the complex lymphovascular system, has remained challenging2. Here we generate a functional small intestinalized colon (SIC) by replacing the native colonic epithelium with ileum-derived organoids. We first find that xenotransplanted human ileum organoids maintain their regional identity and form nascent villus structures in the mouse colon. In vitro culture of an organoid monolayer further reveals an essential role for luminal mechanistic flow in the formation of villi. We then develop a rat SIC model by repositioning the SIC at the ileocaecal junction, where the epithelium is exposed to a constant luminal stream of intestinal juice. This anatomical relocation provides the SIC with organ structures of the small intestine, including intact vasculature and innervation, villous structures, and the lacteal (a fat-absorbing lymphatic structure specific to the small intestine). The SIC has absorptive functions and markedly ameliorates intestinal failure in a rat model of SBS, whereas transplantation of colon organoids instead of ileum organoids invariably leads to mortality. These data provide a proof of principle for the use of intestinal organoids for regenerative purposes, and offer a feasible strategy for SBS treatment.


Subject(s)
Colon/cytology , Ileum/transplantation , Intestinal Mucosa/cytology , Organoids/transplantation , Regeneration , Regenerative Medicine/methods , Short Bowel Syndrome/therapy , Animals , Colon/blood supply , Colon/innervation , Colon/surgery , Disease Models, Animal , Heterografts , Humans , Ileum/cytology , Intestinal Mucosa/blood supply , Intestinal Mucosa/innervation , Intestinal Mucosa/surgery , Male , Organ Culture Techniques , Organoids/cytology , Rats , Rats, Inbred Lew , Short Bowel Syndrome/pathology , Short Bowel Syndrome/surgery
12.
Gastroenterology ; 160(4): 1208-1223.e4, 2021 03.
Article in English | MEDLINE | ID: mdl-32980343

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.


Subject(s)
Colon/innervation , Ganglia, Sympathetic/physiology , Gastrointestinal Motility/physiology , Myenteric Plexus/physiology , Animals , Colon/cytology , Colon/drug effects , Colon/physiology , Female , Ganglia, Sympathetic/drug effects , Gastrointestinal Motility/drug effects , Guanethidine/pharmacology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/innervation , Intestinal Mucosa/physiology , Male , Mice , Models, Animal , Myenteric Plexus/cytology , Myenteric Plexus/drug effects , Neurons/drug effects , Neurons/physiology , Optogenetics , Prazosin/pharmacology , RNA-Seq , Single-Cell Analysis , Yohimbine/pharmacology
13.
Am J Physiol Gastrointest Liver Physiol ; 320(2): G193-G205, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33296267

ABSTRACT

Mucosal damage is a key feature of inflammatory bowel diseases (IBD) and healing of the mucosa is an endpoint of IBD treatment that is often difficult to achieve. Autonomic neurons of the parasympathetic and sympathetic nervous system may influence intestinal epithelial cell growth and modulating epithelial innervation could for that reason serve as an interesting therapeutic option to improve mucosal healing. Understanding of the biological processes triggered by nonspecific and specific epithelial adrenergic and cholinergic receptor activation is of key importance. At present, with rising technological advances, bioelectronic neuromodulation as treatment modality has gained momentum. We discuss the current view on state-of-the-art innervation of the intestinal crypt and its impact on epithelial cell growth and differentiation. Furthermore, we outline bioelectronic technology and review its relevance to wound healing processes.


Subject(s)
Electric Stimulation Therapy , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/injuries , Intestinal Mucosa/innervation , Neurons/physiology , Animals , Humans , Wound Healing
14.
Cell Host Microbe ; 29(2): 179-196.e9, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33352109

ABSTRACT

The intestinal epithelium senses nutritional and microbial stimuli using epithelial sensory enteroendocrine cells (EEC). EECs communicate nutritional information to the nervous system, but whether they also relay signals from intestinal microbes remains unknown. Using in vivo real-time measurements of EEC and nervous system activity in zebrafish, we discovered that the bacteria Edwardsiella tarda activate EECs through the receptor transient receptor potential ankyrin A1 (Trpa1) and increase intestinal motility. Microbial, pharmacological, or optogenetic activation of Trpa1+EECs directly stimulates vagal sensory ganglia and activates cholinergic enteric neurons by secreting the neurotransmitter 5-hydroxytryptamine (5-HT). A subset of indole derivatives of tryptophan catabolism produced by E. tarda and other gut microbes activates zebrafish EEC Trpa1 signaling. These catabolites also directly stimulate human and mouse Trpa1 and intestinal 5-HT secretion. These results establish a molecular pathway by which EECs regulate enteric and vagal neuronal pathways in response to microbial signals.


Subject(s)
Edwardsiella tarda/metabolism , Enteric Nervous System/metabolism , Enteroendocrine Cells/physiology , Intestinal Mucosa/metabolism , TRPA1 Cation Channel/metabolism , Animals , Animals, Genetically Modified , Cholinergic Neurons/metabolism , Enteric Nervous System/cytology , Gastrointestinal Motility/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/innervation , Proto-Oncogene Proteins c-ret/genetics , Serotonin/metabolism , Signal Transduction , Tryptophan/metabolism , Zebrafish , Zebrafish Proteins/genetics
15.
Sci Rep ; 10(1): 21725, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33303794

ABSTRACT

In Hirschsprung's disease (HSCR), postoperative course remains unpredictable. Our aim was to define predictive factors of the main postoperative complications: obstructive symptoms (OS) and Hirschsprung-associated enterocolitis (HAEC). In this prospective multicentre cohort study, samples of resected bowel were collected at time of surgery in 18 neonates with short-segment HSCR in tertiary care hospitals. OS and HAEC were noted during postoperative follow-up. We assessed the enteric nervous system and the intestinal epithelial barrier (IEB) in ganglionic segments by combining immunohistochemical, proteomic and transcriptomic approaches, with functional ex vivo analysis of motility and para/transcellular permeability. Ten HSCR patients presented postoperative complications (median follow-up 23.5 months): 6 OS, 4 HAEC (2 with OS), 2 diarrhoea (without OS/HAEC). Immunohistochemical analysis showed a significant 41% and 60% decrease in median number of nNOS-IR myenteric neurons per ganglion in HSCR with OS as compared to HSCR with HAEC/diarrhoea (without OS) and HSCR without complications (p = 0.0095; p = 0.002, respectively). Paracellular and transcellular permeability was significantly increased in HSCR with HAEC as compared to HSCR with OS/diarrhoea without HAEC (p = 0.016; p = 0.009) and HSCR without complications (p = 0.029; p = 0.017). This pilot study supports the hypothesis that modulating neuronal phenotype and enhancing IEB permeability may treat or prevent postoperative complications in HSCR.


Subject(s)
Enteric Nervous System/physiopathology , Enterocolitis/epidemiology , Hirschsprung Disease/surgery , Intestinal Mucosa/physiopathology , Postoperative Complications/epidemiology , Child, Preschool , Diarrhea/epidemiology , Diarrhea/etiology , Diarrhea/prevention & control , Enterocolitis/etiology , Enterocolitis/prevention & control , Follow-Up Studies , Ganglia/physiopathology , Humans , Infant , Infant, Newborn , Intestinal Mucosa/innervation , Pilot Projects , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Prospective Studies , Time Factors
16.
Int J Med Sci ; 17(14): 2147-2154, 2020.
Article in English | MEDLINE | ID: mdl-32922175

ABSTRACT

Background: Hemorrhagic shock-induced ischemia and hypoxia elicit endoplasmic reticulum stress (ERS) that leads to cell apoptosis, tissue structural damage and organ dysfunction and failure. Stellate ganglion blockade (SGB) has been demonstrated to improve intestinal barrier dysfunction induced by hemorrhagic shock. The present study sought to investigate whether the beneficial effect of SGB on the intestinal mucosal barrier function is via suppression of ERS. Materials and methods: A conscious rat model of hemorrhagic shock (40 ±2 mmHg for 1 hour, followed by resuscitation) was established. The parameters reflecting intestinal morphology and intestinal mucosal barrier function including wet-dry ratio (W/D), intestinal permeability, D-lactic acid (D-LA) and intestinal fatty acid binding protein (I-FABP) in plasma, and expressions of ATF6α, PERK, and IRE1α in intestinal tissues were then observed. Furthermore, the effects of either SGB or ERS inhibitor, 4-phenylbutyric acid (4-PBA), on these parameters in rats with hemorrhagic shock were assessed. The effect of ERS agonist tunicamycin (TM) on the rats subjected with both SGB and hemorrhagic shock was also determined. Results: Either SGB or administration of ERS inhibitor, 4-PBA, alleviated hemorrhagic shock-induced adverse effects such as intestinal mucosal barrier dysfunction and excessive autophagy, which were characterized by damaged intestinal tissue, enhanced intestinal permeability and D-LA and I-FABP levels in plasma, and increased expressions of ATF6α, PERK, IRE1α in intestinal tissue. In contrast, administration of ERS agonist, TM, suppressed the beneficial effects of SGB on intestinal tissue and function during hemorrhagic shock. Conclusion: The SGB repairs intestinal mucosal barrier through suppression of ERS following hemorrhagic shock.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Intestinal Mucosa/pathology , Nerve Block/methods , Shock, Hemorrhagic/therapy , Stellate Ganglion/drug effects , Animals , Apoptosis , Butylamines/administration & dosage , Disease Models, Animal , Humans , Intestinal Mucosa/innervation , Male , Permeability , Rats , Ropivacaine , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/pathology , Tunicamycin/administration & dosage
17.
Front Immunol ; 11: 1268, 2020.
Article in English | MEDLINE | ID: mdl-32676079

ABSTRACT

Neuronal regulation of diverse physiological functions requires complex molecular interactions in innervated tissues to maintain proper organ function. Here we show that loss of the neuronal cell surface adhesion/recognition molecule Contactin-1 (Cntn1) directly impairs intestinal function causing wasting that subsequently results in global immune defects. Loss of Cntn1 results in hematologic alterations and changes in blood metabolites associated with malnourishment. We found thymus and spleen of Cntn1-deficient animals atrophied with severe reductions in lymphocyte populations. Elevated thymic Gilz expression indicated ongoing glucocorticoid signaling in Cntn1-deficient animals, consistent with the malnourishment phenotype. Intestinal Contactin-1 was localized to neurons in the villi and the submucosal/myenteric plexus that innervates smooth muscle. Loss of Cntn1 was associated with reduced intestinal Bdnf and Adrb2, indicating reduced neuromuscular crosstalk. Additionally, loss of Cntn1 resulted in reduced recruitment of CD3+ T cells to villi within the small intestine. Together, these data illustrate the critical role of Contactin-1 function within the gut, and how this is required for normal systemic immune functions.


Subject(s)
Contactin 1/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Animals , Biomarkers , Blood Cell Count , Blood Chemical Analysis , Flow Cytometry , Gene Expression Profiling , Glucocorticoids/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Phenotype , Signal Transduction , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology
18.
Trends Parasitol ; 36(9): 761-772, 2020 09.
Article in English | MEDLINE | ID: mdl-32713764

ABSTRACT

Gastrointestinal helminth infection still constitutes a major public health issue, particularly in the developing world. As these parasites can undergo a large part of their lifecycle within the intestinal tract the host has developed various structural and cellular specializations at the epithelial barrier to contend with infection. Detailed characterization of these cells will provide important insights about their contributions to the protective responses mediated against helminths. Here, we discuss how key components of the intestinal epithelium may function to limit the initial establishment of helminths, and how these cells are altered during an active response to infection.


Subject(s)
Helminthiasis/immunology , Host-Parasite Interactions/immunology , Intestinal Mucosa/innervation , Intestinal Mucosa/parasitology , Animals , Helminthiasis/parasitology , Helminths/immunology , Humans
19.
Bull Exp Biol Med ; 169(1): 104-109, 2020 May.
Article in English | MEDLINE | ID: mdl-32488782

ABSTRACT

Morphological, morphometric, and ultrastructural analysis of the nerve fibers in the colon mucosa was performed in C57BL/6 mice at various terms of development of acute colitis induced by dextran sodium sulphate. The nerve fibers were labeled with antibodies to pan-neuronal marker ßIII-tubulin. The progression of inflammatory and ulcerative processes in the mucosa on days 3-5 was associated with hyperplasia and hypertrophy of nerve fibers that peaked on day 7 after colitis induction. Ultrastructural analysis at all terms of colitis development showed moderate degeneration of axons. Thus, hypertrophy and hyperplasia of the nervous fibers in colon mucosa in experimental acute colitis correlated with aggravation of the ulcerative process in the mucosa. These changes are determined by alteration of histoarchitectonics and regenerative processes in the mucosa.


Subject(s)
Colitis/pathology , Colon/innervation , Intestinal Mucosa/innervation , Nerve Fibers/pathology , Acute Disease , Animals , Colitis, Ulcerative/pathology , Colon/pathology , Disease Models, Animal , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Nerve Fibers/ultrastructure
20.
Peptides ; 131: 170342, 2020 09.
Article in English | MEDLINE | ID: mdl-32522585

ABSTRACT

The gut-brain hormone glucagon-like peptide-1 (GLP-1) has received immense attention over the last couple of decades for its widespread metabolic effects. Notably, intestinal GLP-1 has been recognized as an endogenous satiation signal. Yet, the underlying mechanisms and the pathophysiological relevance of intestinal GLP-1 in obesity remain unclear. This review first recapitulates early findings indicating that intestinal GLP-1 is an endogenous satiation signal, whose eating effects are primarily mediated by vagal afferents. Second, on the basis of recent findings challenging a paracrine action of intestinal GLP-1, a new model for the mediation of GLP-1 effects on eating by two discrete vagal afferent subsets will be proposed. The central mechanisms processing the vagal anorexigenic signals need however to be further delineated. Finally, the idea that intestinal GLP-1 secretion and/or effects on eating are altered in obesity and play a pathophysiological role in the development of obesity will be discussed. In summary, despite the successful therapeutic use of GLP-1 receptor agonists as anti-obesity drugs, the eating effects of intestinal GLP-1 still remain to be elucidated. Specifically, the findings presented here call for a further evaluation of the vago-central neuronal substrates activated by intestinal GLP-1 and for further investigation of its pathophysiological role in obesity.


Subject(s)
Eating/physiology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Obesity/metabolism , Satiation/physiology , Animals , Appetite/drug effects , Brain/drug effects , Brain/physiology , Disease Models, Animal , Eating/drug effects , Gene Expression Regulation , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/genetics , Humans , Incretins/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Liraglutide/pharmacology , Obesity/drug therapy , Obesity/genetics , Obesity/pathology , Satiation/drug effects , Signal Transduction , Vagus Nerve/drug effects , Vagus Nerve/physiology
SELECTION OF CITATIONS
SEARCH DETAIL