51
|
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: 9.5] [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
|
52
|
Abstract
Many studies highlighted that a bidirectional communication between the gut and the central nervous system (CNS) exists. A vigorous immune response to antigens must be avoided, and pathogenic organisms crossing the gut barrier must be detected and killed. For this reason, the immune system developed fine mechanisms able to maintain this delicate balance. The microbiota is beneficial to its host, providing protection against pathogenic bacteria. It is intimately involved in numerous aspects of host physiology, from nutritional status to behavior and stress response. In the last few years, the implication of the gut microbiota and its bioactive microbiota-derived molecules in the progression of multiple diseases, as well as in the development of neurodegenerative disorders, gained increasing attention. The purpose of this review is to provide an overview of the gut microbiota with particular attention toward neurological disorders and mast cells. Relevant roles are played by the mast cells in neuroimmune communication, such as sensors and effectors of cytokines and neurotransmitters. In this context, the intake of beneficial bacterial strains as probiotics could represent a valuable therapeutic approach to adopt in combination with classical therapies. Further studies need to be performed to understand if the gut bacteria are responsible for neurological disorders or if neurological disorders influence the bacterial profile.
Collapse
|
53
|
Parathan P, Wang Y, Leembruggen AJL, Bornstein JC, Foong JPP. The enteric nervous system undergoes significant chemical and synaptic maturation during adolescence in mice. Dev Biol 2020; 458:75-87. [DOI: 10.1016/j.ydbio.2019.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022]
|
54
|
Tsuruta S, Uchida H, Akutsu H. Intestinal Organoids Generated from Human Pluripotent Stem Cells. JMA J 2020; 3:9-19. [PMID: 33324771 PMCID: PMC7733741 DOI: 10.31662/jmaj.2019-0027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal system is one of the most complex organ systems in the human body, and consists of numerous cell types originating from three germ layers. To understand intestinal development and homeostasis and elucidate the pathogenesis of intestinal disorders, including unidentified diseases, several in vitro models have been developed. Human pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have remarkable developmental plasticity and possess the potential for a wide variety of applications. Three-dimensional organs, termed organoids and produced in vitro by PSCs, contain not only epithelium but also mesenchymal tissue and partially recapitulate intestinal functions. Such intestinal organoids have begun to be applied in disease models and drug development and have contributed to a detailed analysis of molecular interactions and findings in the synergistic development of biomedicine for human digestive organs. In this review, we describe gastrointestinal organoid technology derived from PSCs and consider its potential applications.
Collapse
Affiliation(s)
- Satoru Tsuruta
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
- Centre for Regenerative Medicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hajime Uchida
- Transplantation Centre, National Centre for Child Health and Development, Tokyo, Japan
| | - Hidenori Akutsu
- Centre for Regenerative Medicine, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
55
|
Liu YR, Ba F, Cheng LJ, Li X, Zhang SW, Zhang SC. Efficacy of Sox10 Promoter Methylation in the Diagnosis of Intestinal Neuronal Dysplasia From the Peripheral Blood. Clin Transl Gastroenterol 2019; 10:e00093. [PMID: 31789936 PMCID: PMC6970557 DOI: 10.14309/ctg.0000000000000093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/19/2019] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Intestinal neuronal dysplasia (IND) is a common malformation of the enteric nervous system. Diagnosis requires a full-thickness colonic specimen and an experienced pathologist, emphasizing the need for noninvasive analytical methods. Recently, the methylation level of the Sox10 promoter has been found to be critical for enteric nervous system development. However, whether it can be used for diagnostic purposes in IND is unclear. METHODS Blood and colon specimens were collected from 32 patients with IND, 60 patients with Hirschsprung disease (HD), and 60 controls. Sox10 promoter methylation in the blood and the Sox10 expression level in the colon were determined, and their correlation was analyzed. The diagnostic efficacy of blood Sox10 promoter methylation was analyzed by receiver operating characteristic curve. RESULTS The blood level of Sox10 promoter methylation at the 32nd locus was 100% (90%-100%; 95% confidence interval [CI], 92.29%-96.37%) in control, 90% (80%-90%; 95% CI, 82.84%-87.83%) in HD, and 60% (50%-80%; 95% CI, 57.12%-69.76%) in IND specimens. Sox10 promoter methylation in the peripheral blood was negatively correlated with Sox10 expression in the colon, which was low in control, moderate in HD, and high in IND specimens (r = -0.89). The area under the curve of Sox10 promoter methylation in the diagnosis of IND was 0.94 (95% CI, 0.874-1.000, P = 0.000), with a cutoff value of 85% (sensitivity, 90.6%; specificity, 95.0%). By applying a cutoff value of 65%, promoter methylation was more indicative of IND than HD. DISCUSSION The analysis of Sox10 promoter methylation in the peripheral blood can be used as a noninvasive method for IND diagnosis.
Collapse
Affiliation(s)
- Yu-Rong Liu
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang Ba
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lan-Jie Cheng
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xu Li
- Department of Pediatric Surgery, Capital Institute of Pediatrics of Capital Medical University, Beijing, China
| | - Shi-Wei Zhang
- Department of Pediatric Surgery, Harbin Children's Hospital, Harbin, China
| | - Shu-Cheng Zhang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
56
|
Schmitteckert S, Mederer T, Röth R, Günther P, Holland-Cunz S, Metzger M, Samstag Y, Schröder-Braunstein J, Wabnitz G, Kurzhals S, Scheuerer J, Beretta CA, Lasitschka F, Rappold GA, Romero P, Niesler B. Postnatal human enteric neurospheres show a remarkable molecular complexity. Neurogastroenterol Motil 2019; 31:e13674. [PMID: 31318473 DOI: 10.1111/nmo.13674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/26/2019] [Accepted: 06/26/2019] [Indexed: 01/30/2023]
Abstract
BACKGROUND The enteric nervous system (ENS), a complex network of neurons and glial cells, coordinates major gastrointestinal functions. Impaired development or secondary aberrations cause severe enteric neuropathies. Neural crest-derived stem cells as well as enteric neuronal progenitor cells, which form enteric neurospheres, represent a promising tool to unravel molecular pathomechanisms and to develop novel therapy options. However, so far little is known about the detailed cellular composition and the proportional distribution of enteric neurospheres. Comprehensive knowledge will not only be essential for basic research but also for prospective cell replacement therapies to restore or to improve enteric neuronal dysfunction. METHODS Human enteric neurospheres were generated from three individuals with varying age. For detailed molecular characterization, nCounter target gene expression analyses focusing on stem, progenitor, neuronal, glial, muscular, and epithelial cell markers were performed. Corresponding archived paraffin-embedded individuals' specimens were analyzed accordingly. KEY RESULTS Our data revealed a remarkable molecular complexity of enteric neurospheres and archived specimens. Amongst the expression of multipotent stem cell, progenitor cell, neuronal, glial, muscle and epithelial cell markers, moderate levels for the pluripotency marker POU5F1 were observed. Furthermore, besides the interindividual variability, we identified highly distinct intraindividual expression profiles. CONCLUSIONS & INFERENCES Our results emphasize the assessment of molecular signatures to be essential for standardized use, optimization of experimental approaches, and elimination of potential risk factors, as the formation of tumors. Our study pipeline may serve as a blueprint implemented into the characterization procedure of enteric neurospheres for various future applications.
Collapse
Affiliation(s)
- Stefanie Schmitteckert
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Tanja Mederer
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ralph Röth
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,nCounter Core Facility, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Günther
- Division of Pediatric Surgery, Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Holland-Cunz
- Division of Pediatric Surgery, Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Pediatric Surgery, University Children's Hospital Basel, Basel, Switzerland
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research (ISC), Translational Centre Regenerative Therapies (TLC-RT) Wuerzburg, Wuerzburg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Guido Wabnitz
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Kurzhals
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jutta Scheuerer
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carlo A Beretta
- CellNetworks Math-Clinic Core Facility, Bioquant, Heidelberg University, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Philipp Romero
- Division of Pediatric Surgery, Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Niesler
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,nCounter Core Facility, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
57
|
Kozłowska A, Kozera P, Majewski M, Godlewski J. Co-expression of caspase-3 or caspase-8 with galanin in the human stomach section affected by carcinoma. Apoptosis 2019; 23:484-491. [PMID: 30019295 PMCID: PMC6153556 DOI: 10.1007/s10495-018-1470-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Neoplastic process may cause distinct changes in the morphology, i.e. size and number of the neurons of the neuronal plexuses forming the enteric nervous system (ENS) of the human intestine. Moreover, it was also reported that these changes were not directly associated with apoptosis. Thus, the main aim of this study was to determine the atrophic changes of myenteric plexuses (MPs) in the vicinity of cancer invasion and the potential reason which may be responsible for these changes if they occur. Tissue samples from the stomach were collected from ten patients which undergo organ resection due to cancer diagnosis. Samples were taken from the margin of cancer invasion and from a macroscopically-unchanged part of the stomach wall. Triple-immunofluorescence staining of the 10-µm-thick cryostat sections was used to visualize the co-expression of caspase-3 (CASP3) or caspase-8 (CASP8) with galanin (GAL) in the MPs of ENS. Microscopic observations of MPs located closely to gastric cancer invasion showed that they were significantly smaller than plexuses located distally. The percentage of neurons containing CASP3 within MPs located close to cancer-affected regions of the stomach was higher, while containing CASP8 was lower compared to the unchanged regions. Additionally, elevated high expression of CASP3 or CASP8 in the neurons from MPs was accompanied by a decreased expression of GAL. To our knowledge, this is the first report describing the decomposition of MPs within cancer-affected human stomach wall and the possible role of apoptosis in this process.
Collapse
Affiliation(s)
- Anna Kozłowska
- Department of Human Physiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Av 30, 10-082, Olsztyn, Poland.
| | - Piotr Kozera
- Department of Human Physiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Av 30, 10-082, Olsztyn, Poland
| | - Mariusz Majewski
- Department of Human Physiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Av 30, 10-082, Olsztyn, Poland
| | - Janusz Godlewski
- Department of Human Histology and Embryology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Av 30, 10-082, Olsztyn, Poland
| |
Collapse
|
58
|
Veríssimo CP, Carvalho JDS, da Silva FJM, Campanati L, Moura-Neto V, Coelho-Aguiar JDM. Laminin and Environmental Cues Act in the Inhibition of the Neuronal Differentiation of Enteric Glia in vitro. Front Neurosci 2019; 13:914. [PMID: 31551680 PMCID: PMC6733987 DOI: 10.3389/fnins.2019.00914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/16/2019] [Indexed: 12/31/2022] Open
Abstract
The enteric glia, a neural crest-derived cell type that composes the Enteric Nervous System, is involved in controlling gut functions, including motility, gut permeability, and neuronal communication. Moreover this glial cell could to give rise to new neurons. It is believed that enteric neurons are generated up to 21 days postnatally; however, adult gut cells with glial characteristics can give rise to new enteric neurons under certain conditions. The factors that activate this capability of enteric glia to differentiate into neurons remain unknown. Here, we followed the progress of this neuronal differentiation and investigated this ability by challenging enteric glial cells with different culture conditions. We found that, in vitro, enteric glial cells from the gut of adult and neonate mice have a high capability to acquire neuronal markers and undergoing morphological changes. In a co-culture system with 3T3 fibroblasts, the number of glial cells expressing βIIItubulin decreased after 7 days. The effect of 3T3-conditioned medium on adult cells was not significant, and fewer enteric glial cells from neonate mice began the neurogenic process in this medium. Laminin, an extracellular matrix protein that is highly expressed by the niche of the enteric ganglia, seemed to have a large role in inhibiting the differentiation of enteric glia, at least in cells from the adult gut. Our results suggest that, in an in vitro approach that provides conditions more similar to those of enteric glial cells in vivo, these cells could, to some extent, retain their morphology and marker expression, with their neurogenic potential inhibited. Importantly, laminin seemed to inhibit differentiation of adult enteric glial cells. It is possible that the differentiation of enteric glia into neurons is related to severe changes in the microenvironment, leading to disruption of the basement membrane. In summary, our data indicated that the interaction between the enteric glial cells and their microenvironment molecules significantly affects the control of their behavior and functions.
Collapse
Affiliation(s)
- Carla Pires Veríssimo
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Pós-graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana da Silva Carvalho
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Loraine Campanati
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana de Mattos Coelho-Aguiar
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
59
|
Jiang Y, Xu L, Yu L, Xu X, Feng C, Li J. NOX4 inhibition protects enteric glial cells against Clostridium difficile toxin B toxicity via attenuating oxidative and Endoplasmic reticulum stresses. Free Radic Res 2019; 53:932-940. [PMID: 31370714 DOI: 10.1080/10715762.2019.1649670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Enteric glial cells (EGCs), one main cell population of the enteric nervous system (ENS), play a major role in regulating intestinal barrier function. Clostridium difficile toxin B (TcdB) is the major virulence factor produced by C. difficile and estimated to be toxic to EGCs by inducing cell death, cell cycle arrest, and inflammatory cytokine production; however, the detailed mechanism for such effect is still unclear. In this study, we further evaluated the toxic effect of TcdB on EGCs and the involvement of NADPH oxidases in such process using the rat-transformed EGCs (CRL-2690). The results showed that NOX4 was activated by TcdB in EGCs and functioned as the major factor causing cytotoxicity and cell apoptosis. Mechanically, NOX4-generated H2O2 was the inducer of oxidative stress, Ca2+ homeostasis disorder, and ER stress in EGCs upon TcdB treatment, and NOX4 inhibition protected EGCs against TcdB toxicity via attenuating these dysfunctions. These findings contribute to our understanding of the mechanism by which TcdB affects EGCs and suggest the potential value of NOX4 inhibition for treatment against C. difficile infection.
Collapse
Affiliation(s)
- Yanmin Jiang
- Department of Endocrinology, Wuxi People's Hospital Affiliated to Nanjing Medical University , Wuxi , China
| | - Lan Xu
- Department of Endocrinology, Wuxi People's Hospital Affiliated to Nanjing Medical University , Wuxi , China
| | - Lin Yu
- Department of Endocrinology, Wuxi People's Hospital Affiliated to Nanjing Medical University , Wuxi , China
| | - Xiang Xu
- Department of Endocrinology, Wuxi People's Hospital Affiliated to Nanjing Medical University , Wuxi , China
| | - Chen Feng
- Department of Endocrinology, Wuxi People's Hospital Affiliated to Nanjing Medical University , Wuxi , China
| | - Jianbo Li
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| |
Collapse
|
60
|
Liang Y, Tarique I, Vistro WA, Liu Y, Wang Z, haseeb A, Gandahi NS, Iqbal A, Wang S, An T, Yang H, Chen Q, Yang P. Age-associated changes of the intrinsic nervous system in relation with interstitial cells in the pre-weaning goat rumen. Aging (Albany NY) 2019; 11:4641-4653. [PMID: 31305258 PMCID: PMC6660047 DOI: 10.18632/aging.102076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 07/01/2019] [Indexed: 05/04/2023]
Abstract
In this study, we investigated the neural changes and their relationships with interstitial cells (ICs) in the rumen of pre-weaning goats by transmission electron microscopy, western blot and immunofluorescence (antibody: general neuronal marker-Protein Gene Product (PGP9.5)/ IC marker-vimentin). The immunofluorescence results showed that PGP9.5-positive reaction was widely distributed in neuronal soma (NS) and nerve fibre (NF). The NSs were observed in the ganglia of the myenteric plexus (MP) but not in the submucosal plexus. The mean optical density (MOD) of the whole of PGP9.5-positive nerves and the protein expression level of PGP.5 in the rumen wall both decreased significantly with age. However an obvious increase MOD of PGP.5-positive NFs within the rumen epithelium were observed. In the MP, the nerves and ICs were interwoven to form two complex networks that gradually tightened with age. Furthermore, NSs and nerve trunks were surrounded by a ring-boundary layer consisting of several ICs that became physically closer with aging. Moreover, ICs were located nearby NFs within the ML, forming connections between ICs, smooth muscle cells and axons. This study describes the pattern of neural distribution and its association with ICs in the developing rumen which shed light on the postpartum development of ruminants.
Collapse
Affiliation(s)
- Yu Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Imran Tarique
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Waseem Ail Vistro
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Yifei Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Ziyu Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Abdul haseeb
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Noor Samad Gandahi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Adeela Iqbal
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Siyi Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Tianci An
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Huan Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Qiusheng Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Ping Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| |
Collapse
|
61
|
Fisher M, Smeiles C, Jnah AJ, Ruiz ME, Difiore T, Sewell K. Congenital Central Hypoventilation Syndrome: A Case-Based Learning Opportunity for Neonatal Clinicians. Neonatal Netw 2019; 38:217-225. [PMID: 31470390 DOI: 10.1891/0730-0832.38.4.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Congenital central hypoventilation syndrome (CCHS) is a rare and sporadic neurocristopathy characterized by alveolar hypoventilation and autonomic nervous system dysfunction. CCHS manifests quickly after birth, initially as respiratory distress. Mortality risk is estimated at 38 percent, with a median age of death of three months of age. A timely and accurate diagnosis is critical. Genetic testing for PHOX2B gene mutations is necessary to confirm the diagnosis; however, laboratory turnaround time often imposes an additional 7-14-day waiting period on an often anxious family. Neonatal clinicians should recognize that families require disease-specific education, emotional support, and time to rehearse daily caregiving in preparation for discharge. Therefore, this article presents the key clinical, pathophysiologic, and diagnostic factors, as well as a discussion of discharge needs. A case report of an infant, born to parents with no known history of CCHS, is included as a case-based learning opportunity for readers.
Collapse
|
62
|
Mahe MM. Engineering a second brain in a dish. Brain Res 2019; 1693:165-168. [PMID: 29903618 DOI: 10.1016/j.brainres.2018.04.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023]
Abstract
The utilization of human pluripotent stem cells holds great promise in elucidating principles of developmental biology and applications in personalized and regenerative medicine. Breakthroughs from the last decade have allowed the scientific community to better understand and successfully manipulate human pluripotent stem cells using distinct differentiation strategies into a variety of target tissues. This manipulation relies solely on our understanding of developmental processes occurring in model organisms. The in vitro translation of our developmental knowledge upon stem cells provides a new means to generate specific tissue to understand developmental and disease mechanisms, as well as physiological processes. The generation of an integrated human intestinal tissue is one such example. In this review, we highlight the biological motivation behind the generation of human intestinal organoids. We further describe the integration of an enteric nervous system within the organoid to generate a functional intestine. Forthcoming strategies to add additional complexities to the intestinal tissue so as to better understand how our "second brain" functions within the gut are also discussed. The organoid system offers a promising avenue to understand how the enteric nervous system works and patterns the human intestine during both physiology and disease.
Collapse
Affiliation(s)
- Maxime M Mahe
- Department of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, S6.609, Building, 3333 Burnet Avenue, Cincinnati, OH, USA; Inserm UMR 1235 - TENS, INSERM, University of Nantes, Rm427, Faculty of Medicine, 1 Rue Gaston Veil, Nantes, France.
| |
Collapse
|
63
|
Null mutation of the endothelin receptor type B gene causes embryonic death in the GK rat. PLoS One 2019; 14:e0217132. [PMID: 31170185 PMCID: PMC6553694 DOI: 10.1371/journal.pone.0217132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 05/06/2019] [Indexed: 12/28/2022] Open
Abstract
The Hirschsprung disease (HSCR) is an inherited disease that is controlled by multiple genes and has a complicated genetic mechanism. HSCR patients suffer from various extents of constipation due to dysplasia of the enteric nervous system (ENS), which can be so severe as to cause complete intestinal obstruction. Many genes have been identified as playing causative roles in ENS dysplasia and HSCR, among them the endothelin receptor type B gene (Ednrb) has been identified to play an important role. Mutation of Ednrb causes a series of symptoms that include deafness, pigmentary abnormalities, and aganglionosis. In our previous studies of three rat models carrying the same spotting lethal (sl) mutation on Ednrb, the haplotype of a region on chromosome (Chr) 2 was found to be responsible for the differing severities of the HSCR-like symptoms. To confirm that the haplotype of the responsible region on Chr 2 modifies the severity of aganglionosis caused by Ednrb mutation and to recreate a rat model with severe symptoms, we selected the GK inbred strain, whose haplotype in the responsible region on Chr 2 resembles that of the rat strain in which severe symptoms accompany the Ednrbsl mutation. An Ednrb mutation was introduced into the GK rat by crossing with F344-Ednrbsl and by genome editing. The null mutation of Ednrb was found to cause embryonic death in F2 progeny possessing the GK haplotype in the responsible region on Chr 2. The results of this study are unexpected, and they provide new clues and animal models that promise to contribute to studies on the genetic regulatory network in the development of ENS and on embryogenesis.
Collapse
|
64
|
Falomir-Lockhart LJ, Cavazzutti GF, Giménez E, Toscani AM. Fatty Acid Signaling Mechanisms in Neural Cells: Fatty Acid Receptors. Front Cell Neurosci 2019; 13:162. [PMID: 31105530 PMCID: PMC6491900 DOI: 10.3389/fncel.2019.00162] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/08/2019] [Indexed: 12/15/2022] Open
Abstract
Fatty acids (FAs) are typically associated with structural and metabolic roles, as they can be stored as triglycerides, degraded by β-oxidation or used in phospholipids’ synthesis, the main components of biological membranes. It has been shown that these lipids exhibit also regulatory functions in different cell types. FAs can serve as secondary messengers, as well as modulators of enzymatic activities and substrates for cytokines synthesis. More recently, it has been documented a direct activity of free FAs as ligands of membrane, cytosolic, and nuclear receptors, and cumulative evidence has emerged, demonstrating its participation in a wide range of physiological and pathological conditions. It has been long known that the central nervous system is enriched with poly-unsaturated FAs, such as arachidonic (C20:4ω-6) or docosohexaenoic (C22:6ω-3) acids. These lipids participate in the regulation of membrane fluidity, axonal growth, development, memory, and inflammatory response. Furthermore, a whole family of low molecular weight compounds derived from FAs has also gained special attention as the natural ligands for cannabinoid receptors or key cytokines involved in inflammation, largely expanding the role of FAs as precursors of signaling molecules. Nutritional deficiencies, and alterations in lipid metabolism and lipid signaling have been associated with developmental and cognitive problems, as well as with neurodegenerative diseases. The molecular mechanism behind these effects still remains elusive. But in the last two decades, different families of proteins have been characterized as receptors mediating FAs signaling. This review focuses on different receptors sensing and transducing free FAs signals in neural cells: (1) membrane receptors of the family of G Protein Coupled Receptors known as Free Fatty Acid Receptors (FFARs); (2) cytosolic transport Fatty Acid-Binding Proteins (FABPs); and (3) transcription factors Peroxisome Proliferator-Activated Receptors (PPARs). We discuss how these proteins modulate and mediate direct regulatory functions of free FAs in neural cells. Finally, we briefly discuss the advantages of evaluating them as potential targets for drug design in order to manipulate lipid signaling. A thorough characterization of lipid receptors of the nervous system could provide a framework for a better understanding of their roles in neurophysiology and, potentially, help for the development of novel drugs against aging and neurodegenerative processes.
Collapse
Affiliation(s)
- Lisandro Jorge Falomir-Lockhart
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Gian Franco Cavazzutti
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Ezequiel Giménez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Andrés Martín Toscani
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| |
Collapse
|
65
|
Holloway EM, Capeling MM, Spence JR. Biologically inspired approaches to enhance human organoid complexity. Development 2019; 146:dev166173. [PMID: 30992275 PMCID: PMC6503984 DOI: 10.1242/dev.166173] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organoids are complex three-dimensional in vitro organ-like model systems. Human organoids, which are derived from human pluripotent stem cells or primary human donor tissue, have been used to address fundamental questions about human development, stem cell biology and organ regeneration. Focus has now shifted towards implementation of organoids for biological discovery and advancing existing systems to more faithfully recapitulate the native organ. This work has highlighted significant unknowns in human biology and has invigorated new exploration into the cellular makeup of human organs during development and in the adult - work that is crucial for providing appropriate benchmarks for organoid systems. In this Review, we discuss efforts to characterize human organ cellular complexity and attempts to make organoid models more realistic through co-culture, transplantation and bioengineering approaches.
Collapse
Affiliation(s)
- Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
66
|
Barber K, Studer L, Fattahi F. Derivation of enteric neuron lineages from human pluripotent stem cells. Nat Protoc 2019; 14:1261-1279. [PMID: 30911172 DOI: 10.1038/s41596-019-0141-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 01/15/2019] [Indexed: 12/22/2022]
Abstract
The enteric nervous system (ENS) represents a vast network of neuronal and glial cell types that develops entirely from migratory neural crest (NC) progenitor cells. Considerable improvements in the understanding of the molecular mechanisms underlying NC induction and regional specification have recently led to the development of a robust method to re-create the process in vitro using human pluripotent stem cells (hPSCs). Directing the fate of hPSCs toward the enteric NC (ENC) results in an accessible and scalable in vitro model of ENS development. The application of hPSC-derived enteric neural lineages provides a powerful platform for ENS-related disease modeling and drug discovery. Here we present a detailed protocol for the induction of a regionally specific NC intermediate that occurs over the course of a 15-d interval and is an effective source for the in vitro derivation of functional enteric neurons (ENs) from hPSCs. Additionally, we introduce a new and improved protocol that we have developed to optimize the protocol for future applications in regenerative medicine, in which components of undefined activity have been replaced with fully defined culture conditions. This protocol provides access to a broad range of human ENS lineages within a 30-d period.
Collapse
Affiliation(s)
- Kevin Barber
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA. .,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA.
| | - Faranak Fattahi
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA. .,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
67
|
Jin K, Xiang M. Transcription factor Ptf1a in development, diseases and reprogramming. Cell Mol Life Sci 2019; 76:921-940. [PMID: 30470852 PMCID: PMC11105224 DOI: 10.1007/s00018-018-2972-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022]
Abstract
The transcription factor Ptf1a is a crucial helix-loop-helix (bHLH) protein selectively expressed in the pancreas, retina, spinal cord, brain, and enteric nervous system. Ptf1a is preferably assembled into a transcription trimeric complex PTF1 with an E protein and Rbpj (or Rbpjl). In pancreatic development, Ptf1a is indispensable in controlling the expansion of multipotent progenitor cells as well as the specification and maintenance of the acinar cells. In neural tissues, Ptf1a is transiently expressed in the post-mitotic cells and specifies the inhibitory neuronal cell fates, mostly mediated by downstream genes such as Tfap2a/b and Prdm13. Mutations in the coding and non-coding regulatory sequences resulting in Ptf1a gain- or loss-of-function are associated with genetic diseases such as pancreatic and cerebellar agenesis in the rodent and human. Surprisingly, Ptf1a alone is sufficient to reprogram mouse or human fibroblasts into tripotential neural stem cells. Its pleiotropic functions in many biological processes remain to be deciphered in the future.
Collapse
Affiliation(s)
- Kangxin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Mengqing Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
68
|
Saito-Diaz K, Zeltner N. Induced pluripotent stem cells for disease modeling, cell therapy and drug discovery in genetic autonomic disorders: a review. Clin Auton Res 2019; 29:367-384. [PMID: 30631982 DOI: 10.1007/s10286-018-00587-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022]
Abstract
The autonomic nervous system (ANS) regulates all organs in the body independent of consciousness, and is thus essential for maintaining homeostasis of the entire organism. Diseases of the ANS can arise due to environmental insults such as injury, toxins/drugs and infections or due to genetic lesions. Human studies and animal models have been instrumental to understanding connectivity and regulation of the ANS and its disorders. However, research into cellular pathologies and molecular mechanisms of ANS disorders has been hampered by the difficulties in accessing human patient-derived ANS cells in large numbers to conduct meaningful research, mainly because patient neurons cannot be easily biopsied and primary human neuronal cultures cannot be expanded.Human-induced pluripotent stem cell (hiPSC) technology can elegantly bridge these issues, allowing unlimited access of patient-derived ANS cell types for cellular, molecular and biochemical analysis, facilitating the discovery of novel therapeutic targets, and eventually leading to drug discovery. Additionally, such cells may provide a source for cell replacement therapy to replenish lost or injured ANS tissue in patients.Here, we first review the anatomy and embryonic development of the ANS, as this knowledge is crucial for understanding disease modeling approaches. We then review the current advances in human stem cell technology for modeling diseases of the ANS, recent strides toward cell replacement therapy and drug discovery initiatives.
Collapse
Affiliation(s)
- Kenyi Saito-Diaz
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA. .,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA. .,Department of Cellular Biology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
69
|
Abstract
The adult gastrointestinal tract (GI) is a series of connected organs (esophagus, stomach, small intestine, colon) that develop via progressive regional specification of a continuous tubular embryonic organ anlage. This chapter focuses on organogenesis of the small intestine. The intestine arises by folding of a flat sheet of endodermal cells into a tube of highly proliferative pseudostratified cells. Dramatic elongation of this tube is driven by rapid epithelial proliferation. Then, epithelial-mesenchymal crosstalk and physical forces drive a stepwise cascade that results in convolution of the tubular surface into finger-like projections called villi. Concomitant with villus formation, a sharp epithelial transcriptional boundary is defined between stomach and intestine. Finally, flask-like depressions called crypts are established to house the intestinal stem cells needed throughout life for epithelial renewal. New insights into these events are being provided by in vitro organoid systems, which hold promise for future regenerative engineering of the small intestine.
Collapse
Affiliation(s)
- Sha Wang
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| | - Katherine D Walton
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States.
| | - Deborah L Gumucio
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| |
Collapse
|
70
|
Zuber SM, Grikscheit TC. Stem cells for babies and their surgeons: The future is now. J Pediatr Surg 2019; 54:16-20. [PMID: 30497818 DOI: 10.1016/j.jpedsurg.2018.10.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/01/2018] [Indexed: 12/27/2022]
Abstract
Pediatric surgeons are ideal allies for the translation of basic science including stem cell therapies. In the spirit of Robert E. Gross, of applying creative solutions to pediatric problems with technical expertise, we describe the impending cellular therapies that may be derived from stem and progenitor cells. Understanding the types and capabilities of stem and progenitor cells is important for pediatric surgeons to join and facilitate progress for babies. We are developing an induced pluripotent stem cell therapy for enteric neuropathies such as Hirschsprung disease that might be helpful for children in the near future. Our goals, which we hope to share with other surgeons and scientists, include working to establish safe clinical trials and meeting regulatory standards in a thoughtful way that balances patients need and unknown risks.
Collapse
Affiliation(s)
- Samuel M Zuber
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90027, USA
| | - Tracy C Grikscheit
- Division of Pediatric Surgery, Children's Hospital Los Angeles, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90027, USA.
| |
Collapse
|
71
|
Chesné J, Cardoso V, Veiga-Fernandes H. Neuro-immune regulation of mucosal physiology. Mucosal Immunol 2019; 12:10-20. [PMID: 30089849 DOI: 10.1038/s41385-018-0063-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023]
Abstract
Mucosal barriers constitute major body surfaces that are in constant contact with the external environment. Mucosal sites are densely populated by a myriad of distinct neurons and immune cell types that sense, integrate and respond to multiple environmental cues. In the recent past, neuro-immune interactions have been reported to play central roles in mucosal health and disease, including chronic inflammatory conditions, allergy and infectious diseases. Discrete neuro-immune cell units act as building blocks of this bidirectional multi-tissue cross-talk, ensuring mucosal tissue health and integrity. Herein, we will focus on reciprocal neuro-immune interactions in the airways and intestine. Such neuro-immune cross-talk maximizes sensing and integration of environmental aggressions, which can be considered an important paradigm shift in our current views of mucosal physiology and immune regulation.
Collapse
Affiliation(s)
- Julie Chesné
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038, Lisboa, Portugal
| | - Vânia Cardoso
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038, Lisboa, Portugal
| | | |
Collapse
|
72
|
Porokuokka LL, Virtanen HT, Lindén J, Sidorova Y, Danilova T, Lindahl M, Saarma M, Andressoo JO. Gfra1 Underexpression Causes Hirschsprung's Disease and Associated Enterocolitis in Mice. Cell Mol Gastroenterol Hepatol 2018; 7:655-678. [PMID: 30594740 PMCID: PMC6444303 DOI: 10.1016/j.jcmgh.2018.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS RET, the receptor for the glial cell line-derived neurotrophic factor (GDNF) family ligands, is the most frequently mutated gene in congenital aganglionic megacolon or Hirschsprung's disease (HSCR). The leading cause of mortality in HSCR is HSCR-associated enterocolitis (HAEC), which is characterized by altered mucin composition, mucin retention, bacterial adhesion to enterocytes, and epithelial damage, although the order of these events is obscure. In mice, loss of GDNF signaling leads to a severely underdeveloped enteric nervous system and neonatally fatal kidney agenesis, thereby precluding the use of these mice for modeling postnatal HSCR and HAEC. Our aim was to generate a postnatally viable mouse model for HSCR/HAEC and analyze HAEC etiology. METHODS GDNF family receptor alpha-1 (GFRa1) hypomorphic mice were generated by placing a selectable marker gene in the sixth intron of the Gfra1 locus using gene targeting in mouse embryonic stem cells. RESULTS We report that 70%-80% reduction in GDNF co-receptor GFRa1 expression levels in mice results in HSCR and HAEC, leading to death within the first 25 postnatal days. These mice mirror the disease progression and histopathologic findings in children with untreated HSCR/HAEC. CONCLUSIONS In GFRa1 hypomorphic mice, HAEC proceeds from goblet cell dysplasia, with abnormal mucin production and retention, to epithelial damage. Microbial enterocyte adherence and tissue invasion are late events and therefore unlikely to be the primary cause of HAEC. These results suggest that goblet cells may be a potential target for preventative treatment and that reduced expression of GFRa1 may contribute to HSCR susceptibility.
Collapse
Affiliation(s)
| | - Heikki T Virtanen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jere Lindén
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Yulia Sidorova
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Tatiana Danilova
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
73
|
Intracellular emetic signaling cascades by which the selective neurokinin type 1 receptor (NK 1R) agonist GR73632 evokes vomiting in the least shrew (Cryptotis parva). Neurochem Int 2018; 122:106-119. [PMID: 30453005 DOI: 10.1016/j.neuint.2018.11.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/26/2022]
Abstract
To characterize mechanisms involved in neurokinin type 1 receptor (NK1R)-mediated emesis, we investigated the brainstem emetic signaling pathways following treating least shrews with the selective NK1R agonist GR73632. In addition to episodes of vomiting over a 30-min observation period, a significant increase in substance P-immunoreactivity in the emetic brainstem dorsal motor nucleus of the vagus (DMNX) occurred at 15 min post an intraperitoneal (i.p.) injection GR73632 (5 mg/kg). In addition, time-dependent upregulation of phosphorylation of several emesis -associated protein kinases occurred in the brainstem. In fact, Western blots demonstrated significant phosphorylations of Ca2+/calmodulin kinase IIα (CaMKIIα), extracellular signal-regulated protein kinase1/2 (ERK1/2), protein kinase B (Akt) as well as α and βII isoforms of protein kinase C (PKCα/βII). Moreover, enhanced phospho-ERK1/2 immunoreactivity was also observed in both brainstem slices containing the dorsal vagal complex emetic nuclei as well as in jejunal sections from the shrew small intestine. Furthermore, our behavioral findings demonstrated that the following agents suppressed vomiting evoked by GR73632 in a dose-dependent manner: i) the NK1R antagonist netupitant (i.p.); ii) the L-type Ca2+ channel (LTCC) antagonist nifedipine (subcutaneous, s.c.); iii) the inositol trisphosphate receptor (IP3R) antagonist 2-APB (i.p.); iv) store-operated Ca2+ entry inhibitors YM-58483 and MRS-1845, (i.p.); v) the ERK1/2 pathway inhibitor U0126 (i.p.); vi) the PKC inhibitor GF109203X (i.p.); and vii) the inhibitor of phosphatidylinositol 3-kinase (PI3K)-Akt pathway LY294002 (i.p.). Moreover, NK1R, LTCC, and IP3R are required for GR73632-evoked CaMKIIα, ERK1/2, Akt and PKCα/βII phosphorylation. In addition, evoked ERK1/2 phosphorylation was sensitive to inhibitors of PKC and PI3K. These findings indicate that the LTCC/IP3R-dependent PI3K/PKCα/βII-ERK1/2 signaling pathways are involved in NK1R-mediated vomiting.
Collapse
|
74
|
Hu H, Ding Y, Mu W, Li Y, Wang Y, Jiang W, Fu Y, Tou J, Chen W. DRG-Derived Neural Progenitors Differentiate into Functional Enteric Neurons Following Transplantation in the Postnatal Colon. Cell Transplant 2018; 28:157-169. [PMID: 30442032 PMCID: PMC6362519 DOI: 10.1177/0963689718811061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cell therapy has great promise for treating gastrointestinal motility disorders caused by intestinal nervous system (ENS) diseases. However, appropriate sources, other than enteric neural stem cells and human embryonic stem cells, are seldom reported. Here, we show that neural progenitors derived from the dorsal root ganglion (DRG) of EGFP mice survived, differentiated into enteric neurons and glia cells, migrated widely from the site of injection, and established neuron-muscle connections following transplantation into the distal colon of postnatal mice. The exogenous EGFP+ neurons were physiologically functional as shown by the activity of calcium imaging. This study shows that that other tissues besides the postnatal bowel harbor neural crest stem cells or neural progenitors that have the potential to differentiate into functional enteric neurons in vivo and can potentially be used for intestinal nerve regeneration. These DRG-derived neural progenitor cells may be a choice for cell therapy of ENS disease as an allograft. The new knowledge provided by our study is important for the development of neural crest stem cell and cell therapy for the treatment of intestinal neuropathy.
Collapse
Affiliation(s)
- Hui Hu
- 1 Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, School of Medicine, Zhejiang University, China.,2 Institute of Translational Medicine, School of Medicine, Zhejiang University, China
| | - Yuanyuan Ding
- 1 Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, School of Medicine, Zhejiang University, China.,2 Institute of Translational Medicine, School of Medicine, Zhejiang University, China
| | - Wenbo Mu
- 1 Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, School of Medicine, Zhejiang University, China.,2 Institute of Translational Medicine, School of Medicine, Zhejiang University, China
| | - Ying Li
- 1 Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, School of Medicine, Zhejiang University, China.,2 Institute of Translational Medicine, School of Medicine, Zhejiang University, China
| | - Yanpeng Wang
- 3 Department of Gynecology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, China
| | - Weifang Jiang
- 4 Department of Neonatal Surgery, Children's Hospital, School of Medicine, Zhejiang University, China
| | - Yong Fu
- 1 Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, School of Medicine, Zhejiang University, China.,5 Otolaryngological Department, Children's Hospital, School of Medicine, Zhejiang University, China
| | - Jinfa Tou
- 1 Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, School of Medicine, Zhejiang University, China.,4 Department of Neonatal Surgery, Children's Hospital, School of Medicine, Zhejiang University, China
| | - Wei Chen
- 1 Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, School of Medicine, Zhejiang University, China.,2 Institute of Translational Medicine, School of Medicine, Zhejiang University, China.,6 Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, School of Medicine, Zhejiang University, China
| |
Collapse
|
75
|
Hirschsprung's Associated Enterocolitis (HAEC) Personalized Treatment with Probiotics Based on Gene Sequencing Analysis of the Fecal Microbiome. Case Rep Pediatr 2018; 2018:3292309. [PMID: 30405931 PMCID: PMC6204186 DOI: 10.1155/2018/3292309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/19/2018] [Indexed: 11/23/2022] Open
Abstract
Approximately 40% of children with Hirschsprung's disease (HD) suffer from Hirschsprung's associated enterocolitis (HAEC) despite correct surgery. Disturbances of the intestinal microbiome may play a role. Treatment with probiotics based on individual analyses of the fecal microbiome has not been published for HD patients with recurrent HAEC yet. A boy with trisomy 21 received transanal pull-through at the age of 6 months for rectosigmoid HD. With four years, he suffered from recurrent episodes of HAEC. The fecal microbiome was measured during three healthy and three HAEC episodes by next-generation sequencing. The patient was started on daily probiotics for 3 months; the fecal microbiome was measured weekly. The fecal microbiome differed significantly between healthy and HAEC episodes. HAEC episodes were associated with significant decreases of Actinobacteria and significant increases of Bacteroidetes and Proteobacteria. Probiotic treatment led to a significant increase of alpha diversity and a significant increase of Bifidobacterium and Streptococcus as well as decreases of Rikenellaceae, Pseudobutyrivibrio, Blautia, and Lachnospiraceae. A longitudinal observation of the microbiome has never been performed following correction of Hirschsprung's disease. Probiotic treatment significantly changed the fecal microbiome; the alterations were not limited to strains contained in the administered probiotics.
Collapse
|
76
|
López SH, Avetisyan M, Wright CM, Mesbah K, Kelly RG, Moon AM, Heuckeroth RO. Loss of Tbx3 in murine neural crest reduces enteric glia and causes cleft palate, but does not influence heart development or bowel transit. Dev Biol 2018; 444 Suppl 1:S337-S351. [PMID: 30292786 DOI: 10.1016/j.ydbio.2018.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/23/2018] [Accepted: 09/23/2018] [Indexed: 01/12/2023]
Abstract
Transcription factors that coordinate migration, differentiation or proliferation of enteric nervous system (ENS) precursors are not well defined. To identify novel transcriptional regulators of ENS development, we performed microarray analysis at embryonic day (E) 17.5 and identified many genes that were enriched in the ENS compared to other bowel cells. We decided to investigate the T-box transcription factor Tbx3, which is prominently expressed in developing and mature ENS. Haploinsufficiency for TBX3 causes ulnar-mammary syndrome (UMS) in humans, a multi-organ system disorder. TBX3 also regulates several genes known to be important for ENS development. To test the hypothesis that Tbx3 is important for ENS development or function, we inactivated Tbx3 in all neural crest derivatives, including ENS progenitors using Wnt1-Cre and a floxed Tbx3 allele. Tbx3 fl/fl; Wnt1-Cre conditional mutant mice die shortly after birth with cleft palate and difficulty feeding. The ENS of mutants was well-organized with a normal density of enteric neurons and nerve fiber bundles, but small bowel glial cell density was reduced. Despite this, bowel motility appeared normal. Furthermore, although Tbx3 is expressed in cardiac neural crest, Tbx3 fl/fl; Wnt1-Cre mice had structurally normal hearts. Thus, loss of Tbx3 within neural crest has selective effects on Tbx3-expressing neural crest derivatives.
Collapse
Affiliation(s)
- Silvia Huerta López
- The Children's Hospital of Philadelphia Research Institute, 3615 Civic Center Blvd, Abramson Research Center - Suite # 1116I, Philadelphia, PA 19104-4318, United States
| | - Marina Avetisyan
- The Children's Hospital of Philadelphia Research Institute, 3615 Civic Center Blvd, Abramson Research Center - Suite # 1116I, Philadelphia, PA 19104-4318, United States; Department of Pediatrics, Washington University School of Medicine in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, United States
| | - Christina M Wright
- The Children's Hospital of Philadelphia Research Institute, 3615 Civic Center Blvd, Abramson Research Center - Suite # 1116I, Philadelphia, PA 19104-4318, United States; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-4318, United States
| | - Karim Mesbah
- Aix-Marseille Univ, CNRS, IBDM, Marseille, France
| | | | - Anne M Moon
- Weis Center for Research, Geisinger Clinic, Danville, PA, United States; Departments of Pediatrics and Human Genetics, University of Utah, Salt Lake City, United States
| | - Robert O Heuckeroth
- The Children's Hospital of Philadelphia Research Institute, 3615 Civic Center Blvd, Abramson Research Center - Suite # 1116I, Philadelphia, PA 19104-4318, United States; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-4318, United States.
| |
Collapse
|
77
|
Bondurand N, Dufour S, Pingault V. News from the endothelin-3/EDNRB signaling pathway: Role during enteric nervous system development and involvement in neural crest-associated disorders. Dev Biol 2018; 444 Suppl 1:S156-S169. [PMID: 30171849 DOI: 10.1016/j.ydbio.2018.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/27/2018] [Accepted: 08/27/2018] [Indexed: 01/08/2023]
Abstract
The endothelin system is a vertebrate-specific innovation with important roles in regulating the cardiovascular system and renal and pulmonary processes, as well as the development of the vertebrate-specific neural crest cell population and its derivatives. This system is comprised of three structurally similar 21-amino acid peptides that bind and activate two G-protein coupled receptors. In 1994, knockouts of the Edn3 and Ednrb genes revealed their crucial function during development of the enteric nervous system and melanocytes, two neural-crest derivatives. Since then, human and mouse genetics, combined with cellular and developmental studies, have helped to unravel the role of this signaling pathway during development and adulthood. In this review, we will summarize the known functions of the EDN3/EDNRB pathway during neural crest development, with a specific focus on recent scientific advances, and the enteric nervous system in normal and pathological conditions.
Collapse
Affiliation(s)
- Nadege Bondurand
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM U1163, Institut Imagine, Paris, France; Paris Descartes-Sorbonne Paris Cité University, Institut Imagine, Paris, France.
| | - Sylvie Dufour
- INSERM, U955, Equipe 06, Créteil 94000, France; Université Paris Est, Faculté de Médecine, Créteil 94000, France
| | - Veronique Pingault
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM U1163, Institut Imagine, Paris, France; Paris Descartes-Sorbonne Paris Cité University, Institut Imagine, Paris, France; Service de Génétique Moléculaire, Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
78
|
Courtney CM, Onufer EJ, Seiler KM, Warner BW. An anatomic approach to understanding mechanisms of intestinal adaptation. Semin Pediatr Surg 2018; 27:229-236. [PMID: 30342597 DOI: 10.1053/j.sempedsurg.2018.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Cathleen M Courtney
- Division of Pediatric Surgery, St. Louis Children's Hospital, One Children's Place, Suite 6110, St. Louis, 63110 MO, USA; Department of Surgery, Washington University School of Medicine, St. Louis, USA
| | - Emily J Onufer
- Division of Pediatric Surgery, St. Louis Children's Hospital, One Children's Place, Suite 6110, St. Louis, 63110 MO, USA; Department of Surgery, Washington University School of Medicine, St. Louis, USA
| | - Kristen M Seiler
- Division of Pediatric Surgery, St. Louis Children's Hospital, One Children's Place, Suite 6110, St. Louis, 63110 MO, USA; Department of Surgery, Washington University School of Medicine, St. Louis, USA
| | - Brad W Warner
- Division of Pediatric Surgery, St. Louis Children's Hospital, One Children's Place, Suite 6110, St. Louis, 63110 MO, USA; Department of Surgery, Washington University School of Medicine, St. Louis, USA.
| |
Collapse
|
79
|
Cocaine- and amphetamine-regulated transcript (CART) peptide in the enteric nervous system of the porcine esophagus. C R Biol 2018; 341:325-333. [PMID: 29983247 DOI: 10.1016/j.crvi.2018.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 05/10/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022]
Abstract
Cocaine- and amphetamine-regulated transcript peptide (CART) is widely distributed within the central and peripheral nervous system. In the brain, CART is considered as the main anorectic peptide involved in the regulation of food intake. Contrary to the central nervous system, a lot of aspects connected with the distribution and functions of CART within the enteric nervous system (ENS) still remain unknown. The aim of the present study was to investigate, for the first time, the population of CART-like immunoreactive (CART-LI) neurons within the porcine esophagus and the denotation of their neurochemical coding. During this experiment, the distribution of CART-LI neurons and the colocalization of CART with other neuronal active substances were examined using standard double- and triple-immunofluorescence techniques in enteric plexuses of cervical, thoracic, and abdominal esophagus fragments. The obtained results showed that CART is present in a relatively high percentage of esophageal neurons (values fluctuated from 45.2±0.9% in the submucous plexus of the thoracic esophagus to 58.1±5.0% in the myenteric plexus of the same fragment of the esophagus). Moreover, CART colocalized with a wide range of other active neuronal substances, mainly with the vesicular acetylcholine transporter (VAChT, a marker of cholinergic neurons), neuronal isoform of nitric oxide synthase (nNOS, a marker of nitrergic neurons), vasoactive intestinal polypeptide (VIP) and galanin (GAL). The number of CART-positive neuronal cells and their neurochemical coding clearly depended on the fragment of esophagus studied and the type of enteric plexus. The obtained results suggest that CART may play important and multidirectional roles in the neuronal regulation of esophageal functions.
Collapse
|
80
|
Tang CS, Zhuang X, Lam WY, Ngan ESW, Hsu JS, Michelle YU, Man-Ting SO, Cherny SS, Ngo ND, Sham PC, Tam PK, Garcia-Barcelo MM. Uncovering the genetic lesions underlying the most severe form of Hirschsprung disease by whole-genome sequencing. Eur J Hum Genet 2018; 26:818-826. [PMID: 29483666 PMCID: PMC5974185 DOI: 10.1038/s41431-018-0129-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 02/02/2018] [Accepted: 02/13/2018] [Indexed: 11/08/2022] Open
Abstract
Hirschsprung disease (HSCR) is a complex birth defect characterized by the lack of ganglion cells along a variable length of the distal intestine. A large proportion of HSCR patients remain genetically unexplained. We applied whole-genome sequencing (WGS) on 9 trios where the probands are sporadically affected with the most severe form of the disorder and harbor no coding sequence variants affecting the function of known HSCR genes. We found de novo protein-altering variants in three intolerant to change genes-CCT2, VASH1, and CYP26A1-for which a plausible link with the enteric nervous system (ENS) exists. De novo single-nucleotide and indel variants were present in introns and non-coding neighboring regions of ENS-related genes, including NRG1 and ERBB4. Joint analysis with those inherited rare variants found under recessive and/or digenic models revealed both patient-unique and shared genetic features where rare variants were found to be enriched in the extracellular matrix-receptor (ECM-receptor) pathway (p = 3.4 × 10-11). Delineation of the genetic profile of each patient might help finding common grounds that could lead to the discovery of shared molecules that could be used as drug targets for the currently ongoing cell therapy effort which aims at providing an alternative to the surgical treatment.
Collapse
Affiliation(s)
- Clara Sm Tang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, Hong Kong, Hong Kong
| | - Xuehan Zhuang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Wai-Yee Lam
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Elly Sau-Wai Ngan
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jacob Shujui Hsu
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Y U Michelle
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - S O Man-Ting
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Stacey S Cherny
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | | | - Pak C Sham
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Paul Kh Tam
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, Hong Kong, Hong Kong
| | - Maria-Mercè Garcia-Barcelo
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
81
|
Delvalle NM, Dharshika C, Morales-Soto W, Fried DE, Gaudette L, Gulbransen BD. Communication Between Enteric Neurons, Glia, and Nociceptors Underlies the Effects of Tachykinins on Neuroinflammation. Cell Mol Gastroenterol Hepatol 2018; 6:321-344. [PMID: 30116771 PMCID: PMC6091443 DOI: 10.1016/j.jcmgh.2018.05.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 05/18/2018] [Indexed: 12/18/2022]
Abstract
Background & Aims Tachykinins are involved in physiological and pathophysiological mechanisms in the gastrointestinal tract. The major sources of tachykinins in the gut are intrinsic enteric neurons in the enteric nervous system and extrinsic nerve fibers from the dorsal root and vagal ganglia. Although tachykinins are important mediators in the enteric nervous system, how they contribute to neuroinflammation through effects on neurons and glia is not fully understood. Here, we tested the hypothesis that tachykinins contribute to enteric neuroinflammation through mechanisms that involve intercellular neuron-glia signaling. Methods We used immunohistochemistry and quantitative real-time polymerase chain reaction, and studied cellular activity using transient-receptor potential vanilloid-1 (TRPV1)tm1(cre)Bbm/J::Polr2atm1(CAG-GCaMP5g,-tdTomato)Tvrd and Sox10CreERT2::Polr2atm1(CAG-GCaMP5g,-tdTomato)Tvrd mice or Fluo-4. We used the 2,4-di-nitrobenzene sulfonic acid (DNBS) model of colitis to study neuroinflammation, glial reactivity, and neurogenic contractility. We used Sox10::CreERT2+/-/Rpl22tm1.1Psam/J mice to selectively study glial transcriptional changes. Results Tachykinins are expressed predominantly by intrinsic neuronal varicosities whereas neurokinin-2 receptors (NK2Rs) are expressed predominantly by enteric neurons and TRPV1-positive neuronal varicosities. Stimulation of NK2Rs drives responses in neuronal varicosities that are propagated to enteric glia and neurons. Antagonizing NK2R signaling enhanced recovery from colitis and prevented the development of reactive gliosis, neuroinflammation, and enhanced neuronal contractions. Inflammation drove changes in enteric glial gene expression and function, and antagonizing NK2R signaling mitigated these changes. Neurokinin A-induced neurodegeneration requires glial connexin-43 hemichannel activity. Conclusions Our results show that tachykinins drive enteric neuroinflammation through a multicellular cascade involving enteric neurons, TRPV1-positive neuronal varicosities, and enteric glia. Therapies targeting components of this pathway could broadly benefit the treatment of dysmotility and pain after acute inflammation in the intestine.
Collapse
Key Words
- BzATP, 2’(3’)-O-(4-benzoylbenzoyl)adenosine 5’-triphosphate triethylammonium salt
- Ca2+, calcium
- Colitis
- Cx43, connexin-43
- DMEM, Dulbecco's modified Eagle medium
- DNBS, dinitrobenzene sulfonic acid
- EFS, electrical field stimulation
- ENS, enteric nervous system
- Enteric Nervous System
- FGID, functional gastrointestinal disorder
- GFAP, glial fibrillary acidic protein
- GI, gastrointestinal
- Glia
- HA, hemagglutinin
- IPAN, intrinsic primarily afferent neuron
- LMMP, longitudinal muscle–myenteric plexus
- MSU, Michigan State University
- NK1R, neurokinin-1 receptor
- NK2R, neurokinin-2 receptor
- NKA, neurokinin A
- Neurokinins
- SP, substance P
- TRPV1, transient receptor potential vanilloid-1
- mRNA, messenger RNA
Collapse
Affiliation(s)
| | - Christine Dharshika
- Genetics Program, Michigan State University, East Lansing, Michigan
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan
| | | | - David E. Fried
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Lukas Gaudette
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Brian D. Gulbransen
- Neuroscience Program, Michigan State University, East Lansing, Michigan
- Department of Physiology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
82
|
Zhai X, Lin D, Zhao Y, Yang X. Bacterial Cellulose Relieves Diphenoxylate-Induced Constipation in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:4106-4117. [PMID: 29627986 DOI: 10.1021/acs.jafc.8b00385] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This study was to probe the effects of bacterial cellulose (BC) on diphenoxylate-induced constipation in rats. Administration with BC at 500 mg/kg of body weight in diphenoxylate-induced constipation rats distinctly improved the carmine propulsion rate (83.5 ± 5.2%), shortened the defecating time of the first red feces (249.0 ± 23.3 min), and increased the weight of carmine red feces within 5 h (2.7 ± 1.3 g). The levels of aquaporins (AQP-2, AQP-3, and AQP-4) and inhibitory neurotransmitters (nitric oxide, nitric oxide synthetase, vasoactive intestinal peptide, and arginine vasopressin) in the BC-treated groups reduced by 31.9-40.0% ( p < 0.01) and 21.1-67.7% ( p < 0.01) compared to those in the constipation group, respectively. However, the secretion of excitability neurotransmitters (substance P and motilin) in the BC-treated groups was increased by 20.0-39.9% ( p < 0.01). The activities of ATPases in the colon of constipation rats were significantly weakened by BC administration ( p < 0.01). Histological morphology of the colon showed that BC supplementation could effectively increase the length of villus cells and the thickness of colonic mucosa and muscle ( p < 0.01). Moreover, BC supplementation could protect colonic smooth muscle cells against apoptosis. All of the findings suggest that BC supplementation effectively relieves constipation in rats and BC would be used as a great promising dietary fiber for alleviating constipation.
Collapse
|
83
|
Bonafina A, Fontanet PA, Paratcha G, Ledda F. GDNF/GFRα1 Complex Abrogates Self-Renewing Activity of Cortical Neural Precursors Inducing Their Differentiation. Stem Cell Reports 2018; 10:1000-1015. [PMID: 29478900 PMCID: PMC5918270 DOI: 10.1016/j.stemcr.2018.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/30/2022] Open
Abstract
The balance between factors leading to proliferation and differentiation of cortical neural precursors (CNPs) determines the correct cortical development. In this work, we show that GDNF and its receptor GFRα1 are expressed in the neocortex during the period of cortical neurogenesis. We show that the GDNF/GFRα1 complex inhibits the self-renewal capacity of mouse CNP cells induced by fibroblast growth factor 2 (FGF2), promoting neuronal differentiation. While GDNF leads to decreased proliferation of cultured cortical precursor cells, ablation of GFRα1 in glutamatergic cortical precursors enhances its proliferation. We show that GDNF treatment of CNPs promoted morphological differentiation even in the presence of the self-renewal-promoting factor, FGF2. Analysis of GFRα1-deficient mice shows an increase in the number of cycling cells during cortical development and a reduction in dendrite development of cortical GFRα1-expressing neurons. Together, these results indicate that GDNF/GFRα1 signaling plays an essential role in regulating the proliferative condition and the differentiation of cortical progenitors. GFRα1 receptor is expressed in the neocortex during the period of neurogenesis GDNF/GFRα1 complex inhibits self-renewing of cortical neuronal precursors GDNF and GFRα1 promote neurogenic differentiation of cortical neural progenitors Requirement of GFRα1 for proper dendrite development of cortical neurons
Collapse
Affiliation(s)
- Antonela Bonafina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina
| | - Paula Aldana Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina
| | - Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina.
| |
Collapse
|
84
|
Fujiwara N, Nakazawa-Tanaka N, Miyahara K, Arikawa-Hirasawa E, Akazawa C, Yamataka A. Altered expression of laminin alpha1 in aganglionic colon of endothelin receptor-B null mouse model of Hirschsprung's disease. Pediatr Surg Int 2018; 34:137-141. [PMID: 28983681 DOI: 10.1007/s00383-017-4180-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2017] [Indexed: 01/24/2023]
Abstract
PURPOSE Laminin, an extracellular matrix molecule, is essential for normal development of the nervous system. The alpha1 subunit of laminin-1 (LAMA1) has been reported to promote neurites and outgrowth and is expressed only during embryogenesis. Previously, we developed a Sox10 transgenic version of the Endothelin receptor-B (Ednrb) mouse to visualize Enteric neural crest-derived cell (ENCC)s with a green fluorescent protein, Venus. We designed this study to investigate the expression of LAMA1 using Sox10-VENUS mice gut. METHODS We harvested the gut on days 13.5 (E13.5) and 15.5 (E15.5) of gestation. Sox10-VENUS+/Ednrb -/- mice (n = 8) were compared with Sox10-VENUS+/Ednrb +/+ mice (n = 8) as controls. Gene expression of LAMA1 was analysed by real-time RT-PCR. Fluorescent immunohistochemistry was performed to assess protein distribution. RESULTS The relative mRNA expression levels of LAMA1 were significantly increased in HD in the proximal and distal colon on E15.5 compared to controls (p < 0.05), whereas there were no significant differences on E13.5. LAMA1 was expressed in the serosa, submucosa and basal lamina in the gut, and was markedly increased in the proximal and distal colon of HD on E15.5. CONCLUSIONS Altered LAMA1 expression in the aganglionic region may contribute to impaired ENCC migration, resulting in HD. These data could help in understanding the pathophysiologic interactions between LAMA1 and ENCC migration.
Collapse
Affiliation(s)
- Naho Fujiwara
- Department of Pediatric Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Nana Nakazawa-Tanaka
- Department of Pediatric Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Department of Pediatric Surgery, Juntendo Nerima Hospital, Tokyo, Japan
| | - Katsumi Miyahara
- Department of Pediatric Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Eri Arikawa-Hirasawa
- Research Institute for Disease of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Chihiro Akazawa
- Department of Biochemistry and Biophysics, Graduate School of Health Care Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsuyuki Yamataka
- Department of Pediatric Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
85
|
Lasrado R, Boesmans W, Kleinjung J, Pin C, Bell D, Bhaw L, McCallum S, Zong H, Luo L, Clevers H, Vanden Berghe P, Pachnis V. Lineage-dependent spatial and functional organization of the mammalian enteric nervous system. Science 2018; 356:722-726. [PMID: 28522527 DOI: 10.1126/science.aam7511] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022]
Abstract
The enteric nervous system (ENS) is essential for digestive function and gut homeostasis. Here we show that the amorphous neuroglia networks of the mouse ENS are composed of overlapping clonal units founded by postmigratory neural crest-derived progenitors. The spatial configuration of ENS clones depends on proliferation-driven local interactions of ENS progenitors with lineally unrelated neuroectodermal cells, the ordered colonization of the serosa-mucosa axis by clonal descendants, and gut expansion. Single-cell transcriptomics and mutagenesis analysis delineated dynamic molecular states of ENS progenitors and identified RET as a regulator of neurogenic commitment. Clonally related enteric neurons exhibit synchronous activity in response to network stimulation. Thus, lineage relationships underpin the organization of the peripheral nervous system.
Collapse
Affiliation(s)
- Reena Lasrado
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Werend Boesmans
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.,Laboratory for Enteric Neuroscience (LENS), Translational Research in GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - Jens Kleinjung
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Carmen Pin
- Institute of Food Research, Norwich NR4 7UA, UK
| | - Donald Bell
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Leena Bhaw
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sarah McCallum
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Hui Zong
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Hans Clevers
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Centre Utrecht, 3584 CT Utrecht, Netherlands
| | - Pieter Vanden Berghe
- Laboratory for Enteric Neuroscience (LENS), Translational Research in GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - Vassilis Pachnis
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
86
|
Coble JL, Sheldon KE, Yue F, Salameh TJ, Harris LR, Deiling S, Ruggiero FM, Eshelman MA, Yochum GS, Koltun WA, Gerhard GS, Broach JR. Identification of a rare LAMB4 variant associated with familial diverticulitis through exome sequencing. Hum Mol Genet 2018; 26:3212-3220. [PMID: 28595269 DOI: 10.1093/hmg/ddx204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 05/23/2017] [Indexed: 12/13/2022] Open
Abstract
Diverticulitis is a chronic disease of the colon in which diverticuli, or outpouching through the colonic wall, become inflamed. Although recent observations suggest that genetic factors may play a significant role in diverticulitis, few genes have yet been implicated in disease pathogenesis and familial cases are uncommon. Here, we report results of whole exome sequencing performed on members from a single multi-generational family with early onset diverticulitis in order to identify a genetic component of the disease. We identified a rare single nucleotide variant in the laminin β 4 gene (LAMB4) that segregated with disease in a dominant pattern and causes a damaging missense substitution (D435N). Targeted sequencing of LAMB4 in 148 non-familial and unrelated sporadic diverticulitis patients identified two additional rare variants in the gene. Immunohistochemistry indicated that LAMB4 localizes to the myenteric plexus of colonic tissue and patients harboring LAMB4 variants exhibited reduced LAMB4 protein levels relative to controls. Laminins are constituents of the extracellular matrix and play a major role in regulating the development and function of the enteric nervous system. Reduced LAMB4 levels may therefore alter innervation and morphology of the enteric nervous system, which may contribute to colonic dysmotility associated with diverticulitis.
Collapse
Affiliation(s)
- Joel L Coble
- Department of Biochemistry and Molecular Biology
| | | | - Feng Yue
- Department of Biochemistry and Molecular Biology
| | | | | | - Sue Deiling
- Department of Surgery, Division of Colon and Rectal Surgery
| | - Francesca M Ruggiero
- Division of Anatomical Pathology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | - Gregory S Yochum
- Department of Biochemistry and Molecular Biology.,Department of Surgery, Division of Colon and Rectal Surgery
| | | | - Glenn S Gerhard
- Department of Medical Genetics and Molecular Biochemistry, Temple University College of Medicine, Philadelphia, PA 19140, USA
| | | |
Collapse
|
87
|
Uribe RA, Hong SS, Bronner ME. Retinoic acid temporally orchestrates colonization of the gut by vagal neural crest cells. Dev Biol 2018; 433:17-32. [PMID: 29108781 PMCID: PMC5722660 DOI: 10.1016/j.ydbio.2017.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
Abstract
The enteric nervous system arises from neural crest cells that migrate as chains into and along the primitive gut, subsequently differentiating into enteric neurons and glia. Little is known about the mechanisms governing neural crest migration en route to and along the gut in vivo. Here, we report that Retinoic Acid (RA) temporally controls zebrafish enteric neural crest cell chain migration. In vivo imaging reveals that RA loss severely compromises the integrity and migration of the chain of neural crest cells during the window of time window when they are moving along the foregut. After loss of RA, enteric progenitors accumulate in the foregut and differentiate into enteric neurons, but subsequently undergo apoptosis resulting in a striking neuronal deficit. Moreover, ectopic expression of the transcription factor meis3 and/or the receptor ret, partially rescues enteric neuron colonization after RA attenuation. Collectively, our findings suggest that retinoic acid plays a critical temporal role in promoting enteric neural crest chain migration and neuronal survival upstream of Meis3 and RET in vivo.
Collapse
Affiliation(s)
- Rosa A Uribe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Department of Biosciences, Rice University, Houston, TX 77005, USA.
| | - Stephanie S Hong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
88
|
Moeser AJ, Pohl CS, Rajput M. Weaning stress and gastrointestinal barrier development: Implications for lifelong gut health in pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2017; 3:313-321. [PMID: 29767141 PMCID: PMC5941262 DOI: 10.1016/j.aninu.2017.06.003] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 12/14/2022]
Abstract
The gastrointestinal (GI) barrier serves a critical role in survival and overall health of animals and humans. Several layers of barrier defense mechanisms are provided by the epithelial, immune and enteric nervous systems. Together they act in concert to control normal gut functions (e.g., digestion, absorption, secretion, immunity, etc.) whereas at the same time provide a barrier from the hostile conditions in the luminal environment. Breakdown of these critical GI functions is a central pathophysiological mechanism in the most serious GI disorders in pigs. This review will focus on the development and functional properties of the GI barrier in pigs and how common early life production stressors, such as weaning, can alter immediate and long-term barrier function and disease susceptibility. Specific stress-related pathophysiological mechanisms responsible for driving GI barrier dysfunction induced by weaning and the implications to animal health and performance will be discussed.
Collapse
Affiliation(s)
- Adam J. Moeser
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Corresponding author.
| | - Calvin S. Pohl
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Mrigendra Rajput
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
89
|
Espinosa-Medina I, Jevans B, Boismoreau F, Chettouh Z, Enomoto H, Müller T, Birchmeier C, Burns AJ, Brunet JF. Dual origin of enteric neurons in vagal Schwann cell precursors and the sympathetic neural crest. Proc Natl Acad Sci U S A 2017; 114:11980-11985. [PMID: 29078343 PMCID: PMC5692562 DOI: 10.1073/pnas.1710308114] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Most of the enteric nervous system derives from the "vagal" neural crest, lying at the level of somites 1-7, which invades the digestive tract rostro-caudally from the foregut to the hindgut. Little is known about the initial phase of this colonization, which brings enteric precursors into the foregut. Here we show that the "vagal crest" subsumes two populations of enteric precursors with contrasted origins, initial modes of migration, and destinations. Crest cells adjacent to somites 1 and 2 produce Schwann cell precursors that colonize the vagus nerve, which in turn guides them into the esophagus and stomach. Crest cells adjacent to somites 3-7 belong to the crest streams contributing to sympathetic chains: they migrate ventrally, seed the sympathetic chains, and colonize the entire digestive tract thence. Accordingly, enteric ganglia, like sympathetic ones, are atrophic when deprived of signaling through the tyrosine kinase receptor ErbB3, while half of the esophageal ganglia require, like parasympathetic ones, the nerve-associated form of the ErbB3 ligand, Neuregulin-1. These dependencies might bear relevance to Hirschsprung disease, with which alleles of Neuregulin-1 are associated.
Collapse
Affiliation(s)
- Isabel Espinosa-Medina
- Institut de Biologie de l'École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Ben Jevans
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, University College London Great Ormond Street Institute of Child Health, WC1N 1EH London, United Kingdom
| | - Franck Boismoreau
- Institut de Biologie de l'École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Zoubida Chettouh
- Institut de Biologie de l'École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Hideki Enomoto
- Laboratory for Neural Differentiation and Regeneration, Graduate School of Medicine, Kobe University, 650-0017 Kobe City, Japan
| | - Thomas Müller
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz-Association, 13125 Berlin, Germany
| | - Carmen Birchmeier
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz-Association, 13125 Berlin, Germany
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, University College London Great Ormond Street Institute of Child Health, WC1N 1EH London, United Kingdom
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - Jean-François Brunet
- Institut de Biologie de l'École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France;
| |
Collapse
|
90
|
Perea D, Guiu J, Hudry B, Konstantinidou C, Milona A, Hadjieconomou D, Carroll T, Hoyer N, Natarajan D, Kallijärvi J, Walker JA, Soba P, Thapar N, Burns AJ, Jensen KB, Miguel-Aliaga I. Ret receptor tyrosine kinase sustains proliferation and tissue maturation in intestinal epithelia. EMBO J 2017; 36:3029-3045. [PMID: 28899900 PMCID: PMC5641678 DOI: 10.15252/embj.201696247] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 01/25/2023] Open
Abstract
Expression of the Ret receptor tyrosine kinase is a defining feature of enteric neurons. Its importance is underscored by the effects of its mutation in Hirschsprung disease, leading to absence of gut innervation and severe gastrointestinal symptoms. We report a new and physiologically significant site of Ret expression in the intestine: the intestinal epithelium. Experiments in Drosophila indicate that Ret is expressed both by enteric neurons and adult intestinal epithelial progenitors, which require Ret to sustain their proliferation. Mechanistically, Ret is engaged in a positive feedback loop with Wnt/Wingless signalling, modulated by Src and Fak kinases. We find that Ret is also expressed by the developing intestinal epithelium of mice, where its expression is maintained into the adult stage in a subset of enteroendocrine/enterochromaffin cells. Mouse organoid experiments point to an intrinsic role for Ret in promoting epithelial maturation and regulating Wnt signalling. Our findings reveal evolutionary conservation of the positive Ret/Wnt signalling feedback in both developmental and homeostatic contexts. They also suggest an epithelial contribution to Ret loss‐of‐function disorders such as Hirschsprung disease.
Collapse
Affiliation(s)
- Daniel Perea
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Jordi Guiu
- BRIC-Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N, Denmark
| | - Bruno Hudry
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | | | - Alexandra Milona
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Dafni Hadjieconomou
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Thomas Carroll
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Nina Hoyer
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf (UKE), University of Hamburg, Hamburg, Germany
| | - Dipa Natarajan
- Stem Cells and Regenerative Medicine, UCL Institute of Child Health, London, UK
| | - Jukka Kallijärvi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - James A Walker
- Center for Human Genetic Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Soba
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf (UKE), University of Hamburg, Hamburg, Germany
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Institute of Child Health, London, UK
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Institute of Child Health, London, UK
| | - Kim B Jensen
- BRIC-Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N, Denmark.,The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| |
Collapse
|
91
|
Schreiber D, Marx L, Felix S, Clasohm J, Weyland M, Schäfer M, Klotz M, Lilischkis R, Erkel G, Schäfer KH. Anti-inflammatory Effects of Fungal Metabolites in Mouse Intestine as Revealed by In vitro Models. Front Physiol 2017; 8:566. [PMID: 28824460 PMCID: PMC5545603 DOI: 10.3389/fphys.2017.00566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/20/2017] [Indexed: 01/01/2023] Open
Abstract
Inflammatory bowel diseases (IBD), which include Crohn's disease and ulcerative colitis, are chronic inflammatory disorders that can affect the whole gastrointestinal tract or the colonic mucosal layer. Current therapies aiming to suppress the exaggerated immune response in IBD largely rely on compounds with non-satisfying effects or side-effects. Therefore, new therapeutical options are needed. In the present study, we investigated the anti-inflammatory effects of the fungal metabolites, galiellalactone, and dehydrocurvularin in both an in vitro intestinal inflammation model, as well as in isolated myenteric plexus and enterocyte cells. Administration of a pro-inflammatory cytokine mix through the mesenteric artery of intestinal segments caused an up-regulation of inflammatory marker genes. Treatment of the murine intestinal segments with galiellalactone or dehydrocurvularin by application through the mesenteric artery significantly prevented the expression of pro-inflammatory marker genes on the mRNA and the protein level. Comparable to the results in the perfused intestine model, treatment of primary enteric nervous system (ENS) cells from the murine intestine with the fungal compounds reduced expression of cytokines such as IL-6, TNF-α, IL-1β, and inflammatory enzymes such as COX-2 and iNOS on mRNA and protein levels. Similar anti-inflammatory effects of the fungal metabolites were observed in the human colorectal adenocarcinoma cell line DLD-1 after stimulation with IFN-γ (10 ng/ml), TNF-α (10 ng/ml), and IL-1β (5 ng/ml). Our results show that the mesenterially perfused intestine model provides a reliable tool for the screening of new therapeutics with limited amounts of test compounds. Furthermore, we could characterize the anti-inflammatory effects of two novel active compounds, galiellalactone, and dehydrocurvularin which are interesting candidates for studies with chronic animal models of IBD.
Collapse
Affiliation(s)
- Dominik Schreiber
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany.,Department of Biotechnology, Technical University of KaiserslauternKaiserslautern, Germany
| | - Lisa Marx
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Silke Felix
- Department of Biotechnology, Technical University of KaiserslauternKaiserslautern, Germany
| | - Jasmin Clasohm
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Maximilian Weyland
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Maximilian Schäfer
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Markus Klotz
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Rainer Lilischkis
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Gerhard Erkel
- Department of Biotechnology, Technical University of KaiserslauternKaiserslautern, Germany
| | - Karl-Herbert Schäfer
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany.,Pediatric Surgery, University Hospital MannheimMannheim, Germany
| |
Collapse
|
92
|
Roy-Carson S, Natukunda K, Chou HC, Pal N, Farris C, Schneider SQ, Kuhlman JA. Defining the transcriptomic landscape of the developing enteric nervous system and its cellular environment. BMC Genomics 2017; 18:290. [PMID: 28403821 PMCID: PMC5389105 DOI: 10.1186/s12864-017-3653-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Motility and the coordination of moving food through the gastrointestinal tract rely on a complex network of neurons known as the enteric nervous system (ENS). Despite its critical function, many of the molecular mechanisms that direct the development of the ENS and the elaboration of neural network connections remain unknown. The goal of this study was to transcriptionally identify molecular pathways and candidate genes that drive specification, differentiation and the neural circuitry of specific neural progenitors, the phox2b expressing ENS cell lineage, during normal enteric nervous system development. Because ENS development is tightly linked to its environment, the transcriptional landscape of the cellular environment of the intestine was also analyzed. RESULTS Thousands of zebrafish intestines were manually dissected from a transgenic line expressing green fluorescent protein under the phox2b regulatory elements [Tg(phox2b:EGFP) w37 ]. Fluorescence-activated cell sorting was used to separate GFP-positive phox2b expressing ENS progenitor and derivatives from GFP-negative intestinal cells. RNA-seq was performed to obtain accurate, reproducible transcriptional profiles and the unbiased detection of low level transcripts. Analysis revealed genes and pathways that may function in ENS cell determination, genes that may be identifiers of different ENS subtypes, and genes that define the non-neural cellular microenvironment of the ENS. Differential expression analysis between the two cell populations revealed the expected neuronal nature of the phox2b expressing lineage including the enrichment for genes required for neurogenesis and synaptogenesis, and identified many novel genes not previously associated with ENS development. Pathway analysis pointed to a high level of G-protein coupled pathway activation, and identified novel roles for candidate pathways such as the Nogo/Reticulon axon guidance pathway in ENS development. CONCLUSION We report the comprehensive gene expression profiles of a lineage-specific population of enteric progenitors, their derivatives, and their microenvironment during normal enteric nervous system development. Our results confirm previously implicated genes and pathways required for ENS development, and also identify scores of novel candidate genes and pathways. Thus, our dataset suggests various potential mechanisms that drive ENS development facilitating characterization and discovery of novel therapeutic strategies to improve gastrointestinal disorders.
Collapse
Affiliation(s)
- Sweta Roy-Carson
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Kevin Natukunda
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Hsien-Chao Chou
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA.,Present Address: National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Narinder Pal
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA.,Present address: North Central Regional Plant Introduction Station, 1305 State Ave, Ames, IA, 50014, USA
| | - Caitlin Farris
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA.,Present address: Pioneer Hi-Bred International, Johnson, IA, 50131, USA
| | - Stephan Q Schneider
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Julie A Kuhlman
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA. .,642 Science II, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
93
|
Allen AP, Dinan TG, Clarke G, Cryan JF. A psychology of the human brain-gut-microbiome axis. SOCIAL AND PERSONALITY PSYCHOLOGY COMPASS 2017; 11:e12309. [PMID: 28804508 PMCID: PMC5530613 DOI: 10.1111/spc3.12309] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/25/2017] [Accepted: 02/11/2017] [Indexed: 12/15/2022]
Abstract
In recent years, we have seen increasing research within neuroscience and biopsychology on the interactions between the brain, the gastrointestinal tract, the bacteria within the gastrointestinal tract, and the bidirectional relationship between these systems: the brain-gut-microbiome axis. Although research has demonstrated that the gut microbiota can impact upon cognition and a variety of stress-related behaviours, including those relevant to anxiety and depression, we still do not know how this occurs. A deeper understanding of how psychological development as well as social and cultural factors impact upon the brain-gut-microbiome axis will contextualise the role of the axis in humans and inform psychological interventions that improve health within the brain-gut-microbiome axis. Interventions ostensibly aimed at ameliorating disorders in one part of the brain-gut-microbiome axis (e.g., psychotherapy for depression) may nonetheless impact upon other parts of the axis (e.g., microbiome composition and function), and functional gastrointestinal disorders such as irritable bowel syndrome represent a disorder of the axis, rather than an isolated problem either of psychology or of gastrointestinal function. The discipline of psychology needs to be cognisant of these interactions and can help to inform the future research agenda in this emerging field of research. In this review, we outline the role psychology has to play in understanding the brain-gut-microbiome axis, with a focus on human psychology and the use of research in laboratory animals to model human psychology.
Collapse
Affiliation(s)
- Andrew P. Allen
- Dept Psychiatry & Behavioural Neuroscience/APC Microbiome InstituteUniversity College CorkCorkIreland
| | - Timothy G. Dinan
- Dept Psychiatry & Behavioural Neuroscience/APC Microbiome InstituteUniversity College CorkCorkIreland
| | - Gerard Clarke
- Dept Psychiatry & Behavioural Neuroscience/APC Microbiome InstituteUniversity College CorkCorkIreland
| | - John F. Cryan
- Dept Anatomy & Neuroscience/APC Microbiome InstituteUniversity College CorkCorkIreland
| |
Collapse
|
94
|
Stamp LA. Cell therapy for GI motility disorders: comparison of cell sources and proposed steps for treating Hirschsprung disease. Am J Physiol Gastrointest Liver Physiol 2017; 312:G348-G354. [PMID: 28209600 DOI: 10.1152/ajpgi.00018.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/02/2017] [Accepted: 02/08/2017] [Indexed: 01/31/2023]
Abstract
Cell therapeutic approaches to treat a range of congenital and degenerative neuropathies are under intense investigation. There have been recent significant advancements in the development of cell therapy to treat disorders of the enteric nervous system (ENS), enteric neuropathies. These advances include the efficient generation of enteric neural progenitors from pluripotent stem cells and the rescue of a Hirschsprung disease model mouse following their transplantation into the bowel. Furthermore, a recent study provides evidence of functional innervation of the bowel muscle by neurons derived from transplanted ENS-derived neural progenitors. This mini-review discusses these recent findings, compares endogenous ENS-derived progenitors and pluripotent stem cell-derived progenitors as a cell source for therapy, and proposes the key steps for cell therapy to treat Hirschsprung disease.
Collapse
Affiliation(s)
- Lincon A Stamp
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia
| |
Collapse
|
95
|
Moore SW. Advances in understanding functional variations in the Hirschsprung disease spectrum (variant Hirschsprung disease). Pediatr Surg Int 2017; 33:285-298. [PMID: 27988850 DOI: 10.1007/s00383-016-4038-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 12/11/2022]
Abstract
Hirschsprung disease (HSCR) is a fairly well understood congenital, genetically based functional obstruction due to the congenital absence of ganglion cells in the distal bowel. However, although over 90% of Hirschsprung cases conform to the normally accepted histological diagnostic criteria, it has become increasingly clear that in addition to HSCR, there is a group of functional disturbances relating to a number of other congenital neurodysplastic conditions causing some degree of gastrointestinal tract malfunction. Although these represent a variety of possibly separate conditions of the enteric nervous system, this spectrum it would appear to be also influenced by similar developmental processes. The term "variant Hirschsprung" is commonly used to describe these conditions, but ganglion cells are mostly present if abnormal in number and distribution. These conditions are a problem group being amongst the most difficult to diagnose and treat with possible practical and legal consequences. The problem appears to be possibly one of definition which has proven difficult in the relative paucity of normal values, especially when correlated to age and gestation. It is the purpose of this paper to review the current position on these conditions and to explore possible shared common pathogenetic and genetic mechanisms. This article explores those conditions where a similar pathogenetic mechanisms to HSCR can be demonstrated (e.g. hypoganglionosis) as well as other neural features, which appear to represent separate conditions possibly linked to certain syndromes.
Collapse
Affiliation(s)
- S W Moore
- Division of Paediatric Surgery, Faculty of Medicine, University of Stellenbosch, P.O. Box 19063, Tygerberg, 7505, South Africa.
| |
Collapse
|
96
|
Le Berre‐Scoul C, Chevalier J, Oleynikova E, Cossais F, Talon S, Neunlist M, Boudin H. A novel enteric neuron-glia coculture system reveals the role of glia in neuronal development. J Physiol 2017; 595:583-598. [PMID: 27436013 PMCID: PMC5233665 DOI: 10.1113/jp271989] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 07/07/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Unlike astrocytes in the brain, the potential role of enteric glial cells (EGCs) in the formation of the enteric neuronal circuit is currently unknown. To examine the role of EGCs in the formation of the neuronal network, we developed a novel neuron-enriched culture model from embryonic rat intestine grown in indirect coculture with EGCs. We found that EGCs shape axonal complexity and synapse density in enteric neurons, through purinergic- and glial cell line-derived neurotrophic factor-dependent pathways. Using a novel and valuable culture model to study enteric neuron-glia interactions, our study identified EGCs as a key cellular actor regulating neuronal network maturation. ABSTRACT In the nervous system, the formation of neuronal circuitry results from a complex and coordinated action of intrinsic and extrinsic factors. In the CNS, extrinsic mediators derived from astrocytes have been shown to play a key role in neuronal maturation, including dendritic shaping, axon guidance and synaptogenesis. In the enteric nervous system (ENS), the potential role of enteric glial cells (EGCs) in the maturation of developing enteric neuronal circuit is currently unknown. A major obstacle in addressing this question is the difficulty in obtaining a valuable experimental model in which enteric neurons could be isolated and maintained without EGCs. We adapted a cell culture method previously developed for CNS neurons to establish a neuron-enriched primary culture from embryonic rat intestine which was cultured in indirect coculture with EGCs. We demonstrated that enteric neurons grown in such conditions showed several structural, phenotypic and functional hallmarks of proper development and maturation. However, when neurons were grown without EGCs, the complexity of the axonal arbour and the density of synapses were markedly reduced, suggesting that glial-derived factors contribute strongly to the formation of the neuronal circuitry. We found that these effects played by EGCs were mediated in part through purinergic P2Y1 receptor- and glial cell line-derived neurotrophic factor-dependent pathways. Using a novel and valuable culture model to study enteric neuron-glia interactions, our study identified EGCs as a key cellular actor required for neuronal network maturation.
Collapse
|
97
|
McKeown SJ, Mohsenipour M, Bergner AJ, Young HM, Stamp LA. Exposure to GDNF Enhances the Ability of Enteric Neural Progenitors to Generate an Enteric Nervous System. Stem Cell Reports 2017; 8:476-488. [PMID: 28089669 PMCID: PMC5312076 DOI: 10.1016/j.stemcr.2016.12.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 12/13/2016] [Accepted: 12/14/2016] [Indexed: 12/22/2022] Open
Abstract
Cell therapy is a promising approach to generate an enteric nervous system (ENS) and treat enteric neuropathies. However, for translation to the clinic, it is highly likely that enteric neural progenitors will require manipulation prior to transplantation to enhance their ability to migrate and generate an ENS. In this study, we examine the effects of exposure to several factors on the ability of ENS progenitors, grown as enteric neurospheres, to migrate and generate an ENS. Exposure to glial-cell-line-derived neurotrophic factor (GDNF) resulted in a 14-fold increase in neurosphere volume and a 12-fold increase in cell number. Following co-culture with embryonic gut or transplantation into the colon of postnatal mice in vivo, cells derived from GDNF-treated neurospheres showed a 2-fold increase in the distance migrated compared with controls. Our data show that the ability of enteric neurospheres to generate an ENS can be enhanced by exposure to appropriate factors. Enteric neurospheres are likely to require manipulation for clinical applications Exposure to GDNF increased the size and cell number in enteric neurospheres GDNF-treated neurospheres showed enhanced migration after transplantation in vivo Manipulation of enteric neurospheres can enhance the generation of enteric neurons
Collapse
Affiliation(s)
- Sonja J McKeown
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia; Cancer Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| | - Mitra Mohsenipour
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia
| | - Annette J Bergner
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia
| | - Heather M Young
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia
| | - Lincon A Stamp
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
98
|
Nagy N, Goldstein AM. Enteric nervous system development: A crest cell's journey from neural tube to colon. Semin Cell Dev Biol 2017; 66:94-106. [PMID: 28087321 DOI: 10.1016/j.semcdb.2017.01.006] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 12/31/2022]
Abstract
The enteric nervous system (ENS) is comprised of a network of neurons and glial cells that are responsible for coordinating many aspects of gastrointestinal (GI) function. These cells arise from the neural crest, migrate to the gut, and then continue their journey to colonize the entire length of the GI tract. Our understanding of the molecular and cellular events that regulate these processes has advanced significantly over the past several decades, in large part facilitated by the use of rodents, avians, and zebrafish as model systems to dissect the signals and pathways involved. These studies have highlighted the highly dynamic nature of ENS development and the importance of carefully balancing migration, proliferation, and differentiation of enteric neural crest-derived cells (ENCCs). Proliferation, in particular, is critically important as it drives cell density and speed of migration, both of which are important for ensuring complete colonization of the gut. However, proliferation must be tempered by differentiation among cells that have reached their final destination and are ready to send axonal extensions, connect to effector cells, and begin to produce neurotransmitters or other signals. Abnormalities in the normal processes guiding ENCC development can lead to failure of ENS formation, as occurs in Hirschsprung disease, in which the distal intestine remains aganglionic. This review summarizes our current understanding of the factors involved in early development of the ENS and discusses areas in need of further investigation.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Center for Neurointestinal Health, Massachusetts General Hospital, Boston, MA, United States; Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Center for Neurointestinal Health, Massachusetts General Hospital, Boston, MA, United States.
| |
Collapse
|
99
|
Cooper JE, Natarajan D, McCann CJ, Choudhury S, Godwin H, Burns AJ, Thapar N. In vivo transplantation of fetal human gut-derived enteric neural crest cells. Neurogastroenterol Motil 2017; 29:e12900. [PMID: 27380932 PMCID: PMC5215633 DOI: 10.1111/nmo.12900] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022]
Abstract
The prospect of using neural cell replacement for the treatment of severe enteric neuropathies has seen significant progress in the last decade. The ability to harvest and transplant enteric neural crest cells (ENCCs) that functionally integrate within recipient intestine has recently been confirmed by in vivo murine studies. Although similar cells can be harvested from human fetal and postnatal gut, no studies have as yet verified their functional viability upon in vivo transplantation. We sought to determine whether ENCCs harvested from human fetal bowel are capable of engraftment and functional integration within recipient intestine following in vivo transplantation into postnatal murine colon. Enteric neural crest cells selected and harvested from fetal human gut using the neurotrophin receptor p75NTR were lentivirally labeled with either GFP or calcium-sensitive GCaMP and transplanted into the hindgut of Rag2- /γc- /C5- -immunodeficient mice at postnatal day 21. Transplanted intestines were assessed immunohistochemically for engraftment and differentiation of donor cells. Functional viability and integration with host neuromusculature was assessed using calcium imaging. Transplanted human fetal gut-derived ENCC showed engraftment within the recipient postnatal colon in 8/15 mice (53.3%). At 4 weeks posttransplantation, donor cells had spread from the site of transplantation and extended projections over distances of 1.2 ± 0.6 mm (n = 5), and differentiated into enteric nervous system (ENS) appropriate neurons and glia. These cells formed branching networks located with the myenteric plexus. Calcium transients (change in intensity F/F0 = 1.25 ± 0.03; 15 cells) were recorded in transplanted cells upon stimulation of the recipient endogenous ENS demonstrating their viability and establishment of functional connections.
Collapse
Affiliation(s)
- J. E. Cooper
- Stem Cells and Regenerative MedicineUCL Institute of Child HealthLondonUK
| | - D. Natarajan
- Stem Cells and Regenerative MedicineUCL Institute of Child HealthLondonUK
| | - C. J. McCann
- Stem Cells and Regenerative MedicineUCL Institute of Child HealthLondonUK
| | - S. Choudhury
- Stem Cells and Regenerative MedicineUCL Institute of Child HealthLondonUK
| | - H. Godwin
- Department of GastroenterologyGreat Ormond Street Hospital NHS Foundation TrustLondonUK
| | - A. J. Burns
- Stem Cells and Regenerative MedicineUCL Institute of Child HealthLondonUK,Department of Clinical GeneticsErasmus MCRotterdamThe Netherlands
| | - N. Thapar
- Stem Cells and Regenerative MedicineUCL Institute of Child HealthLondonUK,Department of GastroenterologyGreat Ormond Street Hospital NHS Foundation TrustLondonUK
| |
Collapse
|
100
|
Van Den Abeele J, Rubbens J, Brouwers J, Augustijns P. The dynamic gastric environment and its impact on drug and formulation behaviour. Eur J Pharm Sci 2017; 96:207-231. [DOI: 10.1016/j.ejps.2016.08.060] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 02/08/2023]
|