Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
2.
J Neuroinflammation ; 18(1): 261, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34749758

ABSTRACT

BACKGROUND: The aim of the current study was to investigate the effect of macrophage polarization on the expression of oxytocin (OT) and the oxytocin receptor (OTR) in enteric neurons. METHODS: In this study, we used a classic colitis model and D-mannose model to observe the correlation between macrophage polarization and OT signalling system. In order to further demonstrate the effect of macrophages, we examined the expression of OT signalling system after depletion of macrophages. RESULTS: The data showed that, in vitro, following polarization of macrophages to the M1 type by LPS, the macrophage supernatant contained proinflammatory cytokines (IL-1ß, IL-6 and TNF-α) that inhibited the expression of OT and OTR in cultured enteric neurons; following macrophage polarization to the M2 type by IL4, the macrophage supernatant contained anti-inflammatory cytokines (TGF-ß) that promoted the expression of OT and OTR in cultured enteric neurons. Furthermore, M1 macrophages decreased the expression of the OT signalling system mainly through STAT3/NF-κB pathways in cultured enteric neurons; M2 macrophages increased the expression of the OT signalling system mainly through activation of Smad2/3 and inhibition of the expression of Peg3 in cultured enteric neurons. In a colitis model, we demonstrated that macrophages were polarized to the M1 type during the inflammatory phase, with significant decreased in the expression of OT and OTR. When macrophages were polarized to the M2 type during the recovery phase, OT and OTR expression increased significantly. In addition, we found that D-mannose increased the expression of OT and OTR through polarization of macrophages to the M2 type. CONCLUSIONS: This is the first study to demonstrate that macrophage polarization differentially regulates the expression of OT and OTR in enteric neurons.


Subject(s)
Enteric Nervous System/metabolism , Macrophages/immunology , Neurons/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Animals , Cell Differentiation/immunology , Colitis/immunology , Colitis/metabolism , Enteric Nervous System/immunology , Mice , Mice, Inbred C57BL , Neurons/immunology , Oxytocin/immunology , Receptors, Oxytocin/immunology , Signal Transduction/immunology
3.
Immunol Lett ; 240: 77-97, 2021 12.
Article in English | MEDLINE | ID: mdl-34655659

ABSTRACT

Homeostatic regulation of cellular and molecular processes is essential for the efficient physiological functioning of body organs. It requires an intricate balance of several networks throughout the body, most notable being the nervous, immune and metabolic systems. Several studies have reported the interactions between neuro-immune, immune-metabolic and neuro-metabolic pathways. Current review aims to integrate the information and show that neuro, immune and metabolic systems form the triumvirate of homeostasis. It focuses on the cellular and molecular interactions occurring in the extremities and intestine, which are innervated by the peripheral nervous system and for the intestine in particular the enteric nervous system. While the interdependence of neuro-immune-metabolic pathways provides a fallback mechanism in case of disruption of homeostasis, in chronic pathologies of continued disequilibrium, the collapse of one system spreads to the other interacting networks as well. Current review illustrates this domino-effect using diabetes as the main example. Together, this review attempts to provide a holistic picture of the integrated network of neuro-immune-metabolism and attempts to broaden the outlook when devising a scientific study or a treatment strategy.


Subject(s)
Diabetes Mellitus/immunology , Enteric Nervous System/immunology , Homeostasis/immunology , Neuroimmunomodulation , Signal Transduction/immunology , Animals , Humans
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.
Cells ; 10(7)2021 06 23.
Article in English | MEDLINE | ID: mdl-34201851

ABSTRACT

Recently, the involvement of the nervous system in the pathology of allergic diseases has attracted increasing interest. However, the precise pathophysiological role of enteric neurons in food allergies has not been elucidated. We report the presence of functional high-affinity IgE receptors (FcεRIs) in enteric neurons. FcεRI immunoreactivities were observed in approximately 70% of cholinergic myenteric neurons from choline acetyltransferase-eGFP mice. Furthermore, stimulation by IgE-antigen elevated intracellular Ca2+ concentration in isolated myenteric neurons from normal mice, suggesting that FcεRIs are capable of activating myenteric neurons. Additionally, the morphological investigation revealed that the majority of mucosal mast cells were in close proximity to enteric nerve fibers in the colonic mucosa of food allergy mice. Next, using a newly developed coculture system of isolated myenteric neurons and mucosal-type bone-marrow-derived mast cells (mBMMCs) with a calcium imaging system, we demonstrated that the stimulation of isolated myenteric neurons by veratridine caused the activation of mBMMCs, which was suppressed by the adenosine A3 receptor antagonist MRE 3008F20. Moreover, the expression of the adenosine A3 receptor gene was detected in mBMMCs. Therefore, in conclusion, it is suggested that, through interaction with mucosal mast cells, IgE-antigen-activated myenteric neurons play a pathological role in further exacerbating the pathology of food allergy.


Subject(s)
Cell Communication , Enteric Nervous System/physiopathology , Food Hypersensitivity/immunology , Food Hypersensitivity/physiopathology , Intestinal Mucosa/immunology , Intestinal Mucosa/physiopathology , Mast Cells/immunology , Neurons/pathology , Adenosine/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Antigens/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Communication/drug effects , Cells, Cultured , Enteric Nervous System/drug effects , Enteric Nervous System/immunology , Intestinal Mucosa/drug effects , Intracellular Space/metabolism , Male , Mast Cells/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Myenteric Plexus/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Adenosine A3/genetics , Receptor, Adenosine A3/metabolism , Receptors, IgE/metabolism
7.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34196310

ABSTRACT

The gut-brain axis (GBA) refers to the complex interactions between the gut microbiota and the nervous, immune, and endocrine systems, together linking brain and gut functions. Perturbations of the GBA have been reported in people with multiple sclerosis (pwMS), suggesting a possible role in disease pathogenesis and making it a potential therapeutic target. While research in the area is still in its infancy, a number of studies revealed that pwMS are more likely to exhibit altered microbiota, altered levels of short chain fatty acids and secondary bile products, and increased intestinal permeability. However, specific microbes and metabolites identified across studies and cohorts vary greatly. Small clinical and preclinical trials in pwMS and mouse models, in which microbial composition was manipulated through the use of antibiotics, fecal microbiota transplantation, and probiotic supplements, have provided promising outcomes in preventing CNS inflammation. However, results are not always consistent, and large-scale randomized controlled trials are lacking. Herein, we give an overview of how the GBA could contribute to MS pathogenesis, examine the different approaches tested to modulate the GBA, and discuss how they may impact neuroinflammation and demyelination in the CNS.


Subject(s)
Gastrointestinal Microbiome , Multiple Sclerosis/therapy , Animals , Autoimmunity , Disease Models, Animal , Dysbiosis/immunology , Dysbiosis/physiopathology , Endocrine System/immunology , Endocrine System/physiopathology , Enteric Nervous System/immunology , Enteric Nervous System/microbiology , Enteric Nervous System/physiopathology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Models, Neurological , Multiple Sclerosis/etiology , Multiple Sclerosis/microbiology , Neuroimmunomodulation , Probiotics/therapeutic use
8.
Cell Mol Gastroenterol Hepatol ; 12(5): 1617-1641, 2021.
Article in English | MEDLINE | ID: mdl-34246810

ABSTRACT

BACKGROUND & AIMS: Neuroinflammation in the gut is associated with many gastrointestinal (GI) diseases, including inflammatory bowel disease. In the brain, neuroinflammatory conditions are associated with blood-brain barrier (BBB) disruption and subsequent neuronal injury. We sought to determine whether the enteric nervous system is similarly protected by a physical barrier and whether that barrier is disrupted in colitis. METHODS: Confocal and electron microscopy were used to characterize myenteric plexus structure, and FITC-dextran assays were used to assess for presence of a barrier. Colitis was induced with dextran sulfate sodium, with co-administration of liposome-encapsulated clodronate to deplete macrophages. RESULTS: We identified a blood-myenteric barrier (BMB) consisting of extracellular matrix proteins (agrin and collagen-4) and glial end-feet, reminiscent of the BBB, surrounded by a collagen-rich periganglionic space. The BMB is impermeable to the passive movement of 4 kDa FITC-dextran particles. A population of macrophages is present within enteric ganglia (intraganglionic macrophages [IGMs]) and exhibits a distinct morphology from muscularis macrophages, with extensive cytoplasmic vacuolization and mitochondrial swelling but without signs of apoptosis. IGMs can penetrate the BMB in physiological conditions and establish direct contact with neurons and glia. Dextran sulfate sodium-induced colitis leads to BMB disruption, loss of its barrier integrity, and increased numbers of IGMs in a macrophage-dependent process. CONCLUSIONS: In intestinal inflammation, macrophage-mediated degradation of the BMB disrupts its physiological barrier function, eliminates the separation of the intra- and extra-ganglionic compartments, and allows inflammatory stimuli to access the myenteric plexus. This suggests a potential mechanism for the onset of neuroinflammation in colitis and other GI pathologies with acquired enteric neuronal dysfunction.


Subject(s)
Colitis/etiology , Colitis/metabolism , Macrophages/immunology , Macrophages/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Animals , Biomarkers , Colitis/pathology , Disease Models, Animal , Disease Susceptibility , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Extracellular Matrix , Fluorescent Antibody Technique , Immunohistochemistry , Immunophenotyping , Mice , Myenteric Plexus/ultrastructure , Neuroglia/metabolism , Neuroglia/ultrastructure , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neutrophil Infiltration
9.
J Integr Neurosci ; 20(1): 185-196, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33834706

ABSTRACT

Mast cells are the major effectors in allergic reactions through degranulation and release of inflammatory, vasoactive and nociceptive mediators associated with the pathogenesis of a variety of inflammatory disorders. Mast cells are strategically positioned as gatekeepers at host/environment interfaces, like the skin, airways, gastrointestinal and urogenital tracts, and their presence also in the brain allows them to act not only as sentinels of invading microorganisms but also as targets to respond to different allergens, pathogens and other dangerous agents that can be ingested, inhaled or encountered after the breakdown of the epithelial barrier. Mast cells can respond to any change in the environment by communicating with the different cells involved in the immune response and giving rise to an amplification signal network through feedback loops. They secrete both preformed mediators within minutes of stimulation and de novo synthesized molecules acting as effectors in the relationship between nervous, vascular and immune systems. For this peculiarity, mast cells are master regulators and key players of the immune system and important sources of essential and beneficial mediators with crucial roles in regulating various physiological processes.


Subject(s)
Brain , Enteric Nervous System , Gastrointestinal Diseases , Gastrointestinal Microbiome , Inflammation , Mast Cells , Mental Disorders , Nervous System Diseases , Animals , Brain/immunology , Brain/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Enteric Nervous System/physiology , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Gastrointestinal Microbiome/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mental Disorders/immunology , Mental Disorders/metabolism , Nervous System Diseases/immunology , Nervous System Diseases/metabolism
10.
Immunity ; 54(3): 499-513.e5, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691135

ABSTRACT

The immune and enteric nervous (ENS) systems monitor the frontier with commensal and pathogenic microbes in the colon. We investigated whether FoxP3+ regulatory T (Treg) cells functionally interact with the ENS. Indeed, microbe-responsive RORγ+ and Helios+ subsets localized in close apposition to nitrergic and peptidergic nerve fibers in the colon lamina propria (LP). Enteric neurons inhibited in vitro Treg (iTreg) differentiation in a cell-contact-independent manner. A screen of neuron-secreted factors revealed a role for interleukin-6 (IL-6) in modulating iTreg formation and their RORγ+ proportion. Colonization of germfree mice with commensals, especially RORγ+ Treg inducers, broadly diminished colon neuronal density. Closing the triangle, conditional ablation of IL-6 in neurons increased total Treg cells but decreased the RORγ+ subset, as did depletion of two ENS neurotransmitters. Our findings suggest a regulatory circuit wherein microbial signals condition neuronal density and activation, thus tuning Treg cell generation and immunological tolerance in the gut.


Subject(s)
Enteric Nervous System/immunology , Interleukin-6/metabolism , Intestines/immunology , Neurons/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Gastrointestinal Microbiome , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype
11.
Trends Microbiol ; 29(8): 686-699, 2021 08.
Article in English | MEDLINE | ID: mdl-33309188

ABSTRACT

The gastrointestinal tract harbors an intrinsic neuronal network, the enteric nervous system (ENS). The ENS controls motility, fluid homeostasis, and blood flow, but also interacts with other components of the intestine such as epithelial and immune cells. Recent studies indicate that gut microbiota diversification, which occurs alongside postnatal ENS maturation, could be critical for the development and function of the ENS. Here we discuss the possibility that this functional relationship starts in utero, whereby the maternal microbiota would prime the developing ENS and shape its physiology. We review ENS/microbiota interactions and their modulation in physiological and pathophysiological contexts. While microbial modulation of the ENS physiology is now well established, further studies are required to understand the contribution of the gut microbiota to the development and pathology of the ENS and to reveal the precise mechanisms underlying microbiota-to-ENS communications.


Subject(s)
Enteric Nervous System/physiology , Gastrointestinal Microbiome/genetics , Gene Expression Regulation, Bacterial , Homeostasis , Enteric Nervous System/immunology , Enteric Nervous System/microbiology , Gastrointestinal Microbiome/physiology , Humans , Intestines/microbiology , Neurons/physiology
12.
Int J Mol Sci ; 21(24)2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33353157

ABSTRACT

Acrylamide is one of the harmful substances present in food. The present study aimed to establish the effect of acrylamide supplementation in tolerable daily intake (TDI) dose (0.5 µg/kg b.w./day) and a dose ten times higher than TDI (5 µg/kg b.w./day) on the population of vasoactive intestinal peptide-like immunoreactive (VIP-LI) neurons in the porcine small intestine and the degree of the co-localization of VIP with other neuroactive substances (neuronal nitric oxide synthase (nNOS), substance P (SP), and cocaine- and amphetamine-regulated transcript peptide (CART)). In our work, 15 Danish landrace gilts (5 in each experimental group) received capsules (empty or with low or high doses of acrylamide) for a period of 28 days with their morning feeding. Using double immunofluorescence staining, we established that acrylamide supplementation increased the number of neurons showing immunoreactivity towards VIP in all types of enteric nervous system (ENS) plexuses and fragments of the small intestine studied. Moreover, both doses of acrylamide led to changes in the degree of co-localization of VIP with nNOS, SP, and CART in intramural neurons. The observed changes may be the adaptation of neurons to local inflammation, oxidative stress, or the direct toxic effects of acrylamide on intestinal neurons, also referred to as neuronal plasticity.


Subject(s)
Acrylamide/pharmacology , Enteric Nervous System/cytology , Intestine, Small/cytology , Neurons/cytology , Vasoactive Intestinal Peptide/immunology , Animals , Dietary Supplements , Enteric Nervous System/drug effects , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Intestine, Small/drug effects , Intestine, Small/immunology , Intestine, Small/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Substance P/metabolism , Swine
13.
Int J Mol Sci ; 21(23)2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33276665

ABSTRACT

Over recent years, several investigations have suggested that Parkinson's disease (PD) can be regarded as the consequence of a bowel disorder. Indeed, gastrointestinal symptoms can occur at all stages of this neurodegenerative disease and in up to a third of cases, their onset can precede the involvement of the central nervous system. Recent data suggest that enteric glial cells (EGCs) may play a major role in PD-related gastrointestinal disturbances, as well as in the development and progression of the central disease. In addition to their trophic and structural functions, EGCs are crucial for the homeostatic control of a wide range of gastrointestinal activities. The main purpose of this review was to provide a detailed overview of the role of EGCs in intestinal PD-associated alterations, with particular regard for their participation in digestive and central inflammation as well as the dynamic interactions between glial cells and intestinal epithelial barrier. Accumulating evidence suggests that several pathological intestinal conditions, associated with an impairment of barrier permeability, may trigger dysfunctions of EGCs and their shift towards a proinflammatory phenotype. The reactive gliosis is likely responsible for PD-related neuroinflammation and the associated pathological changes in the ENS. Thus, ameliorating the efficiency of mucosal barrier, as well as avoiding IEB disruption and the related reactive gliosis, might theoretically prevent the onset of PD or, at least, counteract its progression.


Subject(s)
Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Neuroglia/immunology , Neuroglia/metabolism , Parkinson Disease/etiology , Parkinson Disease/metabolism , Signal Transduction , Animals , Brain/metabolism , Gastrointestinal Microbiome/immunology , Humans , alpha-Synuclein/metabolism
14.
Cell Mol Life Sci ; 77(22): 4505-4522, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32424438

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.


Subject(s)
Enteric Nervous System/physiology , Signal Transduction/physiology , Animals , Brain/immunology , Brain/physiology , Enteric Nervous System/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiology , Humans , Immune System/immunology , Immune System/physiology , Neuroglia/immunology , Neuroglia/physiology , Neurons/immunology , Neurons/physiology , Signal Transduction/immunology
15.
Int J Mol Sci ; 21(9)2020 May 11.
Article in English | MEDLINE | ID: mdl-32403316

ABSTRACT

Colorectal cancer (CRC) invasion within the large intestine wall results in the replacement of normal tissue architecture by tumour mass. Cancer cells digest the extracellular matrix (ECM) by the release of proteolytic enzymes. The disintegration of matrix ground substance activates several deposited growth factors which stimulate cell proliferation. Stromal (mainly fibroblasts), immune and cancer cells dominate in this area and become involved in a network of multimodal interactions which significantly induce proliferation of colon cancer cells, inhibit their apoptosis and promote their spreading within the local tumour microenvironment. Cancer invasion destroys nerve fibres and neurons of the local enteric nervous system (ENS) and induces subsequent atrophy of the submucosal and myenteric plexuses in areas adjacent to the cancer boundary. Interestingly, the reduction of plexuses' size is accompanied by the increased number of galanin-immunoreactive neurons and increased galanin content in parts of the colon located close to the tumour. Galanin, a neuroprotective peptide, may inhibit the extrinsic pathway of apoptosis and in this way promote cancer cell survival. The possible role of acetylcholine and some ENS neuropeptides was also discussed. Invasion of cancer cells spreads along nerve fibres with the involvement of locally-released neutrophins which promote, via their specific receptors, cancer cell proliferation and pro-survival signalling pathways. Thus, during CRC development cancer cells and neurons of the ENS release many neurotransmitters/neuropeptides which affect key cellular signalling pathways promoting cancer cell proliferation and pro-survival phenotype. The multiple interactions between ENS neurons, cancer cells and other cell types present in the colon wall increase cancer cell invasiveness and have a negative impact on the course of CRC.


Subject(s)
Colorectal Neoplasms/immunology , Enteric Nervous System/immunology , Myenteric Plexus/immunology , Tumor Microenvironment/immunology , Animals , Atrophy/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Disease Progression , Enteric Nervous System/physiopathology , Feedback, Physiological , Humans , Myenteric Plexus/pathology , Myenteric Plexus/physiopathology , Neoplasm Invasiveness
16.
Curr Drug Targets ; 21(14): 1428-1439, 2020.
Article in English | MEDLINE | ID: mdl-32416686

ABSTRACT

Ulcerative colitis (UC) and Crohn's disease (CD) are pathological conditions with an unknown aetiology that are characterised by severe inflammation of the intestinal tract and collectively referred to as inflammatory bowel disease (IBD). Current treatments are mostly ineffective due to their limited efficacy or toxicity, necessitating surgical resection of the affected bowel. The management of IBD is hindered by a lack of prognostic markers for clinical inflammatory relapse. Intestinal inflammation associates with the infiltration of immune cells (leukocytes) into, or surrounding the neuronal ganglia of the enteric nervous system (ENS) termed plexitis or ganglionitis. Histological observation of plexitis in unaffected intestinal regions is emerging as a vital predictive marker for IBD relapses. Plexitis associates with alterations to the structure, cellular composition, molecular expression and electrophysiological function of enteric neurons. Moreover, plexitis often occurs before the onset of gross clinical inflammation, which may indicate that plexitis can contribute to the progression of intestinal inflammation. In this review, the bilateral relationships between the ENS and inflammation are discussed. These include the effects and mechanisms of inflammation-induced enteric neuronal loss and plasticity. Additionally, the role of enteric neurons in preventing antigenic/pathogenic insult and immunomodulation is explored. While all current treatments target the inflammatory pathology of IBD, interventions that protect the ENS may offer an alternative avenue for therapeutic intervention.


Subject(s)
Enteric Nervous System/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/therapy , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/therapy , Animals , Enteric Nervous System/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Neurons/immunology , Neurons/metabolism , Physical Therapy Modalities
17.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32341116

ABSTRACT

The orchestration of host immune responses to enteric bacterial pathogens is a complex process involving the integration of numerous signals, including from the nervous system. Despite the recent progress in understanding the contribution of neuroimmune interactions in the regulation of inflammation, the mechanisms and effects of this communication during enteric bacterial infection are only beginning to be characterized. As part of this neuroimmune communication, neurons specialized to detect painful or otherwise noxious stimuli can respond to bacterial pathogens. Highlighting the complexity of these systems, the immunological consequences of sensory neuron activation can be either host adaptive or maladaptive, depending on the pathogen and organ system. These are but one of many types of neuroimmune circuits, with the vagus nerve and sympathetic innervation of numerous organs now known to modulate immune cell function and therefore dictate immunological outcomes during health and disease. Here, we review the evidence for neuroimmune communication in response to bacterial pathogens, and then discuss the consequences to host morbidity and mortality during infection of the gastrointestinal tract.


Subject(s)
Enteric Nervous System/immunology , Enterobacteriaceae Infections/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Neuroimmunomodulation/genetics , Sensory Receptor Cells/immunology , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/immunology , Citrobacter/growth & development , Citrobacter/immunology , Enteric Nervous System/microbiology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/microbiology , Gene Expression Regulation/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Sensory Receptor Cells/microbiology , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/immunology , TRPV Cation Channels/genetics , TRPV Cation Channels/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
18.
J Infect Dis ; 221(12): 1978-1988, 2020 06 11.
Article in English | MEDLINE | ID: mdl-31960920

ABSTRACT

BACKGROUND: Neurons are an integral component of the immune system that functions to coordinate responses to bacterial pathogens. Sensory nociceptive neurons that can detect bacterial pathogens are found throughout the body with dense innervation of the intestinal tract. METHODS: In this study, we assessed the role of these nerves in the coordination of host defenses to Citrobacter rodentium. Selective ablation of nociceptive neurons significantly increased bacterial burden 10 days postinfection and delayed pathogen clearance. RESULTS: Because the sensory neuropeptide CGRP (calcitonin gene-related peptide) regulates host responses during infection of the skin, lung, and small intestine, we assessed the role of CGRP receptor signaling during C rodentium infection. Although CGRP receptor blockade reduced certain proinflammatory gene expression, bacterial burden and Il-22 expression was unaffected. CONCLUSIONS: Our data highlight that sensory nociceptive neurons exert a significant host protective role during C rodentium infection, independent of CGRP receptor signaling.


Subject(s)
Citrobacter rodentium/immunology , Enteric Nervous System/immunology , Enterobacteriaceae Infections/immunology , Host-Pathogen Interactions/immunology , Nociceptors/immunology , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Disease Models, Animal , Enteric Nervous System/cytology , Enteric Nervous System/drug effects , Enterobacteriaceae Infections/microbiology , Host-Pathogen Interactions/drug effects , Humans , Intestinal Mucosa/innervation , Intestinal Mucosa/microbiology , Intestine, Small/innervation , Intestine, Small/microbiology , Mice , Mice, Knockout , Nociceptors/drug effects , Nociceptors/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , TRPV Cation Channels/genetics
19.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31923399

ABSTRACT

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Subject(s)
Immunity, Mucosal/immunology , Interleukin-18/immunology , Intestinal Mucosa/immunology , Animals , Cytokines/immunology , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Epithelial Cells/immunology , Female , Goblet Cells/immunology , Interleukin-18/biosynthesis , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Male , Mice , Mice, Inbred C57BL , Neurons/immunology , Rats , Rats, Sprague-Dawley , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology
20.
Cell ; 180(1): 64-78.e16, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31923400

ABSTRACT

Enteric-associated neurons (EANs) are closely associated with immune cells and continuously monitor and modulate homeostatic intestinal functions, including motility and nutrient sensing. Bidirectional interactions between neuronal and immune cells are altered during disease processes such as neurodegeneration or irritable bowel syndrome. We investigated the effects of infection-induced inflammation on intrinsic EANs (iEANs) and the role of intestinal muscularis macrophages (MMs) in this context. Using murine models of enteric infections, we observed long-term gastrointestinal symptoms, including reduced motility and loss of excitatory iEANs, which was mediated by a Nlrp6- and Casp11-dependent mechanism, depended on infection history, and could be reversed by manipulation of the microbiota. MMs responded to luminal infection by upregulating a neuroprotective program via ß2-adrenergic receptor (ß2-AR) signaling and mediated neuronal protection through an arginase 1-polyamine axis. Our results identify a mechanism of neuronal death post-infection and point to a role for tissue-resident MMs in limiting neuronal damage.


Subject(s)
Intestinal Mucosa/immunology , Macrophages/immunology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic Agents , Animals , Arginase/metabolism , Caspases, Initiator/immunology , Caspases, Initiator/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Female , Gastrointestinal Diseases , Gastrointestinal Microbiome , Infections , Inflammation/immunology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestines/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microbiota , Neurons/physiology , Receptors, Adrenergic, beta-2/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...