1
|
Reiner S, Linda S, Ebrahim H, Patrick L, Sven W. The role of reactive enteric glia-macrophage interactions in acute and chronic inflammation. Neurogastroenterol Motil 2024:e14947. [PMID: 39428750 DOI: 10.1111/nmo.14947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/18/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Enteric glia are a heterogeneous population of peripheral glia within the enteric nervous system and play pivotal roles in gut homeostasis, tissue integrity, coordination of motility, and intestinal immune responses. Under physiological conditions, they communicate with enteric neurons to control intestinal motility. In contrast, enteric glia undergo reactive changes in response to inflammatory signals during enteric neuroinflammation and participate in immune control. In this state, these glia are called reactive enteric glia, which promote cytokine and chemokine secretion and perpetuate immune cell recruitment, thereby affecting disease progression. Interestingly, reactive glia exhibit a huge plasticity and adapt to or shape the immune environment towards a resolving phenotype during inflammation and neuropathies. Recent studies revealed a bidirectional communication between enteric glia and resident and infiltrating immune cells under healthy conditions and in the context of inflammation-based intestinal disorders and neuropathies. While recent reviews give a superb general overview of enteric glial reactivity, we herein discuss the latest evidence on enteric glial reactivity in two prominent inflammatory conditions: acute postoperative inflammation, resulting in postoperative ileus, and chronic inflammation in inflammatory bowel diseases. We define their plasticity during inflammation and the interplay between reactive enteric glia and intestinal macrophages. Finally, we sketch important questions that should be addressed to clarify further the impact of enteric glial reactivity on intestinal inflammation.
Collapse
Affiliation(s)
| | - Schneider Linda
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Hamza Ebrahim
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Leven Patrick
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Wehner Sven
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
2
|
Baidoo N, Sanger GJ. The human colon: Evidence for degenerative changes during aging and the physiological consequences. Neurogastroenterol Motil 2024:e14848. [PMID: 38887160 DOI: 10.1111/nmo.14848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/16/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The incidence of constipation increases among the elderly (>65 years), while abdominal pain decreases. Causes include changes in lifestyle (e.g., diet and reduced exercise), disease and medications affecting gastrointestinal functions. Degenerative changes may also occur within the colo-rectum. However, most evidence is from rodents, animals with relatively high rates of metabolism and accelerated aging, with considerable variation in time course. In humans, cellular and non-cellular changes in the aging intestine are poorly investigated. PURPOSE To examine all available studies which reported the effects of aging on cellular and tissue functions of human isolated colon, noting the region studied, sex and age of tissue donors and study size. The focus on human colon reflects the ability to access full-thickness tissue over a wide age range, compared with other gastrointestinal regions. Details are important because of natural human variability. We found age-related changes within the muscle, in the enteric and nociceptor innervation, and in the submucosa. Some involve all regions of colon, but the ascending colon appears more vulnerable. Changes can be cell- and sublayer-dependent. Mechanisms are unclear but may include development of "senescent-like" and associated inflammaging, perhaps associated with increased mucosal permeability to harmful luminal contents. In summary, reduced nociceptor innervation can explain diminished abdominal pain among the elderly. Degenerative changes within the colon wall may have little impact on symptoms and colonic functions, because of high "functional reserve," but are likely to facilitate the development of constipation during age-related challenges (e.g., lifestyle, disease, and medications), now operating against a reduced functional reserve.
Collapse
Affiliation(s)
- Nicholas Baidoo
- School of Life Sciences, University of Westminster, London, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth J Sanger
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
3
|
Wang X, Ding C, Li HB. The crosstalk between enteric nervous system and immune system in intestinal development, homeostasis and diseases. SCIENCE CHINA. LIFE SCIENCES 2024; 67:41-50. [PMID: 37672184 DOI: 10.1007/s11427-023-2376-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/19/2023] [Indexed: 09/07/2023]
Abstract
The gut is the largest digestive and absorptive organ, which is essential for induction of mucosal and systemic immune responses, and maintenance of metabolic-immune homeostasis. The intestinal components contain the epithelium, stromal cells, immune cells, and enteric nervous system (ENS), as well as the outers, such as gut microbiota, metabolites, and nutrients. The dyshomeostasis of intestinal microenvironment induces abnormal intestinal development and functions, even colon diseases including dysplasia, inflammation and tumor. Several recent studies have identified that ENS plays a crucial role in maintaining the immune homeostasis of gastrointestinal (GI) microenvironment. The crosstalk between ENS and immune cells, mainly macrophages, T cells, and innate lymphoid cells (ILCs), has been found to exert important regulatory roles in intestinal tissue programming, homeostasis, function, and inflammation. In this review, we mainly summarize the critical roles of the interactions between ENS and immune cells in intestinal homeostasis during intestinal development and diseases progression, to provide theoretical bases and ideas for the exploration of immunotherapy for gastrointestinal diseases with the ENS as potential novel targets.
Collapse
Affiliation(s)
- Xindi Wang
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenbo Ding
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hua-Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Li X, Wang J, Li L, Yang C, Zhao X, Yang B, Zhang P, Liu B, Li Y, Zhang Z, Duan R. Epstein-Barr virus: To be a trigger of autoimmune glial fibrillary acidic protein astrocytopathy? CNS Neurosci Ther 2023; 29:4139-4146. [PMID: 37458208 PMCID: PMC10651959 DOI: 10.1111/cns.14336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/30/2023] [Accepted: 06/18/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy is a novel autoimmune disease of central nervous system (CNS). It is unclear whether Epstein-Barr virus (EBV) is related to autoimmune GFAP astrocytopathy. OBJECTIVE To describe the clinical, laboratory, and imaging characteristics of patients with autoimmune GFAP astrocytopathy. METHODS The clinical, laboratory, and imaging findings of patients are presented. The levels of GFAP in CSF were detected by ELISA. T and B cell subsets in CSF were detected by flow cytometry. GFAP-IgG in serum and cerebrospinal fluid (CSF) were tested by cell-based assay (CBA) and tissue-based assay (TBA). RESULTS All three patients had fever, cognitive dysfunction, limb weakness, and positive GFAP-IgG with EBV infection in CSF. Enteric glia cells may involve in this disease. Typical imaging findings include the gadolinium enhancement of linear perivascular radial perpendicular to the ventricle, meningeal enhancement (especially in midbrain interpeduncal fossa), longitudinally extensive lesions involving spindle cords, and more T2/Flair-hyperintense lesions in the periventricular white matter at late stage. The patients had poor response to antiviral treatment and strong response to steroid pulse therapy. CONCLUSION EBV could induce CNS autoimmune response in autoimmune GFAP astrocytopathy. The detection of GFAP-IgG and EBV may facilitate the early diagnosis in these patients.
Collapse
Affiliation(s)
- Xiao‐Li Li
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
| | - Jun‐Yan Wang
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Liang‐Kang Li
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
| | - Chun‐Lin Yang
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
| | - Xue‐Lu Zhao
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Bing Yang
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
| | - Peng Zhang
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
| | - Bin Liu
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
| | - Yan‐Bin Li
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
| | - Zhao‐Xu Zhang
- Department of NeurologyPeking University People's HospitalBeijingChina
| | - Rui‐Sheng Duan
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
| |
Collapse
|
5
|
Mariant CL, Bacola G, Van Landeghem L. Mini-Review: Enteric glia of the tumor microenvironment: An affair of corruption. Neurosci Lett 2023; 814:137416. [PMID: 37572875 PMCID: PMC10967235 DOI: 10.1016/j.neulet.2023.137416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/07/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
The tumor microenvironment corresponds to a complex mixture of bioactive products released by local and recruited cells whose normal functions have been "corrupted" by cues originating from the tumor, mostly to favor cancer growth, dissemination and resistance to therapies. While the immune and the mesenchymal cellular components of the tumor microenvironment in colon cancer have been under intense scrutiny over the last two decades, the influence of the resident neural cells of the gut on colon carcinogenesis has only very recently begun to draw attention. The vast majority of the resident neural cells of the gastrointestinal tract belong to the enteric nervous system and correspond to enteric neurons and enteric glial cells, both of which have been understudied in the context of colon cancer development and progression. In this review, we especially discuss available evidence on enteric glia impact on colon carcinogenesis. To highlight "corrupted" functioning in enteric glial cells of the tumor microenvironment and its repercussion on tumorigenesis, we first review the main regulatory effects of enteric glial cells on the intestinal epithelium in homeostatic conditions and we next present current knowledge on enteric glia influence on colon tumorigenesis. We particularly examine how enteric glial cell heterogeneity and plasticity require further appreciation to better understand the distinct regulatory interactions enteric glial cell subtypes engage with the various cell types of the tumor, and to identify novel biological targets to block enteric glia pro-carcinogenic signaling.
Collapse
Affiliation(s)
- Chloe L Mariant
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| | - Gregory Bacola
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| | - Laurianne Van Landeghem
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
6
|
Sunardi M, Cirillo C. Mini-review: "Enteric glia functions in nervous tissue repair: Therapeutic target or tool?". Neurosci Lett 2023; 812:137360. [PMID: 37393007 DOI: 10.1016/j.neulet.2023.137360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
In the body, nerve tissue is not only present in the central nervous system, but also in the periphery. The enteric nervous system (ENS) is a highly organized intrinsic network of neurons and glial cells grouped to form interconnected ganglia. Glial cells in the ENS are a fascinating cell population: their neurotrophic role is well established, as well as their plasticity in specific circumstances. Gene expression profiling studies indicate that ENS glia retain neurogenic potential. The identification of neurogenic glial subtype(s) and the molecular basis of glia-derived neurogenesis may have profound biological and clinical implications. In this review, we discuss the potential of using gene-editing for ENS glia and cell transplantation as therapies for enteric neuropathies. Glia in the ENS: target or tool for nerve tissue repair?
Collapse
Affiliation(s)
- Mukhamad Sunardi
- Division of Neural Differentiation and Regeneration (NDR), Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan.
| | - Carla Cirillo
- Division of Neural Differentiation and Regeneration (NDR), Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan; Toulouse NeuroImaging Center (ToNIC), National Institute of Health and Medical Research (INSERM), Toulouse University Paul Sabatier, Toulouse, France.
| |
Collapse
|
7
|
Scantlen MD, Majd H, Fattahi F. Modeling enteric glia development, physiology and disease using human pluripotent stem cells. Neurosci Lett 2023; 811:137334. [PMID: 37315730 DOI: 10.1016/j.neulet.2023.137334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Enteric glia play an integral role in many functions of the gastrointestinal (GI) system, but they have not been characterized comprehensively compared to other cells of the gut. Enteric glia are a specialized type of neuroglia in the enteric nervous system (ENS) that support neurons and interact with other cells of the gut such as immune and epithelial cells. The ENS is diffusely spread throughout the GI tract, making it extremely difficult to access and manipulate. As a result, it has remained extremely understudied. Nevertheless, much more is known about enteric neurons than enteric glia despite the glia being 6 times more abundant in humans [1]. In the past two decades, our understanding of enteric glia has greatly expanded and their many roles in the gut have been described and reviewed elsewhere [2-5]. While the field has made substantial progress, there are still a multitude of open questions about enteric glia biology and their role in disease. Many of these questions have remained intractable due to technical limitations of currently available experimental models of the ENS. In this review, we describe the benefits and limitations of the models commonly used to study enteric glia and discuss the ways in which a human pluripotent stem cell (hPSC) derived enteric glia model could help advance the field.
Collapse
Affiliation(s)
- Megan D Scantlen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Homa Majd
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA 94110, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94110, USA.
| |
Collapse
|
8
|
Kuil LE, Kakiailatu NJ, Windster JD, Bindels E, Zink JT, van der Zee G, Hofstra RM, Shepherd IT, Melotte V, Alves MM. Unbiased characterization of the larval zebrafish enteric nervous system at a single cell transcriptomic level. iScience 2023; 26:107070. [PMID: 37426341 PMCID: PMC10329177 DOI: 10.1016/j.isci.2023.107070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/15/2022] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
The enteric nervous system (ENS) regulates many gastrointestinal functions including peristalsis, immune regulation and uptake of nutrients. Defects in the ENS can lead to severe enteric neuropathies such as Hirschsprung disease (HSCR). Zebrafish have proven to be fruitful in the identification of genes involved in ENS development and HSCR pathogenesis. However, composition and specification of enteric neurons and glial subtypes at larval stages, remains mainly unexplored. Here, we performed single cell RNA sequencing of zebrafish ENS at 5 days post-fertilization. We identified vagal neural crest progenitors, Schwann cell precursors, and four clusters of differentiated neurons. In addition, a previously unrecognized elavl3+/phox2bb-population of neurons and cx43+/phox2bb-enteric glia was found. Pseudotime analysis supported binary neurogenic branching of ENS differentiation, driven by a notch-responsive state. Taken together, we provide new insights on ENS development and specification, proving that the zebrafish is a valuable model for the study of congenital enteric neuropathies.
Collapse
Affiliation(s)
- Laura E. Kuil
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Naomi J.M. Kakiailatu
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Jonathan D. Windster
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
- Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Eric Bindels
- Department of Hematology, Erasmus MC, Rotterdam, the Netherlands
| | - Joke T.M. Zink
- Department of Hematology, Erasmus MC, Rotterdam, the Netherlands
| | - Gaby van der Zee
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Robert M.W. Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
| | | | - Veerle Melotte
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Maria M. Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, the Netherlands
| |
Collapse
|
9
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
10
|
Windster JD, Sacchetti A, Schaaf GJ, Bindels EM, Hofstra RM, Wijnen RM, Sloots CE, Alves MM. A combinatorial panel for flow cytometry-based isolation of enteric nervous system cells from human intestine. EMBO Rep 2023; 24:e55789. [PMID: 36852936 PMCID: PMC10074091 DOI: 10.15252/embr.202255789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 03/01/2023] Open
Abstract
Efficient isolation of neurons and glia from the human enteric nervous system (ENS) is challenging because of their rare and fragile nature. Here, we describe a staining panel to enrich ENS cells from the human intestine by fluorescence-activated cell sorting (FACS). We find that CD56/CD90/CD24 co-expression labels ENS cells with higher specificity and resolution than previous methods. Surprisingly, neuronal (CD24, TUBB3) and glial (SOX10) selective markers appear co-expressed by all ENS cells. We demonstrate that this contradictory staining pattern is mainly driven by neuronal fragments, either free or attached to glial cells, which are the most abundant cell types. Live neurons can be enriched by the highest CD24 and CD90 levels. By applying our protocol to isolate ENS cells for single-cell RNA sequencing, we show that these cells can be obtained with high quality, enabling interrogation of the human ENS transcriptome. Taken together, we present a selective FACS protocol that allows enrichment and discrimination of human ENS cells, opening up new avenues to study this complex system in health and disease.
Collapse
Affiliation(s)
- Jonathan D Windster
- Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Andrea Sacchetti
- Department of Pathology, Josephine Nefkens Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gerben J Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Eric Mj Bindels
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Robert Mw Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Rene Mh Wijnen
- Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Cornelius Ej Sloots
- Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
11
|
Baidoo N, Sanger GJ, Belai A. Effect of old age on the subpopulations of enteric glial cells in human descending colon. Glia 2023; 71:305-316. [PMID: 36128665 PMCID: PMC10087700 DOI: 10.1002/glia.24272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/06/2022]
Abstract
Old age is associated with a higher incidence of lower bowel conditions such as constipation. Recent evidence suggest that colonic motility may be influenced by enteric glial cells (EGCs). Little is known about the effect of aging on the subpopulation of EGCs in the human colon. We assessed and compared the pattern of distribution of EGCs in adult and elderly human colon. Human descending colon were obtained from 23 cancer patients comprising of adults (23-63 years; 6 male, 7 female) and elderly (66-81 year; 6 male, 4 female). Specimens were serially-sectioned and immunolabeled with anti-Sox-10, anti-S100 and anti-GFAP for morphometric analysis. Standardized procedures were utilized to ensure unbiased counting and densitometric evaluation of EGCs. The number of Sox-10 immunoreactive (IR) EGCs were unaltered with age in both the myenteric plexus (MP) (respectively, in adult and elderly patients, 1939 ± 82 and 1760 ± 44/mm length; p > .05) and submucosal plexus; there were no apparent differences between adult males and females. The density of S100-IR EGCs declined among the elderly in the circular muscle and within the MP per ganglionic area. In the adult colon, there were more S100-IR EGCs distributed in the circular muscle per unit area than the Taenia coli. There was little or no GFAP-IR EGCs in both adult and elderly colon. We concluded that aging of the human descending colon does not result in a loss of Sox-10-IR EGCs in the MP and SMP but reduces S100-IR EGCs density within the musculature. This alteration in myenteric EGCs density with age may contribute to colonic dysfunction.
Collapse
Affiliation(s)
- Nicholas Baidoo
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Gareth J Sanger
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Abi Belai
- School of Life and Health Sciences, University of Roehampton, London, UK
| |
Collapse
|
12
|
Zanoletti L, Valdata A, Nehlsen K, Faris P, Casali C, Cacciatore R, Sbarsi I, Carriero F, Arfini D, van Baarle L, De Simone V, Barbieri G, Raimondi E, May T, Moccia F, Bozzola M, Matteoli G, Comincini S, Manai F. Cytological, molecular, cytogenetic, and physiological characterization of a novel immortalized human enteric glial cell line. Front Cell Neurosci 2023; 17:1170309. [PMID: 37153631 PMCID: PMC10158601 DOI: 10.3389/fncel.2023.1170309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/22/2023] [Indexed: 05/10/2023] Open
Abstract
Enteric glial cells (EGCs), the major components of the enteric nervous system (ENS), are implicated in the maintenance of gut homeostasis, thereby leading to severe pathological conditions when impaired. However, due to technical difficulties associated with EGCs isolation and cell culture maintenance that results in a lack of valuable in vitro models, their roles in physiological and pathological contexts have been poorly investigated so far. To this aim, we developed for the first time, a human immortalized EGC line (referred as ClK clone) through a validated lentiviral transgene protocol. As a result, ClK phenotypic glial features were confirmed by morphological and molecular evaluations, also providing the consensus karyotype and finely mapping the chromosomal rearrangements as well as HLA-related genotypes. Lastly, we investigated the ATP- and acetylcholine, serotonin and glutamate neurotransmitters mediated intracellular Ca2+ signaling activation and the response of EGCs markers (GFAP, SOX10, S100β, PLP1, and CCL2) upon inflammatory stimuli, further confirming the glial nature of the analyzed cells. Overall, this contribution provided a novel potential in vitro tool to finely characterize the EGCs behavior under physiological and pathological conditions in humans.
Collapse
Affiliation(s)
- Lisa Zanoletti
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Aurora Valdata
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | | | - Pawan Faris
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Claudio Casali
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Rosalia Cacciatore
- Immunohematology and Transfusion Service, I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | - Ilaria Sbarsi
- Immunohematology and Transfusion Service, I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | - Francesca Carriero
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Davide Arfini
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Lies van Baarle
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Veronica De Simone
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Giulia Barbieri
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Elena Raimondi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | | | - Francesco Moccia
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | | | - Gianluca Matteoli
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Sergio Comincini
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Federico Manai
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
- *Correspondence: Federico Manai,
| |
Collapse
|
13
|
Shi CJ, Lian JJ, Zhang BW, Cha JX, Hua QH, Pi XP, Hou YJ, Xie X, Zhang R. TGFβR-1/ALK5 inhibitor RepSox induces enteric glia-to-neuron transition and influences gastrointestinal mobility in adult mice. Acta Pharmacol Sin 2023; 44:92-104. [PMID: 35794374 DOI: 10.1038/s41401-022-00932-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/30/2022] [Indexed: 01/18/2023] Open
Abstract
Promoting adult neurogenesis in the enteric nervous system (ENS) may be a potential therapeutic approach to cure enteric neuropathies. Enteric glial cells (EGCs) are the most abundant glial cells in the ENS. Accumulating evidence suggests that EGCs can be a complementary source to supply new neurons during adult neurogenesis in the ENS. In the brain, astrocytes have been intensively studied for their neuronal conversion properties, and small molecules have been successfully used to induce the astrocyte-to-neuron transition. However, research on glia-to-neuron conversion in the ENS is still lacking. In this study, we used GFAP-Cre:Rosa-tdTomato mice to trace glia-to-neuron transdifferentiation in the ENS in vivo and in vitro. We showed that GFAP promoter-driven tdTomato exclusively labelled EGCs and was a suitable marker to trace EGCs and their progeny cells in the ENS of adult mice. Interestingly, we discovered that RepSox or other ALK5 inhibitors alone induced efficient transdifferentiation of EGCs into neurons in vitro. Knockdown of ALK5 further confirmed that the TGFβR-1/ALK5 signalling pathway played an essential role in the transition of EGCs to neurons. RepSox-induced neurons were Calbindin- and nNOS-positive and displayed typical neuronal electrophysiological properties. Finally, we showed that administration of RepSox (3, 10 mg· kg-1 ·d-1, i.g.) for 2 weeks significantly promoted the conversion of EGCs to neurons in the ENS and influenced gastrointestinal motility in adult mice. This study provides a method for efficiently converting adult mouse EGCs into neurons by small-molecule compounds, which might be a promising therapeutic strategy for gastrointestinal neuropathy.
Collapse
Affiliation(s)
- Chang-Jie Shi
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jun-Jiang Lian
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bo-Wen Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jia-Xue Cha
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiu-Hong Hua
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiao-Ping Pi
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yu-Jun Hou
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xin Xie
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ru Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
14
|
The Enteric Glia and Its Modulation by the Endocannabinoid System, a New Target for Cannabinoid-Based Nutraceuticals? MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196773. [PMID: 36235308 PMCID: PMC9570628 DOI: 10.3390/molecules27196773] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022]
Abstract
The enteric nervous system (ENS) is a part of the autonomic nervous system that intrinsically innervates the gastrointestinal (GI) tract. Whereas enteric neurons have been deeply studied, the enteric glial cells (EGCs) have received less attention. However, these are immune-competent cells that contribute to the maintenance of the GI tract homeostasis through supporting epithelial integrity, providing neuroprotection, and influencing the GI motor function and sensation. The endogenous cannabinoid system (ECS) includes endogenous classical cannabinoids (anandamide, 2-arachidonoylglycerol), cannabinoid-like ligands (oleoylethanolamide (OEA) and palmitoylethanolamide (PEA)), enzymes involved in their metabolism (FAAH, MAGL, COX-2) and classical (CB1 and CB2) and non-classical (TRPV1, GPR55, PPAR) receptors. The ECS participates in many processes crucial for the proper functioning of the GI tract, in which the EGCs are involved. Thus, the modulation of the EGCs through the ECS might be beneficial to treat some dysfunctions of the GI tract. This review explores the role of EGCs and ECS on the GI tract functions and dysfunctions, and the current knowledge about how EGCs may be modulated by the ECS components, as possible new targets for cannabinoids and cannabinoid-like molecules, particularly those with potential nutraceutical use.
Collapse
|
15
|
Hibberd TJ, Yew WP, Dodds KN, Xie Z, Travis L, Brookes SJ, Costa M, Hu H, Spencer NJ. Quantification of CGRP-immunoreactive myenteric neurons in mouse colon. J Comp Neurol 2022; 530:3209-3225. [PMID: 36043843 DOI: 10.1002/cne.25403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/07/2022]
Abstract
Quantitative data of biological systems provide valuable baseline information for understanding pathology, experimental perturbations, and computational modeling. In mouse colon, calcitonin gene-related peptide (CGRP) is expressed by myenteric neurons with multiaxonal (Dogiel type II) morphology, characteristic of intrinsic primary afferent neurons (IPANs). Analogous neurons in other species and gut regions represent 5-35% of myenteric neurons. We aimed to quantify proportions of CGRP-immunopositive (CGRP+) myenteric neurons. Colchicine-treated wholemount preparations of proximal, mid, and distal colon were labeled for HuC/D, CGRP, nitric oxide synthase (NOS), and peripherin (Per). The pan-neuronal markers (Hu+/Per+) co-labeled 94% of neurons. Hu+/Per- neurons comprised ∼6%, but Hu-/Per+ cells were rare. Thus, quantification was based on Hu+ myenteric neurons (8576 total; 1225 ± 239 per animal, n = 7). CGRP+ cell bodies were significantly larger than the average of all Hu+ neurons (329 ± 13 vs. 261 ± 12 μm2 , p < .0001). CGRP+ neurons comprised 19% ± 3% of myenteric neurons without significant regional variation. NOS+ neurons comprised 42% ± 2% of myenteric neurons overall, representing a lower proportion in proximal colon, compared to mid and distal colon (38% ± 2%, 44% ± 2%, and 44% ± 3%, respectively). Peripherin immunolabeling revealed cell body and axonal morphology in some myenteric neurons. Whether all CGRP+ neurons were multiaxonal could not be addressed using peripherin immunolabeling. However, of 118 putatively multiaxonal neurons first identified based on peripherin immunoreactivity, all were CGRP+ (n = 4). In conclusion, CGRP+ myenteric neurons in mouse colon were comprehensively quantified, occurring within a range expected of a putative IPAN marker. All Per+ multiaxonal neurons, characteristic of Dogiel type II/IPAN morphology, were CGRP+.
Collapse
Affiliation(s)
- Timothy J Hibberd
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Wai Ping Yew
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Kelsi N Dodds
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lee Travis
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Simon J Brookes
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Marcello Costa
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
16
|
Yang W, Pham J, King SK, Newgreen DF, Young HM, Stamp LA, Hao MM. A Novel Method for Identifying the Transition Zone in Long-Segment Hirschsprung Disease: Investigating the Muscle Unit to Ganglion Ratio. Biomolecules 2022; 12:biom12081101. [PMID: 36008996 PMCID: PMC9406109 DOI: 10.3390/biom12081101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Hirschsprung disease (HSCR) is characterised by the absence of enteric ganglia along variable lengths of the distal bowel. Current gold standard treatment involves the surgical resection of the defective, aganglionic bowel. Clear and reliable distinction of the normoganglionated bowel from the transition zone is key for successful resection of the entire defective bowel, and the avoidance of subsequent postoperative complications. However, the intraoperative nature of the tissue analysis and the variability of patient samples, sample preparation, and operator objectivity, make reproducible identification of the transition zone difficult. Here, we have described a novel method for using muscle units as a distinctive landmark for quantifying the density of enteric ganglia in resection specimens from HSCR patients. We show that the muscle unit to ganglion ratio is greater in the transition zone when compared with the proximal, normoganglionated region for long-segment HSCR patients. Patients with short-segment HSCR were also investigated, however, the muscle unit to ganglion ratio was not significantly different in these patients. Immunohistochemical examination of individual ganglia showed that there were no differences in the proportions of either enteric neurons or glial cells through the different regions of the resected colon. In addition, we identified that the size of enteric ganglia was smaller for patients that went on to develop HSCR associated enterocolitis; although the density of ganglia, as determined by the muscle unit to ganglia ratio, was not different when compared with patients that had no further complications. This suggests that subtle changes in the enteric nervous system, even in the “normoganglionated” colon, could be involved in changes in immune function and subsequent bacterial dysbiosis.
Collapse
Affiliation(s)
- Wendy Yang
- Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, Australia
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10663, Taiwan
| | - Jenny Pham
- Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, Australia
| | - Sebastian K. King
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Department of Paediatric Surgery, The Royal Children’s Hospital, Parkville 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville 3010, Australia
| | | | - Heather M. Young
- Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, Australia
| | - Lincon A. Stamp
- Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, Australia
- Correspondence: (L.A.S.); (M.M.H.)
| | - Marlene M. Hao
- Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, Australia
- Correspondence: (L.A.S.); (M.M.H.)
| |
Collapse
|
17
|
Hacene S, Le Friec A, Desmoulin F, Robert L, Colitti N, Fitremann J, Loubinoux I, Cirillo C. Present and future avenues of cell-based therapy for brain injury: The enteric nervous system as a potential cell source. Brain Pathol 2022; 32:e13105. [PMID: 35773942 PMCID: PMC9425017 DOI: 10.1111/bpa.13105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/09/2022] [Indexed: 01/01/2023] Open
Abstract
Cell therapy is a promising strategy in the field of regenerative medicine; however, several concerns limit the effective clinical use, namely a valid cell source. The gastrointestinal tract, which contains a highly organized network of nerves called the enteric nervous system (ENS), is a valuable reservoir of nerve cells. Together with neurons and neuronal precursor cells, it contains glial cells with a well described neurotrophic potential and a newly identified neurogenic one. Recently, enteric glia is looked at as a candidate for cell therapy in intestinal neuropathies. Here, we present the therapeutic potential of the ENS as cell source for brain repair, too. The example of stroke is introduced as a brain injury where cell therapy appears promising. This disease is the first cause of handicap in adults. The therapies developed in recent years allow a partial response to the consequences of the disease. The only prospect of recovery in the chronic phase is currently based on rehabilitation. The urgency to offer other treatments is therefore tangible. In the first part of the review, some elements of stroke pathophysiology are presented. An update on the available therapeutic strategies is provided, focusing on cell‐ and biomaterial‐based approaches. Following, the ENS is presented with its anatomical and functional characteristics, focusing on glial cells. The properties of these cells are depicted, with particular attention to their neurotrophic and, recently identified, neurogenic properties. Finally, preliminary data on a possible therapeutic approach combining ENS‐derived cells and a biomaterial are presented.
Collapse
Affiliation(s)
- Sirine Hacene
- National Veterinary School of Toulouse, University of Toulouse, Toulouse, France.,Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Alice Le Friec
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France.,Department of Biological and Chemical Engineering-Medical Biotechnology, Aarhus University, Aarhus, Denmark
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Lorenne Robert
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Nina Colitti
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Juliette Fitremann
- Laboratoire des IMRCP, CNRS UMR 5623, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Isabelle Loubinoux
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Carla Cirillo
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| |
Collapse
|
18
|
Enteric glia: extent, cohesion, axonal contacts, membrane separations and mitochondria in Auerbach's ganglia of guinea pigs. Cell Tissue Res 2022; 389:409-426. [PMID: 35729372 PMCID: PMC9436829 DOI: 10.1007/s00441-022-03656-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/14/2022] [Indexed: 11/04/2022]
Abstract
Studied by electron microscopy and morphometry, Auerbach’s ganglia comprise nerve cell bodies that occupy ~ 40% of volume; of the neuropil, little over 30% is neural processes (axons, dendrites) and little less than 30% is glia (cell bodies, processes). The amount of surface membrane of neural elements only marginally exceeds that of glia. Glial cells extend laminar processes radially between axons, reaching the ganglion’s surface with specialized membrane domains. Nerve cells and glia are tightly associated, eliminating any free space in ganglia. Glia expands maximally its cell membrane with a minimum of cytoplasm, contacting a maximal number of axons, which, with their near-circular profile, have minimal surface for a given volume. Shape of glia is moulded by the neural elements (predominantly concave the first, predominantly convex the second); the glia extends its processes to maximize contact with neural elements. Yet, a majority of axons is not reached by glia and only few are wrapped by it. Despite the large number of cells, the glia is not sufficiently developed to wrap around or just contact many of the neural elements. Mitochondria are markedly fewer in glia than in neurons, indicating a lower metabolic rate. Compactness of ganglia, their near-circular profile, absence of spaces between elements and ability to withstand extensive deformation suggest strong adhesion between the cellular elements, holding them together and keeping them at a fixed distance. Many axonal varicosities, with vesicles and membrane densities, abut on non-specialized areas of glia, suggesting the possibility of neurotransmitters being released outside synaptic sites.
Collapse
|
19
|
Opioid Use, Gut Dysbiosis, Inflammation, and the Nervous System. J Neuroimmune Pharmacol 2022; 17:76-93. [PMID: 34993905 DOI: 10.1007/s11481-021-10046-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/17/2021] [Indexed: 12/29/2022]
Abstract
Opioid use disorder (OUD) is defined as the chronic use or misuse of prescribed or illicitly obtained opioids and is characterized by clinically significant impairment. The etiology of OUD is multifactorial as it is influenced by genetics, environmental factors, stress response and behavior. Given the profound role of the gut microbiome in health and disease states, in recent years there has been a growing interest to explore interactions between the gut microbiome and the central nervous system as a causal link and potential therapeutic source for OUD. This review describes the role of the gut microbiome and opioid-induced immunopathological disturbances at the gut epithelial surface, which collectively contribute to OUD and perpetuate the vicious cycle of addiction and relapse.
Collapse
|
20
|
Liu C, Yang J. Enteric Glial Cells in Immunological Disorders of the Gut. Front Cell Neurosci 2022; 16:895871. [PMID: 35573829 PMCID: PMC9095930 DOI: 10.3389/fncel.2022.895871] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Enteric glial cells (EGCs) are one of the major cell types of neural crest lineage distributed in the gastrointestinal tract. EGCs represent an integral part of the enteric nervous system (ENS) and significantly outnumber ENS neurons. Studies have suggested that EGCs would exert essential roles in supporting the survival and functions of the ENS neurons. Notably, recent evidence has begun to reveal that EGCs could possess multiple immune functions and thereby may participate in the immune homeostasis of the gut. In this review article, we will summarize the current evidence supporting the potential involvement of EGCs in several important immunological disorders, including inflammatory bowel disease, celiac disease, and autoimmune enteropathy. Further, we highlight critical questions on the immunological aspects of EGCs that warrant future research attention.
Collapse
Affiliation(s)
- Chang Liu
- Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Jing Yang
- Center for Life Sciences, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
- *Correspondence: Jing Yang
| |
Collapse
|
21
|
Sinegubov A, Andreeva D, Burzak N, Vasyutina M, Murashova L, Dyachuk V. Heterogeneity and Potency of Peripheral Glial Cells in Embryonic Development and Adults. Front Mol Neurosci 2022; 15:737949. [PMID: 35401107 PMCID: PMC8990813 DOI: 10.3389/fnmol.2022.737949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the heterogeneity of peripheral glial cell populations, from the emergence of Schwann cells (SCs) in early development, to their involvement, and that of their derivatives in adult glial populations. We focus on the origin of the first glial precursors from neural crest cells (NCCs), and their ability to differentiate into several cell types during development. We also discuss the heterogeneity of embryonic glia in light of the latest data from genetic tracing and transcriptome analysis. Special attention has been paid to the biology of glial populations in adult animals, by highlighting common features of different glial cell types and molecular differences that modulate their functions. Finally, we consider the communication of glial cells with axons of neurons in normal and pathological conditions. In conclusion, the present review details how information available on glial cell types and their functions in normal and pathological conditions may be utilized in the development of novel therapeutic strategies for the treatment of patients with neurodiseases.
Collapse
|
22
|
Goluba K, Kunrade L, Riekstina U, Parfejevs V. Schwann Cells in Digestive System Disorders. Cells 2022; 11:832. [PMID: 35269454 PMCID: PMC8908985 DOI: 10.3390/cells11050832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/18/2022] Open
Abstract
Proper functioning of the digestive system is ensured by coordinated action of the central and peripheral nervous systems (PNS). Peripheral innervation of the digestive system can be viewed as intrinsic and extrinsic. The intrinsic portion is mainly composed of the neurons and glia of the enteric nervous system (ENS), while the extrinsic part is formed by sympathetic, parasympathetic, and sensory branches of the PNS. Glial cells are a crucial component of digestive tract innervation, and a great deal of research evidence highlights the important status of ENS glia in health and disease. In this review, we shift the focus a bit and discuss the functions of Schwann cells (SCs), the glial cells of the extrinsic innervation of the digestive system. For more context, we also provide information on the basic findings regarding the function of innervation in disorders of the digestive organs. We find diverse SC roles described particularly in the mouth, the pancreas, and the intestine. We note that most of the scientific evidence concerns the involvement of SCs in cancer progression and pain, but some research identifies stem cell functions and potential for regenerative medicine.
Collapse
Affiliation(s)
| | | | | | - Vadims Parfejevs
- Faculty of Medicine, University of Latvia, House of Science, Jelgavas Str. 3, LV-1004 Riga, Latvia; (K.G.); (L.K.); (U.R.)
| |
Collapse
|
23
|
Enteric Glia and Enteric Neurons, Associated. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1383:179-190. [PMID: 36587157 DOI: 10.1007/978-3-031-05843-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Peripheral neurons are never found alone and are invariably accompanied by glial cells, with which they are intimately associated in compact, highly deformable structures.Myenteric ganglia of the guinea-pig, examined in situ by electron microscopy, show that in their neuropil (axons and dendrites, and glial cells and processes) the glia constitutes almost half of the volume and almost half of membrane extent.In the glia, the expanse of the cell membrane predominates over that of their cytoplasm, the opposite being the case with the neural elements.The profile of the glial elements is passive and is dictated by the surrounding elements, mainly the axons, and hence it is predominantly concave.The enteric glia is widely developed; however, it is not sufficient to form a full wrapping around all neurons and around all axons (unlike what is found in other autonomic ganglia).Glial processes are radially expanding laminae, irregularly tapering, branching, and penetrating between axons.Some processes have a specialized termination attached to the basal lamina of the ganglion.Mitochondria are markedly more abundant in neural element that in the glia (up to a factor of 2).Many expanded axons, laden with vesicles clustered beneath membrane sites, abut on glial cells and processes, while these show no matching structural specializations.
Collapse
|
24
|
Kovler ML, Gonzalez Salazar AJ, Fulton WB, Lu P, Yamaguchi Y, Zhou Q, Sampah M, Ishiyama A, Prindle T, Wang S, Jia H, Wipf P, Sodhi CP, Hackam DJ. Toll-like receptor 4-mediated enteric glia loss is critical for the development of necrotizing enterocolitis. Sci Transl Med 2021; 13:eabg3459. [PMID: 34550727 PMCID: PMC8859973 DOI: 10.1126/scitranslmed.abg3459] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease of premature infants, whose pathogenesis remains incompletely understood, although activation of the Gram-negative bacterial receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium plays a critical role. Patients with NEC typically display gastrointestinal dysmotility before systemic disease is manifest, suggesting that dysmotility could drive NEC development. Both intestinal motility and inflammation are governed by the enteric nervous system, a network of enteric neurons and glia. We hypothesized here that enteric glia loss in the premature intestine could lead to dysmotility, exaggerated TLR4 signaling, and NEC development. We found that intestinal motility is reduced early in NEC in mice, preceding the onset of intestinal inflammation, whereas pharmacologic restoration of intestinal motility reduced NEC severity. Ileal samples from mouse, piglet, and human NEC revealed enteric glia depletion, and glia-deficient mice (Plp1ΔDTR, Sox10ΔDTR, and BdnfΔDTR) showed increased NEC severity compared with wild-type mice. Mice lacking TLR4 on enteric glia (Sox10-Tlr4ko) did not show NEC-induced enteric glia depletion and were protected from NEC. Mechanistically, brain-derived neurotrophic factor (BDNF) from enteric glia restrained TLR4 signaling on the intestine to prevent NEC. BDNF was reduced in mouse and human NEC, and BDNF administration reduced both TLR4 signaling and NEC severity in enteric glia–deficient mice. Last, we identified an agent (J11) that enhanced enteric glial BDNF release, inhibited intestinal TLR4, restored motility, and prevented NEC in mice. Thus, enteric glia loss might contribute to NEC through intestinal dysmotility and increased TLR4 activation, suggesting enteric glia therapies for this disorder.
Collapse
Affiliation(s)
- Mark L. Kovler
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Andres J. Gonzalez Salazar
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - William B. Fulton
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Peng Lu
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Yukihiro Yamaguchi
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Qinjie Zhou
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Maame Sampah
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Asuka Ishiyama
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Thomas Prindle
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Sanxia Wang
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Hongpeng Jia
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15260
| | - Chhinder P. Sodhi
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| | - David J. Hackam
- Division of Pediatric Surgery, Johns Hopkins University School of Medicine and the Johns Hopkins Children’s Center, Baltimore, MD 21287
| |
Collapse
|
25
|
Abstract
Glia, the non-neuronal cells of the nervous system, were long considered secondary cells only necessary for supporting the functions of their more important neuronal neighbors. Work by many groups over the past two decades has completely overturned this notion, revealing the myriad and vital functions of glia in nervous system development, plasticity, and health. The largest population of glia outside the brain is in the enteric nervous system, a division of the autonomic nervous system that constitutes a key node of the gut-brain axis. Here, we review the latest in the understanding of these enteric glia in mammals with a focus on their putative roles in human health and disease.
Collapse
Affiliation(s)
- Harry J. Rosenberg
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Nutraceuticals and Enteric Glial Cells. Molecules 2021; 26:molecules26123762. [PMID: 34205534 PMCID: PMC8234579 DOI: 10.3390/molecules26123762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
Until recently, glia were considered to be a structural support for neurons, however further investigations showed that glial cells are equally as important as neurons. Among many different types of glia, enteric glial cells (EGCs) found in the gastrointestinal tract, have been significantly underestimated, but proved to play an essential role in neuroprotection, immune system modulation and many other functions. They are also said to be remarkably altered in different physiopathological conditions. A nutraceutical is defined as any food substance or part of a food that provides medical or health benefits, including prevention and treatment of the disease. Following the description of these interesting peripheral glial cells and highlighting their role in physiological and pathological changes, this article reviews all the studies on the effects of nutraceuticals as modulators of their functions. Currently there are only a few studies available concerning the effects of nutraceuticals on EGCs. Most of them evaluated molecules with antioxidant properties in systemic conditions, whereas only a few studies have been performed using models of gastrointestinal disorders. Despite the scarcity of studies on the topic, all agree that nutraceuticals have the potential to be an interesting alternative in the prevention and/or treatment of enteric gliopathies (of systemic or local etiology) and their associated gastrointestinal conditions.
Collapse
|
27
|
Li Y, Wang Y, Chang H, Cheng B, Miao J, Li S, Hu H, Huang L, Wang Q. Inhibitory Effects of Dexmedetomidine and Propofol on Gastrointestinal Tract Motility Involving Impaired Enteric Glia Ca 2+ Response in Mice. Neurochem Res 2021; 46:1410-1422. [PMID: 33656693 DOI: 10.1007/s11064-021-03280-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/20/2021] [Accepted: 02/20/2021] [Indexed: 12/31/2022]
Abstract
Propofol and dexmedetomidine are popular used for sedation in ICU, however, inadequate attention has been paid to their effect on gastrointestinal tract (GIT) motility. Present study aimed to compare the effect of propofol and dexmedetomidine on GIT motility at parallel level of sedation and explore the possible mechanism. Male C57BL/6 mice (8-10 weeks) were randomly divided into control, propofol and dexmedetomidine group. After intraperitoneal injection of propofol or dexmedetomidine, comparable sedative level was confirmed by sedative score, physiological parameters and electroencephalogram (EEG). Different segments of GIT motility in vivo (gastric emptying, small intestine transit, distal colon bead expulsion, stool weight and number of fecal pellets, gastrointestinal transit and whole gut transit time) and colonic migrating motor complexes (CMMCs) pattern in vitro were evaluated. The Ca2+ response of primary enteric glia was examined under the treatment of propofol or dexmedetomidine. There is little difference in physiological parameters and composite permutation entropy index (CPEI) between administration of 50 mg/kg propofol and 40 μg/kg dexmedetomidine, indicated that parallel level of sedation was reached. Data showed that propofol and dexmedetomidine had significantly inhibitory effect on GIT motility while dexmedetomidine was stronger. Also, the amplitude (ΔF/F0) of Ca2+ response in primary enteric glia was attenuated after treated with the sedatives while the effect of dexmedetomidine was greater than propofol. These findings demonstrated that dexmedetomidine caused stronger inhibitory effects on GIT motility in sedative mice, which may involve impaired Ca2+ response in enteric glia. Hence, dexmedetomidine should be carefully applied especially for potential GIT dysmotility patient.
Collapse
Affiliation(s)
- Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yubo Wang
- School of Life Science and Technology, Xidian University, Xi'an, 710061, Shaanxi, China
| | - Haiqing Chang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiwen Miao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shuang Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liyu Huang
- School of Life Science and Technology, Xidian University, Xi'an, 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
28
|
Gao H, Zhang Y, Li Y, Chang H, Cheng B, Li N, Yuan W, Li S, Wang Q. μ-Opioid Receptor-Mediated Enteric Glial Activation Is Involved in Morphine-Induced Constipation. Mol Neurobiol 2021; 58:3061-3070. [PMID: 33624141 DOI: 10.1007/s12035-021-02286-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/08/2021] [Indexed: 01/04/2023]
Abstract
Among all the side effects, opioid-induced constipation (OIC) has the highest incidence rate in people who take chronic opioid therapy. Increasing evidence shows that enteric glial cells (EGCs) play a pivotal role in the modulation of gastrointestinal motility. We aim to investigate whether EGCs are involved in OIC and possible mechanisms. Eight-week male C57BL/6 mice were randomized into four groups: the control group, the morphine group, the gliotoxin fluorocitrate (FC) group, and the FC plus morphine group. OIC was induced by injection of morphine subcutaneously. Colonic motility was evaluated by in vivo motility assays and colonic migrating motor complex (CMMC) in vitro. Both the Ca2+ responses and the release of inflammatory cytokine by EGCs were detected in vitro. Proteins were detected by immunofluorescence staining and Western blot. The morphine group showed prolonged gastrointestinal motility compared with the control group. Once EGCs were disrupted by FC, such inhibitory effect was abolished. There was a remarkable enhancement of the GFAP expression on colonic EGCs. Immunofluorescence exhibited that μ-opioid receptor (MOR) collocated with GFAP, indicating the existence of MOR in EGCs. Moreover, morphine activated the EGCs significantly through enhancing GFAP expression and Ca2+ amplitude. Both effects can be reversed by MOR-siRNA. Morphine treatment elevated the enteric glial release of proinflammatory cytokines notably and this effect was abolished when EGCs were silenced by MOR-siRNA. The activation of EGCs via MOR and the increased proinflammatory cytokine from EGCs may be involved in morphine-induced constipation. These results provided a potential therapeutic target for OIC.
Collapse
Affiliation(s)
- Hui Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Yuxin Zhang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Haiqing Chang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Na Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Wei Yuan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Shuang Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
29
|
Ohno M, Nikaido M, Horiuchi N, Kawakami K, Hatta K. The enteric nervous system in zebrafish larvae can regenerate via migration into the ablated area and proliferation of neural crest-derived cells. Development 2021; 148:dev.195339. [PMID: 33376126 DOI: 10.1242/dev.195339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/10/2020] [Indexed: 12/21/2022]
Abstract
The enteric nervous system (ENS), which is derived from neural crest, is essential for gut function, and its deficiency causes severe congenital diseases. Since the capacity for ENS regeneration in mammals is limited, additional complementary models would be useful. Here, we show that the ENS in zebrafish larvae at 10-15 days postfertilization is highly regenerative. After laser ablation, the number of enteric neurons recovered to ∼50% of the control by 10 days post-ablation (dpa). Using transgenic lines in which enteric neural crest-derived cells (ENCDCs) and enteric neurons are labeled with fluorescent proteins, we live imaged the regeneration process and found covering by neurites that extended from the unablated area and entry of ENCDCs into the ablated areas by 1-3 dpa. BrdU assays suggested that ∼80% of the enteric neurons and ∼90% of the Sox10-positive ENCDCs therein at 7 dpa were generated through proliferation. Thus, ENS regeneration involves proliferation, entrance and neurogenesis of ENCDCs. This is the first report regarding the regeneration process of the zebrafish ENS. Our findings provide a basis for further in vivo research at single-cell resolution in this vertebrate model.
Collapse
Affiliation(s)
- Maria Ohno
- Graduate School of Life Science, University of Hyogo, Ako-gun, Hyogo 678-1297, Japan
| | - Masataka Nikaido
- Graduate School of Life Science, University of Hyogo, Ako-gun, Hyogo 678-1297, Japan
| | - Natsumi Horiuchi
- School of Science, University of Hyogo, Ako-gun, Hyogo 678-1297, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Kohei Hatta
- Graduate School of Life Science, University of Hyogo, Ako-gun, Hyogo 678-1297, Japan
| |
Collapse
|
30
|
Enteric Glia at the Crossroads between Intestinal Immune System and Epithelial Barrier: Implications for Parkinson Disease. Int J Mol Sci 2020; 21:ijms21239199. [PMID: 33276665 PMCID: PMC7730281 DOI: 10.3390/ijms21239199] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
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.
Collapse
|
31
|
Abstract
The gut-brain axis is a coordinated communication system that not only maintains homeostasis, but significantly influences higher cognitive functions and emotions, as well as neurological and behavioral disorders. Among the large populations of sensory and motor neurons that innervate the gut, insights into the function of primary afferent nociceptors, whose cell bodies reside in the dorsal root ganglia and nodose ganglia, have revealed their multiple crosstalk with several cell types within the gut wall, including epithelial, vascular, and immune cells. These bidirectional communications have immunoregulatory functions, control host response to pathogens, and modulate sensations associated with gastrointestinal disorders, through activation of immune cells and glia in the peripheral and central nervous system, respectively. Here, we will review the cellular and neurochemical basis of these interactions at the periphery, in dorsal root ganglia, and in the spinal cord. We will discuss the research gaps that should be addressed to get a better understanding of the multifunctional role of sensory neurons in maintaining gut homeostasis and regulating visceral sensitivity.
Collapse
Affiliation(s)
- Nasser Abdullah
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Graham KD, López SH, Sengupta R, Shenoy A, Schneider S, Wright CM, Feldman M, Furth E, Valdivieso F, Lemke A, Wilkins BJ, Naji A, Doolin E, Howard MJ, Heuckeroth RO. Robust, 3-Dimensional Visualization of Human Colon Enteric Nervous System Without Tissue Sectioning. Gastroenterology 2020; 158:2221-2235.e5. [PMID: 32113825 PMCID: PMC7392351 DOI: 10.1053/j.gastro.2020.02.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Small, 2-dimensional sections routinely used for human pathology analysis provide limited information about bowel innervation. We developed a technique to image human enteric nervous system (ENS) and other intramural cells in 3 dimensions. METHODS Using mouse and human colon tissues, we developed a method that combines tissue clearing, immunohistochemistry, confocal microscopy, and quantitative analysis of full-thickness bowel without sectioning to quantify ENS and other intramural cells in 3 dimensions. RESULTS We provided 280 adult human colon confocal Z-stacks from persons without known bowel motility disorders. Most of our images were of myenteric ganglia, captured using a 20× objective lens. Full-thickness colon images, viewed with a 10× objective lens, were as large as 4 × 5 mm2. Colon from 2 pediatric patients with Hirschsprung disease was used to show distal colon without enteric ganglia, as well as a transition zone and proximal pull-through resection margin where ENS was present. After testing a panel of antibodies with our method, we identified 16 antibodies that bind to molecules in neurons, glia, interstitial cells of Cajal, and muscularis macrophages. Quantitative analyses demonstrated myenteric plexus in 24.5% ± 2.4% of flattened colon Z-stack area. Myenteric ganglia occupied 34% ± 4% of myenteric plexus. Single myenteric ganglion volume averaged 3,527,678 ± 573,832 mm3 with 38,706 ± 5763 neuron/mm3 and 129,321 ± 25,356 glia/mm3. Images of large areas provided insight into why published values of ENS density vary up to 150-fold-ENS density varies greatly, across millimeters, so analyses of small numbers of thin sections from the same bowel region can produce varying results. Neuron subtype analysis revealed that approximately 56% of myenteric neurons stained with neuronal nitric oxide synthase antibody and approximately 33% of neurons produce and store acetylcholine. Transition zone regions from colon tissues of patients with Hirschsprung disease had ganglia in multiple layers and thick nerve fiber bundles without neurons. Submucosal neuron distribution varied among imaged colon regions. CONCLUSIONS We developed a 3-dimensional imaging method for colon that provides more information about ENS structure than tissue sectioning. This approach could improve diagnosis for human bowel motility disorders and may be useful for other bowel diseases as well.
Collapse
Affiliation(s)
- Kahleb D. Graham
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Cincinnati Children’s Hospital Medical Center and the Department of Pediatrics at University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Silvia Huerta López
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318
| | - Rajarshi Sengupta
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,American Association for Cancer Research, 615 Chestnut Street, 17th Floor, Philadelphia, PA 19106-4404
| | - Archana Shenoy
- Department of Pathology, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A., 19104-4318
| | - Sabine Schneider
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| | - Christina M. Wright
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| | - Michael Feldman
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Emma Furth
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Federico Valdivieso
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Amanda Lemke
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318
| | - Benjamin J. Wilkins
- Department of Pathology, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A., 19104-4318
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104-4318
| | - Edward Doolin
- Pediatric General, Thoracic and Fetal Surgery, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A. 19104-4318
| | - Marthe J. Howard
- Department of Neurosciences, University of Toledo, Mail Stop # 1007, 3000 Arlington Avenue, Toledo, OH, U.S.A, 43614-2598
| | - Robert O. Heuckeroth
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| |
Collapse
|
33
|
Persistent Increased Enteric Glial Expression of S100β is Associated With Low-grade Inflammation in Patients With Diverticular Disease. J Clin Gastroenterol 2019. [PMID: 29517710 DOI: 10.1097/mcg.0000000000001011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Diverticular disease (DD) is a common gastrointestinal inflammatory disorder associated with an enteric neuropathy. Although enteric glial cells (EGCs) are essential regulators of intestinal inflammation and motility functions, their contribution to the pathophysiology of DD remains unclear. Therefore, we analyzed the expression of specific EGC markers in patients with DD. MATERIALS AND METHODS Expression of the glial markers S100β, GFAP, Sox10, and Connexin 43 was analyzed by real-time quantitative PCR in colonic specimens of patients with DD and in that of controls. Protein expression levels of S100β, GFAP, and Connexin 43 were further analyzed using immunohistochemistry in the submucosal and myenteric plexus of patients with DD and in that of controls. Expression of the inflammatory cytokines tumor necrosis factor-α and interleukin-6 was quantified using qPCR, and infiltration of CD3+ lymphocytes was determined using immunohistochemistry. RESULTS Expression of S100β was increased in the submucosal and myenteric plexus of patients with DD compared with that in controls, whereas expression of other glial factors remained unchanged. This increased expression of S100β was correlated to CD3+ lymphocytic infiltrates in patients with DD, whereas no correlation was observed in controls. CONCLUSIONS DD is associated with limited but significant alterations of the enteric glial network. The increased expression of S100β is associated with a persistent low-grade inflammation reported in patients with DD, further emphasizing the role of EGCs in intestinal inflammation.
Collapse
|
34
|
Contractile dysfunction and nitrergic dysregulation in small intestine of a primate model of Parkinson's disease. NPJ PARKINSONS DISEASE 2019; 5:10. [PMID: 31231674 PMCID: PMC6557815 DOI: 10.1038/s41531-019-0081-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
Bowel dysfunction is a common non-motor symptom in Parkinson’s disease (PD). The main contractile neurotransmitter in the GI tract is acetylcholine (ACh), while nitric oxide (NO) causes the relaxation of smooth muscle in addition to modulating ACh release. The aim of this study was to characterise functional and neurochemical changes in the isolated ileum of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated marmoset, an established model of PD motor dysfunction. While NO-synthase inhibitor L-NAME concentration dependently augmented the neurogenically-evoked contractions and inhibited the relaxations in normal tissues, it had no effects on the MPTP ileum. Immunohistochemical analyses of the myenteric plexus showed that ChAT-immunoreactivity (-ir) was significantly reduced and the density of the enteric glial cells as shown by SOX-10-ir was increased. However, no change in TH-, 5-HT-, VIP- or nNOS-ir was observed in the MPTP tissues. The enhancement of the neurogenically-evoked contractions and the inhibition of the relaxation phase by L-NAME in the control tissues is in line with NO’s direct relaxing effect on smooth muscle and its indirect inhibitory effect on ACh release. The absence of the relaxation and the inefficacy of L-NAME in the MPTP tissues suggests that central dopaminergic loss dopamine may eventually lead to the impairment of NO signal coupling that affects bowel function, and this may be the result of a complex dysregulation at the level of the neuroeffector junction.
Collapse
|
35
|
Xu Y, Xie MZ, Liang GG. Advances in morphologic study of enteric glial cells. Shijie Huaren Xiaohua Zazhi 2019; 27:521-526. [DOI: 10.11569/wcjd.v27.i8.521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As an important part of the intestinal nervous system, enteric glial cells are about four times as many as intestinal neurons. Furthermore, a large population of astrocyte-like glial cells populate gut muscle layers and the intestinal mucosa, and mounting new evidence points toward enteric glial cells as an active participant in gut pathology. They are similar in morphology and function to the astrocytes of the central nervous system and play an important role in nutrition, supporting gastrointestinal nerve, maintaining gastrointestinal homeostasis, and regulating gastrointestinal function. Because of their complex and diverse roles in the intestinal tract, they have become the focus of research. As the study of their functional mechanism has been extensively deepened, the research methods for intestinal glial cells are also on constant progress and improvement, especially in studying their morphology. This paper mainly introduces the morphological characteristics of enteric glial cells under the conditions of gastrointestinal physiology and pathology, so as to provide a reference for the future study of enteric glial cells and promote the development of this field.
Collapse
Affiliation(s)
- Ying Xu
- Department of Emergency Abdominal Surgery/Institute of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Ming-Zheng Xie
- Department of Emergency Abdominal Surgery/Institute of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Guo-Gang Liang
- Department of Emergency Abdominal Surgery/Institute of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian 116000, Liaoning Province, China
| |
Collapse
|
36
|
Vergnolle N, Cirillo C. Neurons and Glia in the Enteric Nervous System and Epithelial Barrier Function. Physiology (Bethesda) 2019; 33:269-280. [PMID: 29897300 DOI: 10.1152/physiol.00009.2018] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The intestinal epithelial barrier is the largest exchange surface between the body and the external environment. Its functions are regulated by luminal, and also internal, components including the enteric nervous system. This review summarizes current knowledge about the role of the digestive "neuronal-glial-epithelial unit" on epithelial barrier function.
Collapse
Affiliation(s)
- Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse , France.,Department of Physiology and Pharmacology, Faculty of Medicine, University of Calgary , Calgary, Alberta , Canada
| | - Carla Cirillo
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse , France.,Laboratory for Enteric Neuroscience, TARGID, University of Leuven , Leuven , Belgium
| |
Collapse
|
37
|
Valès S, Touvron M, Van Landeghem L. Enteric glia: Diversity or plasticity? Brain Res 2018; 1693:140-145. [PMID: 29425908 DOI: 10.1016/j.brainres.2018.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/27/2018] [Accepted: 02/01/2018] [Indexed: 01/16/2023]
Abstract
Glial cells of the enteric nervous system correspond to a unique glial lineage distinct from other central and peripheral glia, and form a vast and abundant network spreading throughout all the layers of the gastrointestinal wall. Research over the last two decades has demonstrated that enteric glia regulates all major gastrointestinal functions via multiple bi-directional crosstalk with enteric neurons and other neighboring cell types. Recent studies propose that enteric glia represents a heterogeneous population associated with distinct localization within the gut wall, phenotype and activity. Compelling evidence also indicates that enteric glial cells are capable of plasticity leading to phenotypic changes whose pinnacle so far has been shown to be the generation of enteric neurons. While alterations of the glial network have been heavily incriminated in the development of gastrointestinal pathologies, enteric glial cells have also recently emerged as an active player in gut-brain signaling. Therefore, the development of tools and techniques to better appraise enteric glia heterogeneity and plasticity will undoubtedly unveil critical regulatory mechanisms implicated in gut health and disease, as well as disorders of the gut-brain axis.
Collapse
Affiliation(s)
- Simon Valès
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, 1 rue Gaston Veil, 44035 Nantes Cedex, France.
| | - Melissa Touvron
- Department of Molecular Biomedical Sciences, North Carolina State University, College of Veterinary Medicine, CVM Main Building, Campus Box #8401, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| | - Laurianne Van Landeghem
- Department of Molecular Biomedical Sciences, North Carolina State University, College of Veterinary Medicine, CVM Main Building, Campus Box #8401, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| |
Collapse
|
38
|
Pochard C, Coquenlorge S, Freyssinet M, Naveilhan P, Bourreille A, Neunlist M, Rolli-Derkinderen M. The multiple faces of inflammatory enteric glial cells: is Crohn's disease a gliopathy? Am J Physiol Gastrointest Liver Physiol 2018. [PMID: 29517926 DOI: 10.1152/ajpgi.00016.2018] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gone are the days when enteric glial cells (EGC) were considered merely satellites of enteric neurons. Like their brain counterpart astrocytes, EGC express an impressive number of receptors for neurotransmitters and intercellular messengers, thereby contributing to neuroprotection and to the regulation of neuronal activity. EGC also produce different soluble factors that regulate neighboring cells, among which are intestinal epithelial cells. A better understanding of EGC response to an inflammatory environment, often referred to as enteric glial reactivity, could help define the physiological role of EGC and the importance of this reactivity in maintaining gut functions. In chronic inflammatory disorders of the gut such as Crohn's disease (CD) and ulcerative colitis, EGC exhibit abnormal phenotypes, and their neighboring cells are dysfunctional; however, it remains unclear whether EGC are only passive bystanders or active players in the pathophysiology of both disorders. The aim of the present study is to review the physiological roles and properties of EGC, their response to inflammation, and their role in the regulation of the intestinal epithelial barrier and to discuss the emerging concept of CD as an enteric gliopathy.
Collapse
Affiliation(s)
- Camille Pochard
- Inserm, UMR1235 TENS, Nantes , France.,Nantes University , Nantes , France.,Institut des Maladies de l'Appareil Digestif, IMAD, Centre Hospitalier Universitaire de Nantes, Hôpital Hôtel-Dieu, Nantes , France
| | - Sabrina Coquenlorge
- Inserm, UMR1235 TENS, Nantes , France.,Nantes University , Nantes , France.,Institut des Maladies de l'Appareil Digestif, IMAD, Centre Hospitalier Universitaire de Nantes, Hôpital Hôtel-Dieu, Nantes , France
| | - Marie Freyssinet
- Inserm, UMR1235 TENS, Nantes , France.,Nantes University , Nantes , France.,Institut des Maladies de l'Appareil Digestif, IMAD, Centre Hospitalier Universitaire de Nantes, Hôpital Hôtel-Dieu, Nantes , France
| | - Philippe Naveilhan
- Inserm, UMR1235 TENS, Nantes , France.,Nantes University , Nantes , France.,Institut des Maladies de l'Appareil Digestif, IMAD, Centre Hospitalier Universitaire de Nantes, Hôpital Hôtel-Dieu, Nantes , France
| | - Arnaud Bourreille
- Inserm, UMR1235 TENS, Nantes , France.,Nantes University , Nantes , France.,Institut des Maladies de l'Appareil Digestif, IMAD, Centre Hospitalier Universitaire de Nantes, Hôpital Hôtel-Dieu, Nantes , France
| | - Michel Neunlist
- Inserm, UMR1235 TENS, Nantes , France.,Nantes University , Nantes , France.,Institut des Maladies de l'Appareil Digestif, IMAD, Centre Hospitalier Universitaire de Nantes, Hôpital Hôtel-Dieu, Nantes , France
| | - Malvyne Rolli-Derkinderen
- Inserm, UMR1235 TENS, Nantes , France.,Nantes University , Nantes , France.,Institut des Maladies de l'Appareil Digestif, IMAD, Centre Hospitalier Universitaire de Nantes, Hôpital Hôtel-Dieu, Nantes , France
| |
Collapse
|
39
|
Lilli NL, Quénéhervé L, Haddara S, Brochard C, Aubert P, Rolli-Derkinderen M, Durand T, Naveilhan P, Hardouin JB, De Giorgio R, Barbara G, Bruley des Varannes S, Coron E, Neunlist M. Glioplasticity in irritable bowel syndrome. Neurogastroenterol Motil 2018; 30:e13232. [PMID: 29027719 DOI: 10.1111/nmo.13232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/17/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Growing evidence indicates a wide array of cellular remodeling in the mucosal microenvironment during irritable bowel syndrome (IBS), which possibly contributes to pathophysiology and symptom generation. Here, we investigated whether enteric glial cells (EGC) may be altered, and which factors/mechanisms lead to these changes. METHODS Colonic mucosal biopsies of IBS patients (13 IBS-Constipation [IBS-C]; 10 IBS-Diarrhea [IBS-D]; 11 IBS-Mixed [IBS-M]) and 24 healthy controls (HC) were analyzed. Expression of S100β and GFAP was measured. Cultured rat EGC were incubated with supernatants from mucosal biopsies, then proliferation and Ca2+ response to ATP were analyzed using flow cytometry and Ca2+ imaging. Histamine and histamine 1-receptor (H1R) involvement in the effects of supernatant upon EGC was analyzed. KEY RESULTS Compared to HC, the mucosal area immunoreactive for S100β was significantly reduced in biopsies of IBS patients, independently of the IBS subtype. IBS-C supernatants reduced EGC proliferation and IBS-D and IBS-M supernatants reduced Ca2+ response to ATP in EGC. EGC expressed H1R and the effects of supernatant upon Ca2+ response to ATP in EGC were blocked by pyrilamine and reproduced by histamine via H1R. IBS supernatants reduced mRNA expression of connexin-43. The S100β-stained area was negatively correlated with the frequency and intensity of pain and bloating. CONCLUSION AND INFERENCES Changes in EGC occur in IBS, involving mucosal soluble factors. Histamine, via activation of H1R-dependent pathways, partly mediates altered Ca2+ response to ATP in EGC. These changes may contribute to the pathophysiology and the perception of pain and bloating in patients with IBS.
Collapse
Affiliation(s)
- N L Lilli
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - L Quénéhervé
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - S Haddara
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - C Brochard
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France
| | - P Aubert
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - M Rolli-Derkinderen
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - T Durand
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - P Naveilhan
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - J-B Hardouin
- Université de Nantes, INSERM, SPHERE, Université Bretagne Loire, Nantes, France
| | - R De Giorgio
- Department of Medical and Surgical Sciences, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - G Barbara
- Department of Medical and Surgical Sciences, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - S Bruley des Varannes
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - E Coron
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France.,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - M Neunlist
- Université de Nantes, INSERM, IMAD, The enteric nervous system in gut and brain disorders, Université Bretagne Loire, Nantes, France
| |
Collapse
|
40
|
Grubišić V, Verkhratsky A, Zorec R, Parpura V. Enteric glia regulate gut motility in health and disease. Brain Res Bull 2018; 136:109-117. [PMID: 28363846 PMCID: PMC5620110 DOI: 10.1016/j.brainresbull.2017.03.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/20/2017] [Accepted: 03/28/2017] [Indexed: 12/16/2022]
Abstract
The enteric nervous system, often referred to as the second brain, is the largest assembly of neurons and glia outside the central nervous system. The enteric nervous system resides within the wall of the digestive tract and regulates local gut reflexes involved in gastrointestinal motility and fluid transport; these functions can be accomplished in the absence of the extrinsic innervation from the central nervous system. It is neurons and their circuitry within the enteric nervous system that govern the gut reflexes. However, it is becoming clear that enteric glial cells are also actively involved in this process through the bidirectional signaling with neurons and other cells in the gut wall. We synthesize the recently discovered modulatory roles of enteric gliotransmission in gut motility and provide our perspective for future lines of research.
Collapse
Affiliation(s)
- Vladimir Grubišić
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA; Neuroscience Program, Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824, USA
| | - Alexei Verkhratsky
- The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology University of Ljubljana, Ljubljana, Slovenia; Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
41
|
Kobayashi M, Sumiyama K, Shimojima N, Ieiri S, Okano H, Kamba S, Fujimura T, Hirobe S, Kuroda T, Takahashi-Fujigasaki J. Technical feasibility of visualizing myenteric plexus using confocal laser endomicroscopy. J Gastroenterol Hepatol 2017; 32:1604-1610. [PMID: 28142194 DOI: 10.1111/jgh.13754] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/29/2016] [Accepted: 01/27/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIM In preceding studies, we identified that the myenteric plexus (MP) could be visualized with confocal laser endomicroscopy (CLE) by applying neural fluorescent probes lacking clinical safety profiling data from the submucosal side. In this study, we evaluated the technical feasibility of MP visualization using probe-based CLE (pCLE) from the serosal side with cresyl violet (CV), which has been used clinically for chromoendoscopy. METHODS The dye affinity of CV for MP was first explored in an in vivo transgenic mouse model using neural crest derivatives labeled with green fluorescent protein. We also tested the feasibility of CV-assisted visualization of MP in human surgical specimens, wherein the tissue dying and pCLE observation were performed from the serosal side. In the human study, rate of MP visualization by pCLE was evaluated as the primary outcome. We also evaluated the sensitivity and specificity of MP visualization by pCLE, using pathological presence/absence of MP as the gold standard. RESULTS We confirmed the dye affinity of CV to MP in all tested models. The MP appeared as brightly stained ladder-like structures with pCLE, and in the human study, MP was visualized in 12/14 (85.7%) samples, with 92.3% sensitivity and 100% specificity. In positive cases showing the ladder-like structure of MP by pCLE, the mean maximum and minimum widths of nerve strands were 54.3 (± 23.6) and 19.7 (± 6.0) μm, respectively. A ganglion was detected by pCLE in 10 cases (10/12, 83.3%). CONCLUSIONS This study demonstrated the technical feasibility of visualizing the MP in real time by CV-assisted pCLE (UMIN-CTR number, UMIN000015056).
Collapse
Affiliation(s)
- Masakuni Kobayashi
- Department of Endoscopy and Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuki Sumiyama
- Department of Endoscopy, The Jikei University School of Medicine, Tokyo, Japan
| | - Naoki Shimojima
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Surgery, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Kagoshima University, Kagoshima, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Shunsuke Kamba
- Department of Endoscopy and Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takumi Fujimura
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Seiichi Hirobe
- Department of Surgery, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Tatsuo Kuroda
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Junko Takahashi-Fujigasaki
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
42
|
Hao MM, Capoccia E, Cirillo C, Boesmans W, Vanden Berghe P. Arundic Acid Prevents Developmental Upregulation of S100B Expression and Inhibits Enteric Glial Development. Front Cell Neurosci 2017; 11:42. [PMID: 28280459 PMCID: PMC5322270 DOI: 10.3389/fncel.2017.00042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/08/2017] [Indexed: 12/31/2022] Open
Abstract
S100B is expressed in various types of glial cells and is involved in regulating many aspects of their function. However, little is known about its role during nervous system development. In this study, we investigated the effect of inhibiting the onset of S100B synthesis in the development of the enteric nervous system, a network of neurons and glia located in the wall of the gut that is vital for control of gastrointestinal function. Intact gut explants were taken from embryonic day (E)13.5 mice, the day before the first immunohistochemical detection of S100B, and cultured in the presence of arundic acid, an inhibitor of S100B synthesis, for 48 h. The effects on Sox10-immunoreactive enteric neural crest progenitors and Hu-immunoreactive enteric neurons were then analyzed. Culture in arundic acid reduced the proportion of Sox10+ cells and decreased cell proliferation. There was no change in the density of Hu+ enteric neurons, however, a small population of cells exhibited atypical co-expression of both Sox10 and Hu, which was not observed in control cultures. Addition of exogenous S100B to the cultures did not change Sox10+ cell numbers. Overall, our data suggest that cell-intrinsic intracellular S100B is important for maintaining Sox10 and proliferation of the developing enteric glial lineage.
Collapse
Affiliation(s)
- Marlene M Hao
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| | - Elena Capoccia
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU LeuvenLeuven, Belgium; Department of Physiology and Pharmacology, Sapienza University of RomeRome, Italy
| | - Carla Cirillo
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| | - Werend Boesmans
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| |
Collapse
|
43
|
Grubišić V, Gulbransen BD. Enteric glia: the most alimentary of all glia. J Physiol 2017; 595:557-570. [PMID: 27106597 PMCID: PMC5233670 DOI: 10.1113/jp271021] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
Glia (from Greek γλοία meaning 'glue') pertains to non-neuronal cells in the central (CNS) and peripheral nervous system (PNS) that nourish neurons and maintain homeostasis. In addition, glia are now increasingly appreciated as active regulators of numerous physiological processes initially considered exclusively under neuronal regulation. For instance, enteric glia, a collection of glial cells residing within the walls of the intestinal tract, regulate intestinal motility, a well-characterized reflex controlled by enteric neurons. Enteric glia also interact with various non-neuronal cell types in the gut wall such as enterocytes, enteroendocrine and immune cells and are therefore emerging as important local regulators of diverse gut functions. The intricate molecular mechanisms that govern glia-mediated regulation are beginning to be discovered, but much remains unknown about the functions of enteric glia in health and disease. Here we present a current view of the enteric glia and their regulatory roles in gastrointestinal (GI) (patho)physiology; from GI motility and epithelial barrier function to enteric neuroinflammation.
Collapse
Affiliation(s)
- Vladimir Grubišić
- Neuroscience Program, Department of PhysiologyMichigan State University567 Wilson RoadEast LansingMI48824USA
| | - Brian D. Gulbransen
- Neuroscience Program, Department of PhysiologyMichigan State University567 Wilson RoadEast LansingMI48824USA
| |
Collapse
|
44
|
da Cunha Franceschi R, Nardin P, Machado CV, Tortorelli LS, Martinez-Pereira MA, Zanotto C, Gonçalves CA, Zancan DM. Enteric glial reactivity to systemic LPS administration: Changes in GFAP and S100B protein. Neurosci Res 2017; 119:15-23. [PMID: 28063977 DOI: 10.1016/j.neures.2016.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/29/2016] [Accepted: 12/27/2016] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharide (LPS) is used to induce inflammation and promotes nervous system activation. Different regions of the brain present heterogeneous glial responses; thus, in order to verify whether systemic LPS-induced inflammation affects the enteric glia differently across the intestinal segments, we evaluated the expressions of two glial activity markers, GFAP and S100B protein, in different intestine segments, at 1h, 24h and 7days after acute systemic LPS administration (0.25 or 2.5mgkg-1) in rats. Histological inflammatory analysis indicated that the cecum was most affected when compared to the duodenum and proximal colon at the highest doses of LPS. LPS induced an increased S100B content after 24h in all three regions, which decreased at 7days after the highest dose in all regions. Moreover, at 24h, this dose of LPS increased ex-vivo S100B secretion only in the cecum. The highest dose of LPS also increased GFAP in all regions at 24h, but earlier in the cecum, where LPS-induced enteric S100B and GFAP alterations were dependent on dose, time and intestine region. No associated changes in serum S100B were observed. Our results indicate heterogeneous enteric glial responses to inflammatory insult, as observed in distinct brain areas.
Collapse
Affiliation(s)
- Raphaela da Cunha Franceschi
- Laboratory of Comparative Neurobiology, Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Graduate Program in Neuroscience, ICBS, UFRGS, Brazil
| | - Patrícia Nardin
- Laboratory of Calcium-Binding Proteins, Department of Biochemistry, ICBS, UFRGS, Brazil
| | - Clivia Valle Machado
- Laboratory of Comparative Neurobiology, Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Graduate Program in Neuroscience, ICBS, UFRGS, Brazil
| | | | | | - Caroline Zanotto
- Laboratory of Calcium-Binding Proteins, Department of Biochemistry, ICBS, UFRGS, Brazil
| | - Carlos-Alberto Gonçalves
- Graduate Program in Neuroscience, ICBS, UFRGS, Brazil; Laboratory of Calcium-Binding Proteins, Department of Biochemistry, ICBS, UFRGS, Brazil.
| | - Denise Maria Zancan
- Laboratory of Comparative Neurobiology, Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Graduate Program in Neuroscience, ICBS, UFRGS, Brazil
| |
Collapse
|
45
|
Xiao WD, Peng K, Yang H. Enteric glial cells: An emerging key player in intestinal homeostasis modulation under physiological and pathological conditions. Shijie Huaren Xiaohua Zazhi 2016; 24:3657-3665. [DOI: 10.11569/wcjd.v24.i25.3657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The intestine contains multiple components including epithelial cells, microbiome as well as various neuroendocrine pathways, all of which are essential for maintaining dynamic mucosal homeostasis through complex interactions among different components in the gastrointestinal tract. Beyond the basic neurosupportive and neurotrophic effects, growing evidence reveals the key role of enteric glial cells (EGCs) in the modulation of bowel movement, nutrient absorption and secretion, intestinal immunity as well as barrier function. As well, abnormally activated EGCs are believed to be a vital player in the pathogenesis of a variety of diseases including inflammatory bowel disease, intestinal barrier dysfunction and infections. Here we provide a brief overview of recent research progress about the precise role and the molecule mechanisms of EGCs in modulating intestinal homeostasis, and highlight the critical role of EGC in various intestinal diseases.
Collapse
|
46
|
Molecular Signaling and Dysfunction of the Human Reactive Enteric Glial Cell Phenotype: Implications for GI Infection, IBD, POI, Neurological, Motility, and GI Disorders. Inflamm Bowel Dis 2016; 22:1812-34. [PMID: 27416040 PMCID: PMC4993196 DOI: 10.1097/mib.0000000000000854] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Clinical observations or animal studies implicate enteric glial cells in motility disorders, irritable bowel syndrome, inflammatory bowel disease, gastrointestinal (GI) infections, postoperative ileus, and slow transit constipation. Mechanisms underlying glial responses to inflammation in human GI tract are not understood. Our goal was to identify the "reactive human enteric glial cell (rhEGC) phenotype" induced by inflammation, and probe its functional relevance. METHODS Human enteric glial cells in culture from 15 GI-surgical specimens were used to study gene expression, Ca, and purinergic signaling by Ca/fluo-4 imaging and mechanosensitivity. A nanostring panel of 107 genes was designed as a read out of inflammation, transcription, purinergic signaling, vesicular transport protein, channel, antioxidant, and other pathways. A 24-hour treatment with lipopolysaccharide (200 μg/mL) and interferon-γ (10 μg/mL) was used to induce inflammation and study molecular signaling, flow-dependent Ca responses from 3 mL/min to 10 mL/min, adenosine triphosphate (ATP) release, and ATP responses. RESULTS Treatment induced a "rhEGC phenotype" and caused up-regulation in messenger RNA transcripts of 58% of 107 genes analyzed. Regulated genes included inflammatory genes (54%/IP10; IFN-γ; CxCl2; CCL3; CCL2; C3; s100B; IL-1β; IL-2R; TNF-α; IL-4; IL-6; IL-8; IL-10; IL-12A; IL-17A; IL-22; and IL-33), purine-genes (52%/AdoR2A; AdoR2B; P2RY1; P2RY2; P2RY6; P2RX3; P2RX7; AMPD3; ENTPD2; ENTPD3; and NADSYN1), channels (40%/Panx1; CHRNA7; TRPV1; and TRPA1), vesicular transporters (SYT1, SYT2, SNAP25, and SYP), transcription factors (relA/relB, SOCS3, STAT3, GATA_3, and FOXP3), growth factors (IGFBP5 and GMCSF), antioxidant genes (SOD2 and HMOX1), and enzymes (NOS2; TPH2; and CASP3) (P < 0.0001). Treatment disrupted Ca signaling, ATP, and mechanical/flow-dependent Ca responses in human enteric glial cells. ATP release increased 5-fold and s100B decreased 33%. CONCLUSIONS The "rhEGC phenotype" is identified by a complex cascade of pro-inflammatory pathways leading to alterations of important molecular and functional signaling pathways (Ca, purinergic, and mechanosensory) that could disrupt GI motility. Inflammation induced a "purinergic switch" from ATP to adenosine diphosphate/adenosine/uridine triphosphate signaling. Findings have implications for GI infection, inflammatory bowel disease, postoperative ileus, motility, and GI disorders.
Collapse
|
47
|
M Hao M. Development of Neural Activity in the Enteric Nervous System: Similarities and Differences to Other Parts of the Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 891:43-51. [PMID: 27379633 DOI: 10.1007/978-3-319-27592-5_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
All the neurons and glia of the enteric nervous system (ENS) arise from neural crest-derived cells that migrate into the gastrointestinal (GI) tract during development (Yntema and Hammond 1954; Le Douarin and Teillet 1973). Most of the ENS originates from vagal neural crest cells (NCCs), which arise from the caudal hindbrain region of the neural tube, adjacent to somites 1-7. In the developing mouse, vagal NCCs migrate into the developing oesophagus and stomach at embryonic day (E)9.5, enter the small intestine at E10.5, and colonise the developing GI tract in a rostral-to-caudal wave, reaching the anal end of the colon at E14.5 (Serbedzija et al. 1991; Kapur et al. 1992; Anderson et al. 2006). Recent evidence indicates that there is also trans-mesenteric migration of vagal NCCs, where some NCCs leave the small intestine and migrate directly across the mesentery into the colon (Nishiyama et al. 2012). Sacral NCCs also contribute to a small population of neurons and glia in the colon (Burns and Le Douarin 1998; Wang et al. 2011).
Collapse
Affiliation(s)
- Marlene M Hao
- Laboratory for Enteric Neuroscience, TARGID, University of Leuven, Herestraat 49, O&N1, Box 701, Leuven, 3000, Belgium.
| |
Collapse
|
48
|
Cossais F, Durand T, Chevalier J, Boudaud M, Kermarrec L, Aubert P, Neveu I, Naveilhan P, Neunlist M. Postnatal development of the myenteric glial network and its modulation by butyrate. Am J Physiol Gastrointest Liver Physiol 2016; 310:G941-51. [PMID: 27056724 DOI: 10.1152/ajpgi.00232.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 03/16/2016] [Indexed: 02/08/2023]
Abstract
The postnatal period is crucial for the development of gastrointestinal (GI) functions. The enteric nervous system is a key regulator of GI functions, and increasing evidences indicate that 1) postnatal maturation of enteric neurons affect the development of GI functions, and 2) microbiota-derived short-chain fatty acids can be involved in this maturation. Although enteric glial cells (EGC) are central regulators of GI functions, the postnatal evolution of their phenotype remains poorly defined. We thus characterized the postnatal evolution of EGC phenotype in the colon of rat pups and studied the effect of short-chain fatty acids on their maturation. We showed an increased expression of the glial markers GFAP and S100β during the first postnatal week. As demonstrated by immunohistochemistry, a structured myenteric glial network was observed at 36 days in the rat colons. Butyrate inhibited EGC proliferation in vivo and in vitro but had no effect on glial marker expression. These results indicate that the EGC myenteric network continues to develop after birth, and luminal factors such as butyrate endogenously produced in the colon may affect this development.
Collapse
Affiliation(s)
- François Cossais
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Tony Durand
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Julien Chevalier
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Marie Boudaud
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Laetitia Kermarrec
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Philippe Aubert
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Isabelle Neveu
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Philippe Naveilhan
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| | - Michel Neunlist
- INSERM, U913, Nantes, France; Université Nantes, Nantes, France; CHU Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, France; and Centre de Recherche en Nutrition Humaine, Nantes, France
| |
Collapse
|
49
|
Coquenlorge S, Van Landeghem L, Jaulin J, Cenac N, Vergnolle N, Duchalais E, Neunlist M, Rolli-Derkinderen M. The arachidonic acid metabolite 11β-ProstaglandinF2α controls intestinal epithelial healing: deficiency in patients with Crohn's disease. Sci Rep 2016; 6:25203. [PMID: 27140063 PMCID: PMC4853710 DOI: 10.1038/srep25203] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Abstract
In healthy gut enteric glial cells (EGC) are essential to intestinal epithelial barrier (IEB) functions. In Crohn's Disease (CD), both EGC phenotype and IEB functions are altered, but putative involvement of EGC in CD pathogenesis remains unknown and study of human EGC are lacking. EGC isolated from CD and control patients showed similar expression of glial markers and EGC-derived soluble factors (IL6, TGF-β, proEGF, GSH) but CD EGC failed to increase IEB resistance and healing. Lipid profiling showed that CD EGC produced decreased amounts of 15-HETE, 18-HEPE, 15dPGJ2 and 11βPGF2α as compared to healthy EGC. They also had reduced expression of the L-PGDS and AKR1C3 enzymes. Produced by healthy EGC, the 11βPGF2 activated PPARγ receptor of intestinal epithelial cells to induce cell spreading and IEB wound repair. In addition to this novel healing mechanism our data show that CD EGC presented impaired ability to promote IEB functions through defect in L-PGDS-AKR1C3-11βPGF2α dependent pathway.
Collapse
Affiliation(s)
- Sabrina Coquenlorge
- INSERM, UMR913, Nantes, F-44093, France
- Université Nantes, Nantes, F-44093, France
- Institut des Maladies de l’Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, F-44093, France
- Centre de Recherche en Nutrition Humaine, Nantes, F-44093, France
| | - Laurianne Van Landeghem
- INSERM, UMR913, Nantes, F-44093, France
- Université Nantes, Nantes, F-44093, France
- Institut des Maladies de l’Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, F-44093, France
- Centre de Recherche en Nutrition Humaine, Nantes, F-44093, France
| | - Julie Jaulin
- INSERM, UMR913, Nantes, F-44093, France
- Université Nantes, Nantes, F-44093, France
- Institut des Maladies de l’Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, F-44093, France
- Centre de Recherche en Nutrition Humaine, Nantes, F-44093, France
| | - Nicolas Cenac
- Centre de Pathophysiologie, CHU Purpan, Toulouse, France
- INSERM UMR-1043 CNRS UMR-5282, Toulouse, France
| | - Nathalie Vergnolle
- Centre de Pathophysiologie, CHU Purpan, Toulouse, France
- INSERM UMR-1043 CNRS UMR-5282, Toulouse, France
| | - Emilie Duchalais
- INSERM, UMR913, Nantes, F-44093, France
- Université Nantes, Nantes, F-44093, France
- Institut des Maladies de l’Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, F-44093, France
- Centre de Recherche en Nutrition Humaine, Nantes, F-44093, France
| | - Michel Neunlist
- INSERM, UMR913, Nantes, F-44093, France
- Université Nantes, Nantes, F-44093, France
- Institut des Maladies de l’Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, F-44093, France
- Centre de Recherche en Nutrition Humaine, Nantes, F-44093, France
| | - Malvyne Rolli-Derkinderen
- INSERM, UMR913, Nantes, F-44093, France
- Université Nantes, Nantes, F-44093, France
- Institut des Maladies de l’Appareil Digestif, CHU Nantes, Hôpital Hôtel-Dieu, Nantes, F-44093, France
- Centre de Recherche en Nutrition Humaine, Nantes, F-44093, France
| |
Collapse
|
50
|
Uesaka T, Young HM, Pachnis V, Enomoto H. Development of the intrinsic and extrinsic innervation of the gut. Dev Biol 2016; 417:158-67. [PMID: 27112528 DOI: 10.1016/j.ydbio.2016.04.016] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/09/2016] [Accepted: 04/21/2016] [Indexed: 12/16/2022]
Abstract
The gastrointestinal (GI) tract is innervated by intrinsic enteric neurons and by extrinsic efferent and afferent nerves. The enteric (intrinsic) nervous system (ENS) in most regions of the gut consists of two main ganglionated layers; myenteric and submucosal ganglia, containing numerous types of enteric neurons and glial cells. Axons arising from the ENS and from extrinsic neurons innervate most layers of the gut wall and regulate many gut functions. The majority of ENS cells are derived from vagal neural crest cells (NCCs), which proliferate, colonize the entire gut, and first populate the myenteric region. After gut colonization by vagal NCCs, the extrinsic nerve fibers reach the GI tract, and Schwann cell precursors (SCPs) enter the gut along the extrinsic nerves. Furthermore, a subpopulation of cells in myenteric ganglia undergoes a radial (inward) migration to form the submucosal plexus, and the intrinsic and extrinsic innervation to the mucosal region develops. Here, we focus on recent progress in understanding the developmental processes that occur after the gut is colonized by vagal ENS precursors, and provide an up-to-date overview of molecular mechanisms regulating the development of the intrinsic and extrinsic innervation of the GI tract.
Collapse
Affiliation(s)
- Toshihiro Uesaka
- Division of Neural Differentiation and Regeneration, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan.
| | - Heather M Young
- Department of Anatomy and Neuroscience, University of Melbourne, 3010 VIC, Australia
| | - Vassilis Pachnis
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom
| | - Hideki Enomoto
- Division of Neural Differentiation and Regeneration, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan; Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| |
Collapse
|