Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 651
Filter
1.
Neuron ; 112(18): 2993-2995, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39326387

ABSTRACT

While the enteric nervous system (ENS) is highly dynamic during development, the extent to which it is capable of repair remains unclear. In this issue of Neuron, Stavely et al.1 show that enteric neurons can reinnervate damaged regions to regain functionality using a glial positioning system (GPS) as their guide.


Subject(s)
Enteric Nervous System , Nerve Regeneration , Neuroglia , Neuroglia/physiology , Enteric Nervous System/physiology , Enteric Nervous System/cytology , Animals , Nerve Regeneration/physiology , Neurites/physiology , Intestines/physiology , Humans
2.
Neuron ; 112(18): 3143-3160.e6, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39019043

ABSTRACT

Here, we establish that plasticity exists within the postnatal enteric nervous system by demonstrating the reinnervation potential of post-mitotic enteric neurons (ENs). Employing BAF53b-Cre mice for selective neuronal tracing, the reinnervation capabilities of mature postnatal ENs are shown across multiple model systems. Isolated ENs regenerate neurites in vitro, with neurite complexity and direction influenced by contact with enteric glial cells (EGCs). Nerve fibers from transplanted ENs exclusively interface and travel along EGCs within the muscularis propria. Resident EGCs persist after Cre-dependent ablation of ENs and govern the architecture of the myenteric plexus for reinnervating ENs, as shown by nerve fiber projection tracing. Transplantation and optogenetic experiments in vivo highlight the rapid reinnervation potential of post-mitotic neurons, leading to restored gut muscle contractile activity within 2 weeks. These studies illustrate the structural and functional reinnervation capacity of post-mitotic ENs and the critical role of EGCs in guiding and patterning their trajectories.


Subject(s)
Enteric Nervous System , Neuroglia , Neurons , Animals , Neuroglia/physiology , Enteric Nervous System/physiology , Enteric Nervous System/cytology , Mice , Neurons/physiology , Intestines/innervation , Intestines/physiology , Nerve Regeneration/physiology , Myenteric Plexus/cytology , Myenteric Plexus/physiology , Mice, Transgenic , Neurites/physiology
3.
J Vis Exp ; (207)2024 May 17.
Article in English | MEDLINE | ID: mdl-38829111

ABSTRACT

The human enteric nervous system, ENS, is a large network of glial and neuronal cell types with remarkable neurotransmitter diversity. The ENS controls bowel motility, enzyme secretion, and nutrient absorption and interacts with the immune system and the gut microbiome. Consequently, developmental and acquired defects of the ENS are responsible for many human diseases and may contribute to symptoms of Parkinson's disease. Limitations in animal model systems and access to primary tissue pose significant experimental challenges in studies of the human ENS. Here, a detailed protocol is presented for effective in vitro derivation of the ENS lineages from human pluripotent stem cells, hPSC, using defined culture conditions. Our protocol begins with directed differentiation of hPSCs to enteric neural crest cells within 15 days and yields diverse subtypes of functional enteric neurons within 30 days. This platform provides a scalable resource for developmental studies, disease modeling, drug discovery, and regenerative applications.


Subject(s)
Cell Differentiation , Enteric Nervous System , Neural Crest , Pluripotent Stem Cells , Humans , Enteric Nervous System/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Neural Crest/cytology , Cytological Techniques/methods , Neurons/cytology
4.
Dev Cell ; 59(13): 1689-1706.e8, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38636517

ABSTRACT

During enteric nervous system (ENS) development, pioneering wavefront enteric neural crest cells (ENCCs) initiate gut colonization. However, the molecular mechanisms guiding their specification and niche interaction are not fully understood. We used single-cell RNA sequencing and spatial transcriptomics to map the spatiotemporal dynamics and molecular landscape of wavefront ENCCs in mouse embryos. Our analysis shows a progressive decline in wavefront ENCC potency during migration and identifies transcription factors governing their specification and differentiation. We further delineate key signaling pathways (ephrin-Eph, Wnt-Frizzled, and Sema3a-Nrp1) utilized by wavefront ENCCs to interact with their surrounding cells. Disruptions in these pathways are observed in human Hirschsprung's disease gut tissue, linking them to ENS malformations. Additionally, we observed region-specific and cell-type-specific transcriptional changes in surrounding gut tissues upon wavefront ENCC arrival, suggesting their role in shaping the gut microenvironment. This work offers a roadmap of ENS development, with implications for understanding ENS disorders.


Subject(s)
Cell Movement , Enteric Nervous System , Neural Crest , Signal Transduction , Animals , Neural Crest/metabolism , Neural Crest/cytology , Mice , Enteric Nervous System/metabolism , Enteric Nervous System/embryology , Enteric Nervous System/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Cell Differentiation , Gene Expression Regulation, Developmental , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Humans
5.
Eur J Neurosci ; 59(10): 2465-2482, 2024 May.
Article in English | MEDLINE | ID: mdl-38487941

ABSTRACT

The enteric nervous system (ENS) comprises a complex network of neurons whereby a subset appears to be dopaminergic although the characteristics, roles, and implications in disease are less understood. Most investigations relating to enteric dopamine (DA) neurons rely on immunoreactivity to tyrosine hydroxylase (TH)-the rate-limiting enzyme in the production of DA. However, TH immunoreactivity is likely to provide an incomplete picture. This study herein provides a comprehensive characterization of DA neurons in the gut using a reporter mouse line, expressing a fluorescent protein (tdTomato) under control of the DA transporter (DAT) promoter. Our findings confirm a unique localization of DA neurons in the gut and unveil the discrete subtypes of DA neurons in this organ, which we characterized using both immunofluorescence and single-cell transcriptomics, as well as validated using in situ hybridization. We observed distinct subtypes of DAT-tdTomato neurons expressing co-transmitters and modulators across both plexuses; some of them likely co-releasing acetylcholine, while others were positive for a slew of canonical DAergic markers (TH, VMAT2 and GIRK2). Interestingly, we uncovered a seemingly novel population of DA neurons unique to the ENS which was ChAT/DAT-tdTomato-immunoreactive and expressed Grp, Calcb, and Sst. Given the clear heterogeneity of DAergic gut neurons, further investigation is warranted to define their functional signatures and decipher their implication in disease.


Subject(s)
Dopamine Plasma Membrane Transport Proteins , Dopaminergic Neurons , Enteric Nervous System , Animals , Mice , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Dopaminergic Neurons/metabolism , Enteric Nervous System/metabolism , Enteric Nervous System/cytology , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Mice, Transgenic , Tyrosine 3-Monooxygenase/metabolism , Vesicular Monoamine Transport Proteins/metabolism , Vesicular Monoamine Transport Proteins/genetics , Genes, Reporter
6.
Stem Cells Transl Med ; 13(5): 490-504, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38387006

ABSTRACT

Regenerative cell therapy to replenish the missing neurons and glia in the aganglionic segment of Hirschsprung disease represents a promising treatment option. However, the success of cell therapies for this condition are hindered by poor migration of the transplanted cells. This limitation is in part due to a markedly less permissive extracellular environment in the postnatal gut than that of the embryo. Coordinated interactions between enteric neural crest-derived cells (ENCDCs) and their local environment drive migration along the embryonic gut during development of the enteric nervous system. Modifying transplanted cells, or the postnatal extracellular environment, to better recapitulate embryonic ENCDC migration could be leveraged to improve the engraftment and coverage of stem cell transplants. We compared the transcriptomes of ENCDCs from the embryonic intestine to that of postnatal-derived neurospheres and identified 89 extracellular matrix (ECM)-associated genes that are differentially expressed. Agrin, a heparin sulfate proteoglycan with a known inhibitory effect on ENCDC migration, was highly over-expressed by postnatal-derived neurospheres. Using a function-blocking antibody and a shRNA-expressing lentivirus, we show that inhibiting agrin promotes ENCDC migration in vitro and following cell transplantation ex vivo and in vivo. This enhanced migration is associated with an increased proportion of GFAP + cells, whose migration is especially enhanced.


Subject(s)
Agrin , Cell Movement , Neural Stem Cells , Animals , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Mice , Agrin/metabolism , Enteric Nervous System/metabolism , Enteric Nervous System/cytology , Colon/metabolism , Colon/cytology , Neural Crest/metabolism , Neural Crest/cytology , Hirschsprung Disease/metabolism , Hirschsprung Disease/therapy , Stem Cell Transplantation/methods
7.
Nature ; 618(7966): 818-826, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37316669

ABSTRACT

Correct development and maturation of the enteric nervous system (ENS) is critical for survival1. At birth, the ENS is immature and requires considerable refinement to exert its functions in adulthood2. Here we demonstrate that resident macrophages of the muscularis externa (MMϕ) refine the ENS early in life by pruning synapses and phagocytosing enteric neurons. Depletion of MMϕ before weaning disrupts this process and results in abnormal intestinal transit. After weaning, MMϕ continue to interact closely with the ENS and acquire a neurosupportive phenotype. The latter is instructed by transforming growth factor-ß produced by the ENS; depletion of the ENS and disruption of transforming growth factor-ß signalling result in a decrease in neuron-associated MMϕ associated with loss of enteric neurons and altered intestinal transit. These findings introduce a new reciprocal cell-cell communication responsible for maintenance of the ENS and indicate that the ENS, similarly to the brain, is shaped and maintained by a dedicated population of resident macrophages that adapts its phenotype and transcriptome to the timely needs of the ENS niche.


Subject(s)
Enteric Nervous System , Intestines , Macrophages , Enteric Nervous System/cytology , Enteric Nervous System/growth & development , Enteric Nervous System/physiology , Intestines/innervation , Lymphotoxin-alpha/metabolism , Macrophages/metabolism , Macrophages/physiology , Neurons/physiology , Weaning , Cell Communication , Transcriptome , Phenotype , Phagocytosis , Synapses , Neuronal Plasticity , Gastrointestinal Transit
8.
Neurosci Lett ; 806: 137221, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37031943

ABSTRACT

Enteric glia are a unique population of peripheral neuroglia associated with the enteric nervous system (ENS) throughout the digestive tract. The emerging data from the latest glial biology studies unveiled enteric glia as a heterogenic population with plastic and adaptative abilities that display phenotypic and functional changes upon distinct extrinsic cues. This aspect is essential in the dynamic signaling that enteric glia engage with neurons and other neighboring cells within the intestinal wall, such as epithelial, endocrine, and immune cells to maintain local homeostasis. Likewise, enteric glia sense signals from luminal microbes, although the extent of this active communication is still unclear. In this minireview, we discuss the recent findings that support glia-microbes crosstalk in the intestine in health and disease, pointing out the critical aspects that require further investigation.


Subject(s)
Disease , Enteric Nervous System , Gastrointestinal Microbiome , Health , Neuroglia , Humans , Biodiversity , Enteric Nervous System/cytology , Enteric Nervous System/physiology , Enteric Nervous System/physiopathology , Gastrointestinal Microbiome/physiology , Host Microbial Interactions , Inflammation/microbiology , Neuroglia/physiology , Probiotics , Animals
9.
Cell Rep ; 42(3): 112194, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36857184

ABSTRACT

The enteric nervous system (ENS) consists of glial cells (EGCs) and neurons derived from neural crest precursors. EGCs retain capacity for large-scale neurogenesis in culture, and in vivo lineage tracing has identified neurons derived from glial cells in response to inflammation. We thus hypothesize that EGCs possess a chromatin structure poised for neurogenesis. We use single-cell multiome sequencing to simultaneously assess transcription and chromatin accessibility in EGCs undergoing spontaneous neurogenesis in culture, as well as small intestine myenteric plexus EGCs. Cultured EGCs maintain open chromatin at genomic loci accessible in neurons, and neurogenesis from EGCs involves dynamic chromatin rearrangements with a net decrease in accessible chromatin. A subset of in vivo EGCs, highly enriched within the myenteric ganglia and that persist into adulthood, have a gene expression program and chromatin state consistent with neurogenic potential. These results clarify the mechanisms underlying EGC potential for neuronal fate transition.


Subject(s)
Enteric Nervous System , Ganglia , Multiomics , Neurogenesis , Neuroglia , Single-Cell Analysis , Neuroglia/classification , Neuroglia/cytology , Neuroglia/metabolism , Neurogenesis/genetics , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , RNA/analysis , RNA/genetics , Ganglia/cytology , Male , Female , Animals , Mice , Enteric Nervous System/cytology , Single-Cell Gene Expression Analysis , Cell Culture Techniques , Intestine, Small/cytology , Weaning
10.
Development ; 148(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34758081

ABSTRACT

The developmental programs that build and sustain animal forms also encode the capacity to sense and adapt to the microbial world within which they evolved. This is abundantly apparent in the development of the digestive tract, which typically harbors the densest microbial communities of the body. Here, we review studies in human, mouse, zebrafish and Drosophila that are revealing how the microbiota impacts the development of the gut and its communication with the nervous system, highlighting important implications for human and animal health.


Subject(s)
Brain-Gut Axis/physiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/growth & development , Animals , Cell Lineage , Enteric Nervous System/cytology , Enteric Nervous System/growth & development , Enteric Nervous System/physiology , Gastrointestinal Motility , Gastrointestinal Tract/innervation , Gastrointestinal Tract/microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Neurons/cytology , Neurons/physiology
11.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Article in English | MEDLINE | ID: mdl-34593632

ABSTRACT

Glia in the central nervous system exert precise spatial and temporal regulation over neural circuitry on a synapse-specific basis, but it is unclear if peripheral glia share this exquisite capacity to sense and modulate circuit activity. In the enteric nervous system (ENS), glia control gastrointestinal motility through bidirectional communication with surrounding neurons. We combined glial chemogenetics with genetically encoded calcium indicators expressed in enteric neurons and glia to study network-level activity in the intact myenteric plexus of the proximal colon. Stimulation of neural fiber tracts projecting in aboral, oral, and circumferential directions activated distinct populations of enteric glia. The majority of glia responded to both oral and aboral stimulation and circumferential pathways, while smaller subpopulations were activated only by ascending and descending pathways. Cholinergic signaling functionally specifies glia to the descending circuitry, and this network plays an important role in repressing the activity of descending neural pathways, with some degree of cross-inhibition imposed upon the ascending pathway. Glial recruitment by purinergic signaling functions to enhance activity within ascending circuit pathways and constrain activity within descending networks. Pharmacological manipulation of glial purinergic and cholinergic signaling differentially altered neuronal responses in these circuits in a sex-dependent manner. Collectively, our findings establish that the balance between purinergic and cholinergic signaling may differentially control specific circuit activity through selective signaling between networks of enteric neurons and glia. Thus, enteric glia regulate the ENS circuitry in a network-specific manner, providing profound insights into the functional breadth and versatility of peripheral glia.


Subject(s)
Enteric Nervous System/physiology , Gastrointestinal Motility/physiology , Myenteric Plexus/physiology , Neuroglia/physiology , Animals , Cell Communication , Enteric Nervous System/cytology , Female , Male , Mice , Myenteric Plexus/cytology , Neuroglia/cytology , Neurons/cytology , Signal Transduction
13.
Acta Med Okayama ; 75(5): 549-556, 2021.
Article in English | MEDLINE | ID: mdl-34703037

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. The loss of nigrostriatal dopaminergic neurons produces its characteristic motor symptoms, but PD patients also have non-motor symptoms such as constipation and orthostatic hypotension. The pathological hallmark of PD is the presence of α-synuclein-containing Lewy bodies and neurites in the brain. However, the PD pathology is observed in not only the central nervous system (CNS) but also in parts of the peripheral nervous system such as the enteric nervous system (ENS). Since constipation is a typical prodromal non-motor symptom in PD, often preceding motor symptoms by 10-20 years, it has been hypothesized that PD pathology propagates from the ENS to the CNS via the vagal nerve. Discovery of pharmacological and other methods to halt this progression of neurodegeneration in PD has the potential to improve millions of lives. Astrocytes protect neurons in the CNS by secretion of neurotrophic and antioxidative factors. Similarly, astrocyte-like enteric glial cells (EGCs) are known to secrete neuroprotective factors in the ENS. In this article, we summarize the neuroprotective function of astrocytes and EGCs and discuss therapeutic strategies for the prevention of neurodegeneration in PD targeting neurotrophic and antioxidative molecules in glial cells.


Subject(s)
Antioxidants/metabolism , Central Nervous System/drug effects , Enteric Nervous System/drug effects , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Central Nervous System/cytology , Enteric Nervous System/cytology , Humans
14.
Int J Mol Sci ; 22(18)2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34575824

ABSTRACT

The development of the enteric nervous system (ENS) is highly modulated by the synchronized interaction between the enteric neural crest cells (ENCCs) and the neural stem cell niche comprising the gut microenvironment. Genetic defects dysregulating the cellular behaviour(s) of the ENCCs result in incomplete innervation and hence ENS dysfunction. Hirschsprung disease (HSCR) is a rare complex neurocristopathy in which the enteric neural crest-derived cells fail to colonize the distal colon. In addition to ENS defects, increasing evidence suggests that HSCR patients may have intrinsic defects in the niche impairing the extracellular matrix (ECM)-cell interaction and/or dysregulating the cellular niche factors necessary for controlling stem cell behaviour. The niche defects in patients may compromise the regenerative capacity of the stem cell-based therapy and advocate for drug- and niche-based therapies as complementary therapeutic strategies to alleviate/enhance niche-cell interaction. Here, we provide a summary of the current understandings of the role of the enteric neural stem cell niche in modulating the development of the ENS and in the pathogenesis of HSCR. Deciphering the contribution of the niche to HSCR may provide important implications to the development of regenerative medicine for HSCR.


Subject(s)
Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Hirschsprung Disease/genetics , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Stem Cell Niche , Animals , Biomarkers , Cell Differentiation , Disease Management , Disease Susceptibility , Endothelin-3/metabolism , Genetic Predisposition to Disease , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hirschsprung Disease/diagnosis , Hirschsprung Disease/metabolism , Hirschsprung Disease/therapy , Humans , Neural Crest/cytology , Neural Crest/metabolism , Receptor, Endothelin B/metabolism , Regenerative Medicine , Signal Transduction
15.
Nature ; 597(7875): 250-255, 2021 09.
Article in English | MEDLINE | ID: mdl-34497389

ABSTRACT

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Subject(s)
Aging , Enteric Nervous System/cytology , Fetus/cytology , Health , Intestines/cytology , Intestines/growth & development , Lymph Nodes/cytology , Lymph Nodes/growth & development , Adult , Animals , Child , Crohn Disease/pathology , Datasets as Topic , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/embryology , Humans , Intestines/embryology , Intestines/innervation , Lymph Nodes/embryology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Organogenesis , Receptors, IgG/metabolism , Signal Transduction , Spatio-Temporal Analysis , Time Factors
16.
Sci Rep ; 11(1): 17189, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433854

ABSTRACT

Neuronal nitric oxide synthase (nNOS) neurons play a fundamental role in inhibitory neurotransmission, within the enteric nervous system (ENS), and in the establishment of gut motility patterns. Clinically, loss or disruption of nNOS neurons has been shown in a range of enteric neuropathies. However, the effects of nNOS loss on the composition and structure of the ENS remain poorly understood. The aim of this study was to assess the structural and transcriptional consequences of loss of nNOS neurons within the murine ENS. Expression analysis demonstrated compensatory transcriptional upregulation of pan neuronal and inhibitory neuronal subtype targets within the Nos1-/- colon, compared to control C57BL/6J mice. Conventional confocal imaging; combined with novel machine learning approaches, and automated computational analysis, revealed increased interconnectivity within the Nos1-/- ENS, compared to age-matched control mice, with increases in network density, neural projections and neuronal branching. These findings provide the first direct evidence of structural and molecular remodelling of the ENS, upon loss of nNOS signalling. Further, we demonstrate the utility of machine learning approaches, and automated computational image analysis, in revealing previously undetected; yet potentially clinically relevant, changes in ENS structure which could provide improved understanding of pathological mechanisms across a host of enteric neuropathies.


Subject(s)
Enteric Nervous System/metabolism , Nitric Oxide Synthase Type I/genetics , Animals , Enteric Nervous System/cytology , Machine Learning , Mice , Mice, Inbred C57BL , Nerve Net/cytology , Nerve Net/metabolism , Neurons/cytology , Neurons/metabolism , Nitric Oxide Synthase Type I/deficiency
17.
Sci Rep ; 11(1): 15889, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354183

ABSTRACT

Enteric neural stem cells (ENSC) have been identified as a possible treatment for enteric neuropathies. After in vivo transplantation, ENSC and their derivatives have been shown to engraft within colonic tissue, migrate and populate endogenous ganglia, and functionally integrate with the enteric nervous system. However, the mechanisms underlying the integration of donor ENSC, in recipient tissues, remain unclear. Therefore, we aimed to examine ENSC integration using an adapted ex vivo organotypic culture system. Donor ENSC were obtained from Wnt1cre/+;R26RYFP/YFP mice allowing specific labelling, selection and fate-mapping of cells. YFP+ neurospheres were transplanted to C57BL6/J (6-8-week-old) colonic tissue and maintained in organotypic culture for up to 21 days. We analysed and quantified donor cell integration within recipient tissues at 7, 14 and 21 days, along with assessing the structural and molecular consequences of ENSC integration. We found that organotypically cultured tissues were well preserved up to 21-days in ex vivo culture, which allowed for assessment of donor cell integration after transplantation. Donor ENSC-derived cells integrated across the colonic wall in a dynamic fashion, across a three-week period. Following transplantation, donor cells displayed two integrative patterns; longitudinal migration and medial invasion which allowed donor cells to populate colonic tissue. Moreover, significant remodelling of the intestinal ECM and musculature occurred upon transplantation, to facilitate donor cell integration within endogenous enteric ganglia. These results provide critical evidence on the timescale and mechanisms, which regulate donor ENSC integration, within recipient gut tissue, which are important considerations in the future clinical translation of stem cell therapies for enteric disease.


Subject(s)
Colon/cytology , Intestinal Pseudo-Obstruction/therapy , Neural Stem Cells/cytology , Animals , Cell Culture Techniques/methods , Colon/physiology , Enteric Nervous System/cytology , Enteric Nervous System/physiology , Female , Intestinal Pseudo-Obstruction/physiopathology , Intestine, Small/cytology , Male , Mice , Mice, Inbred C57BL , Neural Crest/cytology , Neural Stem Cells/physiology , Neurons/cytology , Organoids/cytology , Organoids/metabolism , Stem Cell Transplantation/methods
18.
Cell Mol Gastroenterol Hepatol ; 12(4): 1215-1237, 2021.
Article in English | MEDLINE | ID: mdl-34166814

ABSTRACT

BACKGROUND & AIMS: Enteric glial cells express type II major histocompatibility complex (MHC-II) molecules in Crohn's disease and Chagas disease, but it is unclear whether the expressed molecules are functional. We examined the capabilities of enteric glia to act as an antigen-presenting cell in vivo and whether glial MHC-II has immunomodulatory effects. METHODS: We generated Sox10CreERT2;IABfl/fl mice to ablate MHC-II in enteric glia after exposure to tamoxifen. We measured phagocytic activity and autophagy activation to assess potential peptide sources loaded onto glial MHC-II and measured T- and B-lymphocyte activation and serum and colonic tissue cytokine levels to study enteric glial immunomodulatory capabilities. RESULTS: Enteric glia express MHC-II molecules in response to a subclinical dose of interferon-γ and lipopolysaccharide in vivo. Glial MHC-II expression contributes to effective B-lymphocyte and T-lymphocyte activation with marked effects on T-helper cell (Th)17 and regulatory T cell subtypes. No effect on Th1 or Th2 subtypes was observed. Enteric glial MHC-II does not have a major effect on serum or colonic tissue cytokine levels but may influence local cytokine levels. Glial MHC-II expression requires the activation of autophagy pathways, but activating autophagy alone is not sufficient to drive glial MHC-II expression. CONCLUSIONS: Enteric glia express MHC-II as a mechanism to tune intestinal immune responses. Glial autophagy is triggered in response to proinflammatory stimuli and induces glial antigen presentation, which functions to modulate the activation of T-lymphocyte subsets involved in tolerance. These observations suggest that enteric glia may express MHC-II to maintain immune homeostasis during inflammatory conditions such as Crohn's disease.


Subject(s)
Autophagy , Enteric Nervous System/cytology , Histocompatibility Antigens Class II/genetics , Lymphocyte Activation/immunology , Neuroglia/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cell Communication , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunophenotyping , Interferon-gamma/metabolism , Lipopolysaccharides/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Knockout , Models, Biological , Phagocytosis
19.
Neuroreport ; 32(10): 875-881, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34029286

ABSTRACT

OBJECTIVE: Enteric glial cells (EGCs) can activate multiple pathways to inhibit the deleterious effects of acute and chronic insults. Our aim was to test the effect of EGCs on hyperglycemia-induced neuron damage and its underlying intracellular mechanisms. METHODS: A coculture model composed of EGCs and neuroblastoma cells (SH-SY5Y) was established to examine glial-mediated neuroprotection under high glucose conditions. The cell counting assay kit CCK-8 was used to measure cell viability. Flow cytometry was used to measure the induction of reactive oxygen species (ROS), change of mitochondrial membrane potential (MMP), cell cycle distribution, and apoptosis. The expressions of cyclin D1, cyclin E2, Bax, cleaved caspase-3, AKT, p-AKT, GSK-3ß, and p-GSK-3ß were tested using western blot. RESULTS: Exposure to high glucose (≥35 mM) reduced the viability of SH-SY5Y cells in a concentration- and time-dependent manner. Meanwhile, enhanced ROS generation and decrease of MMP were observed in SH-SY5Y cells when treated with high glucose. Furthermore, high glucose also caused SH-SY5Y cells arrest in G2 phase and apoptosis, accompanied by decreasing cyclin D1 and E2, and upregulating Bax and cleaved caspase-3. Coculture EGC lines or EGC-conditioned medium with SH-SY5Y prevented the neurotoxic effects. The p-AKT/AKT and p-GSK-3ß/GSK-3ß ratios were dramatically decreased in SH-SY5Y cells after high glucose incubation, which was restored after coculture with EGCs. CONCLUSIONS: EGCs can protect neurons from hyperglycemia-induced injury by activating the Akt/GSK-3ß pathway.


Subject(s)
Enteric Nervous System/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hyperglycemia/metabolism , Neuroglia/metabolism , Neuroprotection/physiology , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Coculture Techniques , Enteric Nervous System/cytology , Enteric Nervous System/drug effects , Glucose/toxicity , Humans , Hyperglycemia/chemically induced , Neuroglia/drug effects , Neuroprotection/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Neurochem Res ; 46(7): 1781-1793, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33864170

ABSTRACT

Increasing evidences indicate that the enteric nervous system (ENS) and enteric glial cells (EGC) play important regulatory roles in intestinal inflammation. Mercaptopurine (6-MP) is a cytostatic compound clinically used for the treatment of inflammatory bowel diseases (IBD), such as ulcerative colitis and Crohn's disease. However, potential impacts of 6-MP on ENS response to inflammation have not been evaluated yet. In this study, we aimed to gain deeper insights into the profile of inflammatory mediators expressed by the ENS and on the potential anti-inflammatory impact of 6-MP in this context. Genome-wide expression analyses were performed on ENS primary cultures exposed to lipopolysaccharide (LPS) and 6-MP alone or in combination. Differential expression of main hits was validated by quantitative real-time PCR (qPCR) using a cell line for EGC. ENS cells expressed a broad spectrum of cytokines and chemokines of the C-X-C motif ligand (CXCL) family under inflammatory stress. Induction of Cxcl5 and Cxcl10 by inflammatory stimuli was confirmed in EGC. Inflammation-induced protein secretion of TNF-α and Cxcl5 was partly inhibited by 6-MP in ENS primary cultures but not in EGC. Further work is required to identify the cellular mechanisms involved in this regulation. These findings extend our knowledge of the anti-inflammatory properties of 6-MP related to the ENS and in particular of the EGC-response to inflammatory stimuli.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression/drug effects , Interleukin-1beta/genetics , Mercaptopurine/pharmacology , Neurons/drug effects , Tumor Necrosis Factor-alpha/genetics , Animals , Cells, Cultured , Enteric Nervous System/cytology , Inflammation/chemically induced , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Lipopolysaccharides , Mice , Rats , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL