1
|
Bu L, He L, Wang X, Du G, Wu R, Liu W. Proteomic Analysis Provides a New Sight Into the CRABP1 Expression in the Pathogenesis of Hirschsprung Disease. Biochem Genet 2024:10.1007/s10528-024-10913-3. [PMID: 39298027 DOI: 10.1007/s10528-024-10913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024]
Abstract
Hirschsprung's disease (HSCR) is the most common developmental disorder of the enteric nervous system and its etiology and pathogenesis remain largely unknown. This study aims to identify the differential proteomic patterns linked to the occurrence and development of Hirschsprung disease in colonic tissues. Biopsies were obtained from the aganglionic colon in human HSCR and the corresponding ganglionic colon segments for direct quantitative determination of the data-independent acquisition (DIA) followed by bioinformatics analysis. The differentially expressed main proteins were confirmed by Western blot and immunostaining. A total of 5832 proteins were identified in human colon tissues. Among them, 97 differentially expressed proteins (DEP) with fold change (FC) > 1.2 were screened, including 18 upregulated proteins and 79 downregulated proteins, and GO and KEGG enrichment analyses were performed on differential proteins. By comparing down-regulated proteins with highly connected protein nodes in the PPI network with those related to intracellular metabolic processes in the above analysis, we identified cellular retinoic acid binding protein 1(CRABP1). Its expression was verified in the aganglionic part of the colon by western blotting in an expanded sample set (P = 0.0031). The immunostaining results revealed that CRABP1 was highly expressed in the myenteric plexus ganglion in ganglionic colons compared to aganglionic segments (P = 0.0004). This study demonstrated the down-regulation of CRABP1 in the aganglionic hindgut of HSCR, which could provide potential markers or promising new candidate actors for the pathogenesis of HSCR.
Collapse
Affiliation(s)
- Lingyun Bu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, China
| | - Lingxiao He
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, China
| | - Xiaoqing Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, China
| | - Guoqiang Du
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, China
| | - Rongde Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, China
| | - Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, China.
| |
Collapse
|
2
|
Mainali BB, Yoo JJ, Ladd MR. Tissue engineering and regenerative medicine approaches in colorectal surgery. Ann Coloproctol 2024; 40:336-349. [PMID: 39228197 PMCID: PMC11375227 DOI: 10.3393/ac.2024.00437.0062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Tissue engineering and regenerative medicine (TERM) is an emerging field that has provided new therapeutic opportunities by delivering innovative solutions. The development of nontraditional therapies for previously unsolvable diseases and conditions has brought hope and excitement to countless individuals globally. Many regenerative medicine therapies have been developed and delivered to patients clinically. The technology platforms developed in regenerative medicine have been expanded to various medical areas; however, their applications in colorectal surgery remain limited. Applying TERM technologies to engineer biological tissue and organ substitutes may address the current therapeutic challenges and overcome some complications in colorectal surgery, such as inflammatory bowel diseases, short bowel syndrome, and diseases of motility and neuromuscular function. This review provides a comprehensive overview of TERM applications in colorectal surgery, highlighting the current state of the art, including preclinical and clinical studies, current challenges, and future perspectives. This article synthesizes the latest findings, providing a valuable resource for clinicians and researchers aiming to integrate TERM into colorectal surgical practice.
Collapse
Affiliation(s)
- Bigyan B Mainali
- Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
- Department of Biomedical Engineering, Wake Forest University, Winston-Salem, NC, USA
| | - Mitchell R Ladd
- Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
- Department of Biomedical Engineering, Wake Forest University, Winston-Salem, NC, USA
| |
Collapse
|
3
|
Dershowitz LB, Kaltschmidt JA. Enteric Nervous System Striped Patterning and Disease: Unexplored Pathophysiology. Cell Mol Gastroenterol Hepatol 2024; 18:101332. [PMID: 38479486 PMCID: PMC11176954 DOI: 10.1016/j.jcmgh.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
The enteric nervous system (ENS) controls gastrointestinal (GI) motility, and defects in ENS development underlie pediatric GI motility disorders. In disorders such as Hirschsprung's disease (HSCR), pediatric intestinal pseudo-obstruction (PIPO), and intestinal neuronal dysplasia type B (INDB), ENS structure is altered with noted decreased neuronal density in HSCR and reports of increased neuronal density in PIPO and INDB. The developmental origin of these structural deficits is not fully understood. Here, we review the current understanding of ENS development and pediatric GI motility disorders incorporating new data on ENS structure. In particular, emerging evidence demonstrates that enteric neurons are patterned into circumferential stripes along the longitudinal axis of the intestine during mouse and human development. This novel understanding of ENS structure proposes new questions about the pathophysiology of pediatric GI motility disorders. If the ENS is organized into stripes, could the observed changes in enteric neuron density in HSCR, PIPO, and INDB represent differences in the distribution of enteric neuronal stripes? We review mechanisms of striped patterning from other biological systems and propose how defects in striped ENS patterning could explain structural deficits observed in pediatric GI motility disorders.
Collapse
Affiliation(s)
- Lori B Dershowitz
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California; Wu Tsai Neurosciences Institute, Stanford University, Stanford, California
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California; Wu Tsai Neurosciences Institute, Stanford University, Stanford, California.
| |
Collapse
|
4
|
Dershowitz LB, Li L, Pasca AM, Kaltschmidt JA. Anatomical and functional maturation of the mid-gestation human enteric nervous system. Nat Commun 2023; 14:2680. [PMID: 37160892 PMCID: PMC10170115 DOI: 10.1038/s41467-023-38293-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/14/2023] [Indexed: 05/11/2023] Open
Abstract
Immature gastrointestinal motility impedes preterm infant survival. The enteric nervous system controls gastrointestinal motility, yet it is unknown when the human enteric nervous system matures enough to carry out vital functions. Here we demonstrate that the second trimester human fetal enteric nervous system takes on a striped organization akin to the embryonic mouse. Further, we perform ex vivo functional assays of human fetal tissue and find that human fetal gastrointestinal motility matures in a similar progression to embryonic mouse gastrointestinal motility. Together, this provides critical knowledge, which facilitates comparisons with common animal models to advance translational disease investigations and testing of pharmacological agents to enhance gastrointestinal motility in prematurity.
Collapse
Affiliation(s)
- Lori B Dershowitz
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305, USA
| | - Li Li
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Anca M Pasca
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
5
|
Deguchi K, Zambaiti E, De Coppi P. Regenerative medicine: current research and perspective in pediatric surgery. Pediatr Surg Int 2023; 39:167. [PMID: 37014468 PMCID: PMC10073065 DOI: 10.1007/s00383-023-05438-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/05/2023]
Abstract
The field of regenerative medicine, encompassing several disciplines including stem cell biology and tissue engineering, continues to advance with the accumulating research on cell manipulation technologies, gene therapy and new materials. Recent progress in preclinical and clinical studies may transcend the boundaries of regenerative medicine from laboratory research towards clinical reality. However, for the ultimate goal to construct bioengineered transplantable organs, a number of issues still need to be addressed. In particular, engineering of elaborate tissues and organs requires a fine combination of different relevant aspects; not only the repopulation of multiple cell phenotypes in an appropriate distribution but also the adjustment of the host environmental factors such as vascularisation, innervation and immunomodulation. The aim of this review article is to provide an overview of the recent discoveries and development in stem cells and tissue engineering, which are inseparably interconnected. The current status of research on tissue stem cells and bioengineering, and the possibilities for application in specific organs relevant to paediatric surgery have been specifically focused and outlined.
Collapse
Affiliation(s)
- Koichi Deguchi
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Elisa Zambaiti
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK
- UOC Chirurgia Pediatrica, Ospedale Infantile Regina Margherita, Turin, Italy
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK.
- NIHR BRC SNAPS Great Ormond Street Hospitals, London, UK.
- Stem Cells and Regenerative Medicine Section, Faculty of Population Health Sciences, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
6
|
The Potential Role of Microorganisms on Enteric Nervous System Development and Disease. Biomolecules 2023; 13:biom13030447. [PMID: 36979382 PMCID: PMC10046024 DOI: 10.3390/biom13030447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/14/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The enteric nervous system (ENS), the inherent nervous system of the gastrointestinal (GI) tract is a vast nervous system that controls key GI functions, including motility. It functions at a critical interface between the gut luminal contents, including the diverse population of microorganisms deemed the microbiota, as well as the autonomic and central nervous systems. Critical development of this axis of interaction, a key determinant of human health and disease, appears to occur most significantly during early life and childhood, from the pre-natal through to the post-natal period. These factors that enable the ENS to function as a master regulator also make it vulnerable to damage and, in turn, a number of GI motility disorders. Increasing attention is now being paid to the potential of disruption of the microbiota and pathogenic microorganisms in the potential aetiopathogeneis of GI motility disorders in children. This article explores the evidence regarding the relationship between the development and integrity of the ENS and the potential for such factors, notably dysbiosis and pathogenic bacteria, viruses and parasites, to impact upon them in early life.
Collapse
|
7
|
Caillaud M, Le Dréan ME, De-Guilhem-de-Lataillade A, Le Berre-Scoul C, Montnach J, Nedellec S, Loussouarn G, Paillé V, Neunlist M, Boudin H. A functional network of highly pure enteric neurons in a dish. Front Neurosci 2023; 16:1062253. [PMID: 36685225 PMCID: PMC9853279 DOI: 10.3389/fnins.2022.1062253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/02/2022] [Indexed: 01/09/2023] Open
Abstract
The enteric nervous system (ENS) is the intrinsic nervous system that innervates the entire digestive tract and regulates major digestive functions. Recent evidence has shown that functions of the ENS critically rely on enteric neuronal connectivity; however, experimental models to decipher the underlying mechanisms are limited. Compared to the central nervous system, for which pure neuronal cultures have been developed for decades and are recognized as a reference in the field of neuroscience, an equivalent model for enteric neurons is lacking. In this study, we developed a novel model of highly pure rat embryonic enteric neurons with dense and functional synaptic networks. The methodology is simple and relatively fast. We characterized enteric neurons using immunohistochemical, morphological, and electrophysiological approaches. In particular, we demonstrated the applicability of this culture model to multi-electrode array technology as a new approach for monitoring enteric neuronal network activity. This in vitro model of highly pure enteric neurons represents a valuable new tool for better understanding the mechanisms involved in the establishment and maintenance of enteric neuron synaptic connectivity and functional networks.
Collapse
Affiliation(s)
- Martial Caillaud
- Nantes Université, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France,*Correspondence: Martial Caillaud,
| | - Morgane E. Le Dréan
- Nantes Université, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | | | - Catherine Le Berre-Scoul
- Nantes Université, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Jérôme Montnach
- Nantes Université, CNRS, INSERM, L’institut du Thorax, Nantes, France
| | - Steven Nedellec
- Nantes Université, CHU Nantes, CNRS, INSERM, BioCore, US16, SFR Bonamy, Nantes, France
| | - Gildas Loussouarn
- Nantes Université, CNRS, INSERM, L’institut du Thorax, Nantes, France
| | - Vincent Paillé
- Nantes Université, INRAE, IMAD, CRNH-O, UMR 1280, PhAN, Nantes, France
| | - Michel Neunlist
- Nantes Université, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Hélène Boudin
- Nantes Université, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| |
Collapse
|
8
|
Hamnett R, Dershowitz LB, Sampathkumar V, Wang Z, Gomez-Frittelli J, De Andrade V, Kasthuri N, Druckmann S, Kaltschmidt JA. Regional cytoarchitecture of the adult and developing mouse enteric nervous system. Curr Biol 2022; 32:4483-4492.e5. [PMID: 36070775 PMCID: PMC9613618 DOI: 10.1016/j.cub.2022.08.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 07/19/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022]
Abstract
The organization and cellular composition of tissues are key determinants of their biological function. In the mammalian gastrointestinal (GI) tract, the enteric nervous system (ENS) intercalates between muscular and epithelial layers of the gut wall and can control GI function independent of central nervous system (CNS) input.1 As in the CNS, distinct regions of the GI tract are highly specialized and support diverse functions, yet the regional and spatial organization of the ENS remains poorly characterized.2 Cellular arrangements,3,4 circuit connectivity patterns,5,6 and diverse cell types7-9 are known to underpin ENS functional complexity and GI function, but enteric neurons are most typically described only as a uniform meshwork of interconnected ganglia. Here, we present a bird's eye view of the mouse ENS, describing its previously underappreciated cytoarchitecture and regional variation. We visually and computationally demonstrate that enteric neurons are organized in circumferential neuronal stripes. This organization emerges gradually during the perinatal period, with neuronal stripe formation in the small intestine (SI) preceding that in the colon. The width of neuronal stripes varies throughout the length of the GI tract, and distinct neuronal subtypes differentially populate specific regions of the GI tract, with stark contrasts between SI and colon as well as within subregions of each. This characterization provides a blueprint for future understanding of region-specific GI function and identifying ENS structural correlates of diverse GI disorders.
Collapse
Affiliation(s)
- Ryan Hamnett
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Lori B Dershowitz
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Vandana Sampathkumar
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA; Biosciences Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Ziyue Wang
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Julieta Gomez-Frittelli
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Vincent De Andrade
- Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Narayanan Kasthuri
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA; Biosciences Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Shaul Druckmann
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
9
|
Słupecka-Ziemilska M, Pierzynowski SG, Szczurek P, Pierzynowska K, Wychowański P, Seklecka B, Koperski M, Starzyńska A, Szkopek D, Donaldson J, Andrzejewski K, Woliński J. Milk Formula Enriched with Sodium Butyrate Influences Small Intestine Contractility in Neonatal Pigs. Nutrients 2022; 14:nu14204301. [PMID: 36296985 PMCID: PMC9608939 DOI: 10.3390/nu14204301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Butyrate, a by-product of gut bacteria fermentation as well as the digestion of fat in mother’s milk, exerts a wide spectrum of beneficial effects in the gastrointestinal tissues. The present study aimed to determine the effects of sodium butyrate on small intestine contractility in neonatal piglets. Piglets were fed milk formula alone (group C) or milk formula supplemented with sodium butyrate (group B). After a 7-day treatment period, isometric recordings of whole-thickness segments of the duodenum and middle jejunum were obtained by electric field stimulation under the influence of increasing doses of Ach (acetylocholine) in the presence of TTX (tetrodotoxin) and atropine. Moreover, structural properties of the intestinal wall were assessed, together with the expression of cholinergic and muscarinic receptors (M1 and M2). In both intestinal segments (duodenum and middle jejunum), EFS (electric field stimulation) impulses resulted in increased contractility and amplitude of contractions in group B compared to group C. Additionally, exposure to dietary butyrate led to a significant increase in tunica muscularis thickness in the duodenum, while mitotic and apoptotic indices were increased in the middle jejunum. The expression of M1 and M2 receptors in the middle jejunum was significantly higher after butyrate treatment. The results indicate increased cholinergic signaling and small intestinal growth and renewal in response to feeding with milk formula enriched with sodium butyrate in neonatal piglets.
Collapse
Affiliation(s)
- Monika Słupecka-Ziemilska
- Department of Human Epigenetics, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warszawa, Poland
| | - Stefan Grzegorz Pierzynowski
- Department of Medical Biology, Institute of Rural Health, 20-090 Lublin, Poland
- SGP + Group, 231 32 Trelleborg, Sweden
- Department of Biology, Lund University, Sölvegatan 35, 223 62 Lund, Sweden
| | - Paulina Szczurek
- Department of Animal Nutrition and Feed Sciences, National Research Institute of Animal Production, 32-083 Balice, Poland
| | - Kateryna Pierzynowska
- Department of Biology, Lund University, Sölvegatan 35, 223 62 Lund, Sweden
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
- Correspondence: (K.P.); (J.W.)
| | - Piotr Wychowański
- Division of Oral Surgery and Implantology, Department of Head and Neck, Institute of Clinical Dentistry, Oral Surgery and Implantology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS-Universita Cattolica del Sacro Coure, 00168 Rome, Italy
| | | | - Maciej Koperski
- Department of Human Epigenetics, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warszawa, Poland
| | - Anna Starzyńska
- Departament of Oral Surgery, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland
| | - Dominika Szkopek
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
| | - Janine Donaldson
- SGP + Group, 231 32 Trelleborg, Sweden
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Krzysztof Andrzejewski
- Department of Orthopedics and Traumatology, Veteran’s Memorial Hospital, Medical University of Łódź, 90-549 Łódź, Poland
| | - Jarosław Woliński
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
- Correspondence: (K.P.); (J.W.)
| |
Collapse
|
10
|
Sicard P, Falco A, Faure S, Thireau J, Lindsey SE, Chauvet N, de Santa Barbara P. High-resolution ultrasound and speckle tracking: a non-invasive approach to assess in vivo gastrointestinal motility during development. Development 2022; 149:dev200625. [PMID: 35912573 PMCID: PMC10655954 DOI: 10.1242/dev.200625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2023]
Abstract
Gastrointestinal motor activity has been extensively studied in adults; however, only few studies have investigated fetal motor skills. It is unknown when the gastrointestinal tract starts to contract during the embryonic period and how this function evolves during development. Here, we adapted a non-invasive high-resolution echography technique combined with speckle tracking analysis to examine the gastrointestinal tract motor activity dynamics during chick embryo development. We provided the first recordings of fetal gastrointestinal motility in living embryos without anesthesia. We found that, although gastrointestinal contractions appear very early during development, they become synchronized only at the end of the fetal period. To validate this approach, we used various pharmacological inhibitors and BAPX1 gene overexpression in vivo. We found that the enteric nervous system determines the onset of the synchronized contractions in the stomach. Moreover, alteration of smooth muscle fiber organization led to an impairment of this functional activity. Altogether, our findings show that non-invasive high-resolution echography and speckle tracking analysis allows visualization and quantification of gastrointestinal motility during development and highlight the progressive acquisition of functional and coordinated gastrointestinal motility before birth.
Collapse
Affiliation(s)
- Pierre Sicard
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
- IPAM, Biocampus Montpellier, CNRS, INSERM, University of Montpellier, 34295 Montpellier, France
| | - Amandine Falco
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Sandrine Faure
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Jérome Thireau
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Stéphanie E. Lindsey
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
- Department of Mechanical and Aerospace Engineering, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Norbert Chauvet
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | | |
Collapse
|
11
|
Theodorou CM, Jackson JE, Stokes SC, Pivetti CD, Kumar P, Paxton ZJ, Matsukuma KE, Yamashiro KJ, Reynaga L, Hyllen AA, de Lorimier AJ, Hassan M, Wang A, Farmer DL, Saadai P. Early investigations into improving bowel and bladder function in fetal ovine myelomeningocele repair. J Pediatr Surg 2022; 57:941-948. [PMID: 35093254 PMCID: PMC10372624 DOI: 10.1016/j.jpedsurg.2021.12.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Fetal myelomeningocele (MMC) repair improves lower extremity motor function. We have previously demonstrated that augmentation of fetal MMC repair with placental mesenchymal stromal cells (PMSCs) seeded on extracellular matrix (PMSC-ECM) further improves motor function in the ovine model. However, little progress has been made in improving bowel and bladder function, with many patients suffering from neurogenic bowel and bladder. We hypothesized that fetal MMC repair with PMSC-ECM would also improve bowel and bladder function. METHODS MMC defects were surgically created in twelve ovine fetuses at median gestational age (GA) 73 days, followed by defect repair at GA101 with PMSC-ECM. Fetuses were delivered at GA141. Primary bladder function outcomes were voiding posture and void volumes. Primary bowel function outcome was anorectal manometry findings including resting anal pressure and presence of rectoanal inhibitory reflex (RAIR). Secondary outcomes were anorectal and bladder detrusor muscle thickness. PMSC-ECM lambs were compared to normal lambs (n = 3). RESULTS Eighty percent of PMSC-ECM lambs displayed normal voiding posture compared to 100% of normal lambs (p = 1). Void volumes were similar (PMSC-ECM 6.1 ml/kg vs. normal 8.8 ml/kg, p = 0.4). Resting mean anal pressures were similar between cohorts (27.0 mmHg PMSC-ECM vs. normal 23.5 mmHg, p = 0.57). RAIR was present in 3/5 PMSC-ECM lambs that underwent anorectal manometry and all normal lambs (p = 0.46). Thicknesses of anal sphincter complex, rectal wall muscles, and bladder detrusor muscles were similar between cohorts. CONCLUSION Ovine fetal MMC repair augmented with PMSC-ECM results in near-normal bowel and bladder function. Further work is needed to evaluate these outcomes in human patients.
Collapse
Affiliation(s)
- Christina M Theodorou
- Department of Surgery, Division of Pediatric General, Thoracic, and Fetal Surgery. University of California Davis Medical Center. Sacramento, CA, United States.
| | - Jordan E Jackson
- Department of Surgery, Division of Pediatric General, Thoracic, and Fetal Surgery. University of California Davis Medical Center. Sacramento, CA, United States
| | - Sarah C Stokes
- Department of Surgery, Division of Pediatric General, Thoracic, and Fetal Surgery. University of California Davis Medical Center. Sacramento, CA, United States
| | - Christopher D Pivetti
- Surgical Bioengineering Laboratory, University of California Davis Medical Center. Sacramento, CA, United States
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, University of California Davis Medical Center. Sacramento, CA, United States
| | - Zachary J Paxton
- Surgical Bioengineering Laboratory, University of California Davis Medical Center. Sacramento, CA, United States
| | - Karen E Matsukuma
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, United States
| | - Kaeli J Yamashiro
- Department of Surgery, Division of Pediatric General, Thoracic, and Fetal Surgery. University of California Davis Medical Center. Sacramento, CA, United States
| | - Lizette Reynaga
- Surgical Bioengineering Laboratory, University of California Davis Medical Center. Sacramento, CA, United States
| | - Alicia A Hyllen
- Surgical Bioengineering Laboratory, University of California Davis Medical Center. Sacramento, CA, United States
| | - Arthur J de Lorimier
- Department of Pediatrics, Division of Gastroenterology. University of California Davis Medical Center. Sacramento, CA, United States
| | - Maheen Hassan
- Department of Pediatrics, Division of Gastroenterology. University of California Davis Medical Center. Sacramento, CA, United States
| | - Aijun Wang
- Surgical Bioengineering Laboratory, University of California Davis Medical Center. Sacramento, CA, United States
| | - Diana L Farmer
- Department of Surgery, Division of Pediatric General, Thoracic, and Fetal Surgery. University of California Davis Medical Center. Sacramento, CA, United States
| | - Payam Saadai
- Department of Surgery, Division of Pediatric General, Thoracic, and Fetal Surgery. University of California Davis Medical Center. Sacramento, CA, United States
| |
Collapse
|
12
|
Chandramowlishwaran P, Raja S, Maheshwari A, Srinivasan S. Enteric Nervous System in Neonatal Necrotizing Enterocolitis. Curr Pediatr Rev 2022; 18:9-24. [PMID: 34503418 DOI: 10.2174/1573396317666210908162745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/26/2021] [Accepted: 06/08/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The pathophysiology of necrotizing enterocolitis (NEC) is not clear, but increasing information suggests that the risk and severity of NEC may be influenced by abnormalities in the enteric nervous system (ENS). OBJECTIVE The purpose of this review was to scope and examine the research related to ENS-associated abnormalities that have either been identified in NEC or have been noted in other inflammatory bowel disorders (IBDs) with histopathological abnormalities similar to NEC. The aim was to summarize the research findings, identify research gaps in existing literature, and disseminate them to key knowledge end-users to collaborate and address the same in future studies. METHODS Articles that met the objectives of the study were identified through an extensive literature search in the databases PubMed, EMBASE, and Scopus. RESULTS The sources identified through the literature search revealed that: (1) ENS may be involved in NEC development and post-NEC complications, (2) NEC development is associated with changes in the ENS, and (3) NEC-associated changes could be modulated by the ENS. CONCLUSION The findings from this review identify the enteric nervous as a target in the development and progression of NEC. Thus, factors that can protect the ENS can potentially prevent and treat NEC and post-NEC complications. This review serves to summarize the existing literature and highlights a need for further research on the involvement of ENS in NEC.
Collapse
Affiliation(s)
- Pavithra Chandramowlishwaran
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| | - Shreya Raja
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| | - Akhil Maheshwari
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Shanthi Srinivasan
- Department of Medicine, Emory University School of Medicine, Decatur, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
13
|
Lionarons JM, Hoogland G, Slegers RJ, Steinbusch H, Claessen SMH, Vles JSH. Dystrophin in the Neonatal and Adult Rat Intestine. Life (Basel) 2021; 11:life11111155. [PMID: 34833031 PMCID: PMC8622973 DOI: 10.3390/life11111155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Gastrointestinal (GI) complaints are frequently noted in aging dystrophinopathy patients, yet their underlying molecular mechanisms are largely unknown. As dystrophin protein isoform 71 (Dp71) is particularly implicated in the development of smooth muscle cells, we evaluated its distribution in the neonatal and adult rat intestine in this study. Methods: Dp71 expression levels were assessed in the proximal (duodenum, jejunum and ileum) and distal (caecum, colon and rectum) intestine by Western blotting and qPCR. In addition, the cellular distribution of total Dp was evaluated in the duodenum and colon by immunohistochemical colocalization studies with alpha-smooth muscle actin (aSMA), Hu RNA binding proteins C and D (HuC/HuD) for neurons and vimentin (VIM) for interstitial cells. Results: In neonatal and adult rats, the distal intestine expressed 2.5 times more Dp71 protein than the proximal part (p < 0.01). This regional difference was not observed in Dp71 mRNA. During both stages, Dp-immunoreactivity was predominant in the muscularis propria, where it co-localized with aSMA and HuC/HuD. Conclusions: In neonatal and adult rats, Dp71 was expressed highest in the distal intestine. Together with the observation that Dp may be expressed by myenteric neurons, this warrants a paradigm shift in the treatment of GI comorbidities.
Collapse
Affiliation(s)
- Judith M. Lionarons
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
- Correspondence: (J.M.L.); (G.H.); Tel.: +31-(0)43-3875058 (J.M.L.); +31-(0)43-3881024 (G.H.)
| | - Govert Hoogland
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
- Department of Neurosurgery, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Correspondence: (J.M.L.); (G.H.); Tel.: +31-(0)43-3875058 (J.M.L.); +31-(0)43-3881024 (G.H.)
| | - Rutger J. Slegers
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| | - Hellen Steinbusch
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| | - Sandra M. H. Claessen
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| | - Johan S. H. Vles
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.J.S.); (H.S.); (S.M.H.C.); (J.S.H.V.)
| |
Collapse
|
14
|
Fujii K, Nakajo K, Egashira Y, Yamamoto Y, Kitada K, Taniguchi K, Kawai M, Tomiyama H, Kawakami K, Uchiyama K, Ono F. Gastrointestinal Neurons Expressing HCN4 Regulate Retrograde Peristalsis. Cell Rep 2021; 30:2879-2888.e3. [PMID: 32130893 DOI: 10.1016/j.celrep.2020.02.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/30/2019] [Accepted: 02/06/2020] [Indexed: 12/31/2022] Open
Abstract
Peristalsis is indispensable for physiological function of the gut. The enteric nervous system (ENS) plays an important role in regulating peristalsis. While the neural network regulating anterograde peristalsis, which migrates from the oral end to the anal end, is characterized to some extent, retrograde peristalsis remains unresolved with regards to its neural regulation. Using forward genetics in zebrafish, we reveal that a population of neurons expressing a hyperpolarization-activated nucleotide-gated channel HCN4 specifically regulates retrograde peristalsis. When HCN4 channels are blocked by an HCN channel inhibitor or morpholinos blocking the protein expression, retrograde peristalsis is specifically attenuated. Conversely, when HCN4(+) neurons expressing channelrhodopsin are activated by illumination, retrograde peristalsis is enhanced while anterograde peristalsis remains unchanged. We propose that HCN4(+) neurons in the ENS forward activating signals toward the oral end and simultaneously stimulate local circuits regulating the circular muscle.
Collapse
Affiliation(s)
- Kensuke Fujii
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Koichi Nakajo
- Department of Physiology, Osaka Medical College, Takatsuki, Japan; Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | | | | | - Kazuya Kitada
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Kohei Taniguchi
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Masaru Kawai
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Hideki Tomiyama
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan
| | - Kazuhisa Uchiyama
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Fumihito Ono
- Department of Physiology, Osaka Medical College, Takatsuki, Japan.
| |
Collapse
|
15
|
Li Y, Toothaker JM, Ben-Simon S, Ozeri L, Schweitzer R, McCourt BT, McCourt CC, Werner L, Snapper SB, Shouval DS, Khatib S, Koren O, Agnihorti S, Tseng G, Konnikova L. In utero human intestine harbors unique metabolome, including bacterial metabolites. JCI Insight 2020; 5:138751. [PMID: 33001863 PMCID: PMC7710283 DOI: 10.1172/jci.insight.138751] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
Symbiotic microbial colonization through the establishment of the intestinal microbiome is critical to many intestinal functions, including nutrient metabolism, intestinal barrier integrity, and immune regulation. Recent studies suggest that education of intestinal immunity may be ongoing in utero. However, the drivers of this process are unknown. The microbiome and its byproducts are one potential source. Whether a fetal intestinal microbiome exists is controversial, and whether microbially derived metabolites are present in utero is unknown. Here, we aimed to determine whether bacterial DNA and microbially derived metabolites can be detected in second trimester human intestinal samples. Although we were unable to amplify bacterial DNA from fetal intestines, we report a fetal metabolomic intestinal profile with an abundance of bacterially derived and host-derived metabolites commonly produced in response to microbiota. Though we did not directly assess their source and function, we hypothesize that these microbial-associated metabolites either come from the maternal microbiome and are vertically transmitted to the fetus to prime the fetal immune system and prepare the gastrointestinal tract for postnatal microbial encounters or are produced locally by bacteria that were below our detection threshold.
Collapse
Affiliation(s)
| | - Jessica M. Toothaker
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shira Ben-Simon
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Lital Ozeri
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Ron Schweitzer
- Analytical Chemistry Laboratory, Tel-Hai College, Upper Galilee, Israel
| | - Blake T. McCourt
- Department of Pediatrics, Yale University, New Haven, Connecticut, USA
| | - Collin C. McCourt
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lael Werner
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Scott B. Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Dror S. Shouval
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Soliman Khatib
- Analytical Chemistry Laboratory, Tel-Hai College, Upper Galilee, Israel
- Department of Natural Compounds and Analytical Chemistry, Migal Galilee Research Institute, Kiryat Shmona, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | | | | | - Liza Konnikova
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, Yale University, New Haven, Connecticut, USA
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Reproductive Sciences and
- Program in Human and Translational Immunology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
16
|
Heymans C, de Lange IH, Lenaerts K, Kessels LCGA, Hadfoune M, Rademakers G, Melotte V, Boesmans W, Kramer BW, Jobe AH, Saito M, Kemp MW, van Gemert WG, Wolfs TGAM. Chorioamnionitis induces enteric nervous system injury: effects of timing and inflammation in the ovine fetus. Mol Med 2020; 26:82. [PMID: 32883198 PMCID: PMC7469100 DOI: 10.1186/s10020-020-00206-x] [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] [Received: 02/05/2020] [Accepted: 07/28/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chorioamnionitis, inflammation of the chorion and amnion, which often results from intrauterine infection, is associated with premature birth and contributes to significant neonatal morbidity and mortality, including necrotizing enterocolitis (NEC). Recently, we have shown that chronic chorioamnionitis is associated with significant structural enteric nervous system (ENS) abnormalities that may predispose to later NEC development. Understanding time point specific effects of an intra-amniotic (IA) infection on the ENS is important for further understanding the pathophysiological processes and for finding a window for optimal therapeutic strategies for an individual patient. The aim of this study was therefore to gain insight in the longitudinal effects of intrauterine LPS exposure (ranging from 5 h to 15 days before premature delivery) on the intestinal mucosa, submucosa, and ENS in fetal lambs by use of a well-established translational ovine chorioamnionitis model. METHODS We used an ovine chorioamnionitis model to assess outcomes of the fetal ileal mucosa, submucosa and ENS following IA exposure to one dose of 10 mg LPS for 5, 12 or 24 h or 2, 4, 8 or 15 days. RESULTS Four days of IA LPS exposure causes a decreased PGP9.5- and S100β-positive surface area in the myenteric plexus along with submucosal and mucosal intestinal inflammation that coincided with systemic inflammation. These changes were preceded by a glial cell reaction with early systemic and local gut inflammation. ENS changes and inflammation recovered 15 days after the IA LPS exposure. CONCLUSIONS The pattern of mucosal and submucosal inflammation, and ENS alterations in the fetus changed over time following IA LPS exposure. Although ENS damage seemed to recover after prolonged IA LPS exposure, additional postnatal inflammatory exposure, which a premature is likely to encounter, may further harm the ENS and influence functional outcome. In this context, 4 to 8 days of IA LPS exposure may form a period of increased ENS vulnerability and a potential window for optimal therapeutic strategies.
Collapse
Affiliation(s)
- C Heymans
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - I H de Lange
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.,Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, P.O. Box 616, Universiteitssingel 50, 6200, MD, Maastricht, The Netherlands
| | - K Lenaerts
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - L C G A Kessels
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, P.O. Box 616, Universiteitssingel 50, 6200, MD, Maastricht, The Netherlands
| | - M Hadfoune
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - G Rademakers
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, the Netherlands
| | - V Melotte
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, the Netherlands
| | - W Boesmans
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, the Netherlands.,Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - B W Kramer
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, P.O. Box 616, Universiteitssingel 50, 6200, MD, Maastricht, The Netherlands.,Neonatology, Department of Pediatrics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - A H Jobe
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Australia.,Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - M Saito
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - M W Kemp
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Australia.,School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia
| | - W G van Gemert
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands.,Pediatric surgery, Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Surgery, University Hospital Aachen, Aachen, Germany
| | - T G A M Wolfs
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, P.O. Box 616, Universiteitssingel 50, 6200, MD, Maastricht, The Netherlands. .,Department of Biomedical Engineering (BMT), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
17
|
Heymans C, de Lange IH, Hütten MC, Lenaerts K, de Ruijter NJE, Kessels LCGA, Rademakers G, Melotte V, Boesmans W, Saito M, Usuda H, Stock SJ, Spiller OB, Beeton ML, Payne MS, Kramer BW, Newnham JP, Jobe AH, Kemp MW, van Gemert WG, Wolfs TGAM. Chronic Intra-Uterine Ureaplasma parvum Infection Induces Injury of the Enteric Nervous System in Ovine Fetuses. Front Immunol 2020; 11:189. [PMID: 32256485 PMCID: PMC7089942 DOI: 10.3389/fimmu.2020.00189] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/24/2020] [Indexed: 01/18/2023] Open
Abstract
Background: Chorioamnionitis, inflammation of the fetal membranes during pregnancy, is often caused by intra-amniotic (IA) infection with single or multiple microbes. Chorioamnionitis can be either acute or chronic and is associated with adverse postnatal outcomes of the intestine, including necrotizing enterocolitis (NEC). Neonates with NEC have structural and functional damage to the intestinal mucosa and the enteric nervous system (ENS), with loss of enteric neurons and glial cells. Yet, the impact of acute, chronic, or repetitive antenatal inflammatory stimuli on the development of the intestinal mucosa and ENS has not been studied. The aim of this study was therefore to investigate the effect of acute, chronic, and repetitive microbial exposure on the intestinal mucosa, submucosa and ENS in premature lambs. Materials and Methods: A sheep model of pregnancy was used in which the ileal mucosa, submucosa, and ENS were assessed following IA exposure to lipopolysaccharide (LPS) for 2 or 7 days (acute), Ureaplasma parvum (UP) for 42 days (chronic), or repetitive microbial exposure (42 days UP with 2 or 7 days LPS). Results: IA LPS exposure for 7 days or IA UP exposure for 42 days caused intestinal injury and inflammation in the mucosal and submucosal layers of the gut. Repetitive microbial exposure did not further aggravate injury of the terminal ileum. Chronic IA UP exposure caused significant structural ENS alterations characterized by loss of PGP9.5 and S100β immunoreactivity, whereas these changes were not found after re-exposure of chronic UP-exposed fetuses to LPS for 2 or 7 days. Conclusion: The in utero loss of PGP9.5 and S100β immunoreactivity following chronic UP exposure corresponds with intestinal changes in neonates with NEC and may therefore form a novel mechanistic explanation for the association of chorioamnionitis and NEC.
Collapse
Affiliation(s)
- Cathelijne Heymans
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Ilse H de Lange
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands.,Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Matthias C Hütten
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands.,Neonatology, Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands.,Neonatology, Department of Pediatrics, University Hospital Aachen, Aachen, Germany.,Neonatology, Department of Pediatrics, University Children's Hospital Würzburg, Würzburg, Germany
| | - Kaatje Lenaerts
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Nadine J E de Ruijter
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Lilian C G A Kessels
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Glenn Rademakers
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, Netherlands
| | - Veerle Melotte
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, Netherlands
| | - Werend Boesmans
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, Netherlands.,Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Masatoshi Saito
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Sarah J Stock
- Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Owen B Spiller
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael L. Beeton
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Matthew S Payne
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia
| | - Boris W Kramer
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands.,Neonatology, Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands
| | - John P Newnham
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan.,School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
| | - Wim G van Gemert
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands.,Pediatric Surgery, Department of Surgery, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Surgery, University Hospital Aachen, Aachen, Germany
| | - Tim G A M Wolfs
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands.,Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
18
|
Li YW, Yu YJ, Fei F, Zheng MY, Zhang SW. High-resolution colonic manometry and its clinical application in patients with colonic dysmotility: A review. World J Clin Cases 2019; 7:2675-2686. [PMID: 31616684 PMCID: PMC6789394 DOI: 10.12998/wjcc.v7.i18.2675] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/26/2019] [Accepted: 08/20/2019] [Indexed: 02/05/2023] Open
Abstract
The detailed process and mechanism of colonic motility are still unclear, and colonic motility disorders are associated with numerous clinical diseases. Colonic manometry is considered to the most direct means of evaluating colonic peristalsis. Colonic manometry has been studied for more than 30 years; however, the long duration of the examination, high risk of catheterization, huge amount of real-time data, strict catheter sterilization, and high cost of disposable equipment restrict its wide application in clinical practice. Recently, high-resolution colonic manometry (HRCM) has rapidly developed into a major technique for obtaining more effective information involved in the physiology and/or pathophysiology of colonic contractile activity in colonic dysmotility patients. This review focuses on colonic motility, manometry, operation, and motor patterns, and the clinical application of HRCM. Furthermore, the limitations, future directions, and potential usefulness of HRCM in the evaluation of clinical treatment effects are also discussed.
Collapse
Affiliation(s)
- Yu-Wei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300071, China
| | - Yong-Jun Yu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300071, China
| | - Fei Fei
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300071, China
- Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Min-Ying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300071, China
| | - Shi-Wu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300071, China
- Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
19
|
Kulkarni S, Ganz J, Bayrer J, Becker L, Bogunovic M, Rao M. Advances in Enteric Neurobiology: The "Brain" in the Gut in Health and Disease. J Neurosci 2018; 38:9346-9354. [PMID: 30381426 PMCID: PMC6209840 DOI: 10.1523/jneurosci.1663-18.2018] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/20/2018] [Accepted: 09/22/2018] [Indexed: 12/14/2022] Open
Abstract
The enteric nervous system (ENS) is a large, complex division of the peripheral nervous system that regulates many digestive, immune, hormonal, and metabolic functions. Recent advances have elucidated the dynamic nature of the mature ENS, as well as the complex, bidirectional interactions among enteric neurons, glia, and the many other cell types that are important for mediating gut behaviors. Here, we provide an overview of ENS development and maintenance, and focus on the latest insights gained from the use of novel model systems and live-imaging techniques. We discuss major advances in the understanding of enteric glia, and the functional interactions among enteric neurons, glia, and enteroendocrine cells, a large class of sensory epithelial cells. We conclude by highlighting recent work on muscularis macrophages, a group of immune cells that closely interact with the ENS in the gut wall, and the importance of neurological-immune system communication in digestive health and disease.
Collapse
Affiliation(s)
- Subhash Kulkarni
- Department of Medicine, The John Hopkins University School of Medicine, Baltimore, Maryland 21205,
| | - Julia Ganz
- Department of Integrative Biology, Michigan State University, East Lansing, Michigan 48824
| | - James Bayrer
- Department of Pediatrics, University of California, San Francisco, San Francisco, California 94143
| | - Laren Becker
- Department of Medicine, Stanford University, Stanford, California 94305
| | - Milena Bogunovic
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania 17033, and
| | - Meenakshi Rao
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
20
|
Parsons SP, Huizinga JD. Phase waves and trigger waves: emergent properties of oscillating and excitable networks in the gut. J Physiol 2018; 596:4819-4829. [PMID: 30055053 PMCID: PMC6187044 DOI: 10.1113/jp273425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 07/18/2018] [Indexed: 12/30/2022] Open
Abstract
The gut is enmeshed by a number of cellular networks, but there is only a limited understanding of how these networks generate the complex patterns of activity that drive gut contractile functions. Here we review two fundamental types of cell behaviour, excitable and oscillating, and the patterns that networks of such cells generate, trigger waves and phase waves, respectively. We use both the language of biophysics and the theory of nonlinear dynamics to define these behaviours and understand how they generate patterns. Based on this we look for evidence of trigger and phase waves in the gut, including some of our recent work on the small intestine.
Collapse
Affiliation(s)
- Sean P. Parsons
- Farncombe Family Digestive Health Research InstituteDepartment of MedicineMcMaster UniversityHamiltonONCanada
| | - Jan D. Huizinga
- Farncombe Family Digestive Health Research InstituteDepartment of MedicineMcMaster UniversityHamiltonONCanada
| |
Collapse
|
21
|
Abstract
Enteric duplications have been described throughout the entire gastrointestinal tract. The usual perinatal presentation is an abdominal mass. Duplications associated with the foregut have associated respiratory symptoms, whereas duplications in the midgut and hindgut can present with obstructive symptoms, perforation, nausea, emesis, hemorrhage, or be asymptomatic, and identified as an incidental finding. These are differentiated from other cystic lesions by the presence of a normal gastrointestinal mucosal epithelium. Enteric duplications are located on the mesenteric side of the native structures and are often singular with tubular or cystic characteristics. Management of enteric duplications often requires operative intervention with preservation of the native blood supply and intestine. These procedures are usually very well tolerated with low morbidity.
Collapse
Affiliation(s)
- Paul M Jeziorczak
- Division of Pediatric Surgery, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri
| | - Brad W Warner
- Division of Pediatric Surgery, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri
| |
Collapse
|
22
|
Nam SH. A novel and simple method using a transanal intestinal long tube for protecting intestinal anastomosis and decompressing the small bowel. Ann Surg Treat Res 2017; 93:137-142. [PMID: 28932729 PMCID: PMC5597537 DOI: 10.4174/astr.2017.93.3.137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/21/2017] [Accepted: 03/30/2017] [Indexed: 11/30/2022] Open
Abstract
PURPOSE I introduce the use of transanal intestinal long tube (TILT) using nasogastric tube. TILT passes from anus to the anastomosis, helping to decompress a dilated bowel loop. METHODS TILT procedure was limited to those patients predicting a severe luminal size discrepancy after intestinal anastomosis, and who had postoperative prolonged ileus. We retrospectively reviewed the medical records of 10 infants (7 male an 3 female patients) who were treated using the TILT procedure between 2012 and 2016. RESULTS Median gestational age was 27+5 weeks and birth weight was 940 g. The first operation was done at a median of 4.5 days after birth due to necrotizing enterocolitis perforation (4 cases), isolated intestinal perforation (3 cases), meconium related ileus (1 case), congenital ileal volvulus (1 case), and ileal atresia (1 case). Nine cases of ileostomy closure were planned at a median of 130.5 days with a body weight of 3,060 g. For the ileal atresia case, TILT procedure without additional small bowel resection was performed to treat postoperative prolonged ileus. Nine out of ten were well functioned and defecation via anus was observed in a median of 4.5 days. Milk feeding began at a median of 6 days and the long intestinal tube was removed in a median of 14.5 days. CONCLUSION I suggested that TILT procedure could be a noninvasive operative option, predicting of size mismatched anastomosis causing prolonged ileus. Passive drainage of proximal intestinal contents might be helpful for decompress endoluminal pressure during the time of anastomosis healing with bowel movement recovery.
Collapse
Affiliation(s)
- So Hyun Nam
- Division of Pediatric Surgery, Department of General Surgery, Dong-A University College of Medicine, Dong-A University Hospital, Busan, Korea
| |
Collapse
|
23
|
Schlieve CR, Fowler KL, Thornton M, Huang S, Hajjali I, Hou X, Grubbs B, Spence JR, Grikscheit TC. Neural Crest Cell Implantation Restores Enteric Nervous System Function and Alters the Gastrointestinal Transcriptome in Human Tissue-Engineered Small Intestine. Stem Cell Reports 2017; 9:883-896. [PMID: 28803915 PMCID: PMC5599241 DOI: 10.1016/j.stemcr.2017.07.017] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 07/20/2017] [Accepted: 07/21/2017] [Indexed: 01/20/2023] Open
Abstract
Acquired or congenital disruption in enteric nervous system (ENS) development or function can lead to significant mechanical dysmotility. ENS restoration through cellular transplantation may provide a cure for enteric neuropathies. We have previously generated human pluripotent stem cell (hPSC)-derived tissue-engineered small intestine (TESI) from human intestinal organoids (HIOs). However, HIO-TESI fails to develop an ENS. The purpose of our study is to restore ENS components derived exclusively from hPSCs in HIO-TESI. hPSC-derived enteric neural crest cell (ENCC) supplementation of HIO-TESI establishes submucosal and myenteric ganglia, repopulates various subclasses of neurons, and restores neuroepithelial connections and neuron-dependent contractility and relaxation in ENCC-HIO-TESI. RNA sequencing identified differentially expressed genes involved in neurogenesis, gliogenesis, gastrointestinal tract development, and differentiated epithelial cell types when ENS elements are restored during in vivo development of HIO-TESI. Our findings validate an effective approach to restoring hPSC-derived ENS components in HIO-TESI and may implicate their potential for the treatment of enteric neuropathies.
Collapse
Affiliation(s)
- Christopher R Schlieve
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, 4650 W. Sunset Boulevard, MS#100, Los Angeles, CA 90027, USA; Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Kathryn L Fowler
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, 4650 W. Sunset Boulevard, MS#100, Los Angeles, CA 90027, USA
| | - Matthew Thornton
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sha Huang
- 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; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Ibrahim Hajjali
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, 4650 W. Sunset Boulevard, MS#100, Los Angeles, CA 90027, USA
| | - Xiaogang Hou
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, 4650 W. Sunset Boulevard, MS#100, Los Angeles, CA 90027, USA
| | - Brendan Grubbs
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jason R Spence
- 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; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Tracy C Grikscheit
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, 4650 W. Sunset Boulevard, MS#100, Los Angeles, CA 90027, USA; Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA.
| |
Collapse
|
24
|
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: 3.3] [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
|
25
|
Extracerebral Dysfunction in Animal Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:159-187. [PMID: 28551756 DOI: 10.1007/978-3-319-52498-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Genetic factors might be largely responsible for the development of autism spectrum disorder (ASD) that alone or in combination with specific environmental risk factors trigger the pathology. Multiple mutations identified in ASD patients that impair synaptic function in the central nervous system are well studied in animal models. How these mutations might interact with other risk factors is not fully understood though. Additionally, how systems outside of the brain are altered in the context of ASD is an emerging area of research. Extracerebral influences on the physiology could begin in utero and contribute to changes in the brain and in the development of other body systems and further lead to epigenetic changes. Therefore, multiple recent studies have aimed at elucidating the role of gene-environment interactions in ASD. Here we provide an overview on the extracerebral systems that might play an important associative role in ASD and review evidence regarding the potential roles of inflammation, trace metals, metabolism, genetic susceptibility, enteric nervous system function and the microbiota of the gastrointestinal (GI) tract on the development of endophenotypes in animal models of ASD. By influencing environmental conditions, it might be possible to reduce or limit the severity of ASD pathology.
Collapse
|
26
|
Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nat Med 2016; 23:49-59. [PMID: 27869805 DOI: 10.1038/nm.4233] [Citation(s) in RCA: 446] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023]
Abstract
The enteric nervous system (ENS) of the gastrointestinal tract controls many diverse functions, including motility and epithelial permeability. Perturbations in ENS development or function are common, yet there is no human model for studying ENS-intestinal biology and disease. We used a tissue-engineering approach with embryonic and induced pluripotent stem cells (PSCs) to generate human intestinal tissue containing a functional ENS. We recapitulated normal intestinal ENS development by combining human-PSC-derived neural crest cells (NCCs) and developing human intestinal organoids (HIOs). NCCs recombined with HIOs in vitro migrated into the mesenchyme, differentiated into neurons and glial cells and showed neuronal activity, as measured by rhythmic waves of calcium transients. ENS-containing HIOs grown in vivo formed neuroglial structures similar to a myenteric and submucosal plexus, had functional interstitial cells of Cajal and had an electromechanical coupling that regulated waves of propagating contraction. Finally, we used this system to investigate the cellular and molecular basis for Hirschsprung's disease caused by a mutation in the gene PHOX2B. This is, to the best of our knowledge, the first demonstration of human-PSC-derived intestinal tissue with a functional ENS and how this system can be used to study motility disorders of the human gastrointestinal tract.
Collapse
|
27
|
von Sochaczewski CO, Wenke K, Grieve A, Westgarth-Taylor C, Loveland JA, Metzger R, Kluth D. Regenerative capacity of the enteric nervous system: is immaturity defining the point of no return? J Surg Res 2016; 209:112-121. [PMID: 28032547 DOI: 10.1016/j.jss.2016.09.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 09/14/2016] [Accepted: 09/21/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND Intestinal obstruction in newborns is associated with intestinal motility disorders after surgery. Alterations in the enteric nervous system (ENS) might cause abnormal peristalsis, which may then result in intestinal motility disorders. We aimed to quantify alterations in the myenteric plexus after a ligation and to test if these alterations were reversible. METHODS Small intestines of chicken embryos were ligated in ovo at embryonic day (ED) 11 for either 4 d (ED 11-15) or 8 d (ED 11-19). Both treated groups and control group were sacrificed and intestinal segments examined by means of both light and electron microscopy. RESULTS The number of proximal myenteric ganglia increased (ED 19, 30.7 ± 3.16 versus 23.1 ± 2.03; P < 0.001) in the 8-d ligature group but had values similar to the control group in the 4-d ligature group. The size distribution was skewed toward small ganglia in the 8-d ligature group (ED 19, 83.71 ± 11.60% versus 3.88 ± 4.74% in the control group; P < 0.001) but comparable with the control group in the 4-d ligature group. Subcellular alterations in the 4-d ligature group were reversible. CONCLUSIONS The pathologic alterations in the ENS were fully reversible in the 4-d ligature group. This reversibility might be linked to the degree of immaturity of the ENS.
Collapse
Affiliation(s)
| | - Katharina Wenke
- Department of Pediatric Surgery, University Hospital of Hamburg, Hamburg, Germany
| | - Andrew Grieve
- Department of Pediatric Surgery, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Chris Westgarth-Taylor
- Department of Pediatric Surgery, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Jerome A Loveland
- Department of Pediatric Surgery, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Roman Metzger
- Department of Pediatric and Adolescent Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Dietrich Kluth
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| |
Collapse
|
28
|
Touré AM, Charrier B, Pilon N. Male-specific colon motility dysfunction in the TashT mouse line. Neurogastroenterol Motil 2016; 28:1494-507. [PMID: 27278627 DOI: 10.1111/nmo.12847] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 04/10/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND In Hirschsprung disease (HSCR), the absence of myenteric neural ganglia in the distal bowel prevents motility and thereby causes functional intestinal obstruction. Although surgical resection of the aganglionic segment allows HSCR children to survive this condition, a number of patients still suffer from impaired motility despite having myenteric ganglia in their postoperative distal bowel. Such phenomenon is also observed in patients suffering from other enteric neuropathies and, in both cases, colonic dysmotility is believed to result from abnormalities of myenteric ganglia and/or associated interstitial cells of Cajal (ICC). To better understand this, we used a recently described HSCR mouse model called TashT. METHODS Intestinal motility parameters were assessed and correlated with extent of aganglionosis and with neuronal density in ganglionated regions. The neural composition of the myenteric plexus and the status of ICC networks was also evaluated using immunofluorescence. KEY RESULTS TashT(Tg/Tg) mice display a strong male bias in the severity of both colonic aganglionosis and hypoganglionosis, which are associated with male-specific reduced colonic motility. TashT(Tg/Tg) male mice also exhibit a specific increase in nNos(+) neurons that is restricted to the most distal ganglionated regions. In contrast, Calretinin(+) myenteric neurons, Sox10(+) myenteric glial cells, and cKit(+) ICC are not affected in TashT(Tg/Tg) mice. CONCLUSIONS AND INFERENCES Male-specific impairment of colonic motility in TashT(Tg/Tg) mice is associated with both severe hypoganglionosis and myenteric neuronal imbalance. Considering these parameters in the clinic might be important for the management of postoperative HSCR patients.
Collapse
Affiliation(s)
- A M Touré
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, Faculty of Sciences, University of Quebec at Montreal (UQAM), Montreal, Canada
| | - B Charrier
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, Faculty of Sciences, University of Quebec at Montreal (UQAM), Montreal, Canada
| | - N Pilon
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, Faculty of Sciences, University of Quebec at Montreal (UQAM), Montreal, Canada.
| |
Collapse
|
29
|
Ramalhosa F, Soares-Cunha C, Seixal RM, Sousa N, Carvalho AF. The Impact of Prenatal Exposure to Dexamethasone on Gastrointestinal Function in Rats. PLoS One 2016; 11:e0161750. [PMID: 27584049 PMCID: PMC5008745 DOI: 10.1371/journal.pone.0161750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 08/11/2016] [Indexed: 01/23/2023] Open
Abstract
Antenatal treatment with synthetic glucocorticoids is commonly used in pregnant women at risk of preterm delivery to accelerate tissue maturation. Exposure to glucocorticoids during development has been hypothesized to underlie different functional gastrointestinal (GI) and motility disorders. Herein, we investigated the impact of in utero exposure to synthetic glucocorticoids (iuGC) on GI function of adult rats. Wistar male rats, born from pregnant dams treated with dexamethasone (DEX), were studied at different ages. Length, histologic analysis, proliferation and apoptosis assays, GI transit, permeability and serotonin (5-HT) content of GI tract were measured. iuGC treatment decreased small intestine size and decreased gut transit. However, iuGC had no impact on intestinal permeability. iuGC differentially impacts the structure and function of the GI tract, which leads to long-lasting alterations in the small intestine that may predispose subjects prone to disorders of the GI tract.
Collapse
Affiliation(s)
- Fátima Ramalhosa
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui Miguel Seixal
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Franky Carvalho
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biometrics Associate Laboratory, Braga/Guimarães, Portugal
- General Surgery Department, Hospital of Braga, Braga, Portugal
| |
Collapse
|
30
|
Nikiforou M, Willburger C, de Jong AE, Kloosterboer N, Jellema RK, Ophelders DRMG, Steinbusch HWM, Kramer BW, Wolfs TGAM. Global hypoxia-ischemia induced inflammation and structural changes in the preterm ovine gut which were not ameliorated by mesenchymal stem cell treatment. Mol Med 2016; 22:244-257. [PMID: 27257938 PMCID: PMC5023518 DOI: 10.2119/molmed.2015.00252] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/11/2016] [Indexed: 12/13/2022] Open
Abstract
Perinatal asphyxia, a condition of impaired gas exchange during birth, leads to fetal hypoxia-ischemia (HI) and is associated with postnatal adverse outcomes including intestinal dysmotility and necrotizing enterocolitis (NEC). Evidence from adult animal models of transient, locally-induced intestinal HI has shown that inflammation is essential in HI-induced injury of the gut. Importantly, mesenchymal stem cell (MSC) treatment prevented this HI-induced intestinal damage. We therefore assessed whether fetal global HI induced inflammation, injury and developmental changes in the gut and whether intravenous MSC administration ameliorated these HI-induced adverse intestinal effects. In a preclinical ovine model, fetuses were subjected to umbilical cord occlusion (UCO), with or without MSC treatment, and sacrificed 7 days after UCO. Global HI increased the number of myeloperoxidase positive cells in the mucosa, upregulated mRNA levels of interleukin (IL)-1β and IL-17 in gut tissue and caused T-cell invasion in the intestinal muscle layer. Intestinal inflammation following global HI was associated with increased Ki67+ cells in the muscularis and subsequent muscle hyperplasia. Global HI caused distortion of glial fibrillary acidic protein immunoreactivity in the enteric glial cells and increased synaptophysin and serotonin expression in the myenteric ganglia. Intravenous MSC treatment did not ameliorate these HI-induced adverse intestinal events. Global HI resulted in intestinal inflammation and enteric nervous system abnormalities which are clinically associated with postnatal complications including feeding intolerance, altered gastrointestinal transit and NEC. The intestinal histopathological changes were not prevented by intravenous MSC treatment directly after HI, indicating that alternative treatment regimens for cell-based therapies should be explored.
Collapse
Affiliation(s)
- Maria Nikiforou
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Carolin Willburger
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Anja E de Jong
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nico Kloosterboer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Reint K Jellema
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Daan RMG Ophelders
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Harry WM Steinbusch
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Tim GAM Wolfs
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
31
|
Medland JE, Pohl CS, Edwards LL, Frandsen S, Bagley K, Li Y, Moeser AJ. Early life adversity in piglets induces long-term upregulation of the enteric cholinergic nervous system and heightened, sex-specific secretomotor neuron responses. Neurogastroenterol Motil 2016; 28:1317-29. [PMID: 27134125 PMCID: PMC5002263 DOI: 10.1111/nmo.12828] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/04/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Early life adversity (ELA) is a risk factor for the later-life onset of gastrointestinal (GI) diseases such as irritable bowel syndrome (IBS); however, the mechanisms are poorly understood. Here, we utilized a porcine model of ELA, early weaning stress (EWS), to investigate the influence of ELA on the development and function of the enteric nervous system (ENS). METHODS Female and castrated male (Male-C) piglets were weaned from their sow either at 15 days of age (EWS) or 28 days of age (late weaning control, LWC). At 60 and 170 days of age, ileal mucosa-submucosa preparations were mounted in Ussing chambers and veratridine- and corticotropin releasing factor (CRF)-releasing factor-evoked short circuit current (Isc ) responses were recorded as indices of secretomotor neuron function. Enteric neuron numbers and the expression of select neurotransmitters and their receptors were also measured. KEY RESULTS Compared with LWC pigs, female, but not Male-C EWS, pigs exhibited heightened veratridine-induced Isc responses at 60 and 170 days of age that were inhibited with tetrodotoxin and atropine. Ileum from EWS pigs had higher numbers of enteric neurons that were choline acetyltransferase positive. Markers of increased cholinergic signaling (increased acetylcholinesterase) and downregulated mucosal muscarinic receptor 3 gene expression were also observed in EWS pigs. CONCLUSIONS & INFERENCES This study demonstrated that EWS in pigs induces lasting and sex-specific hypersensitivity of secretomotor neuron function and upregulation of the cholinergic ENS. These findings may represent a mechanistic link between ELA and lifelong susceptibility to GI diseases such as IBS.
Collapse
Affiliation(s)
- Julia E. Medland
- Comparative Biomedical Sciences Program, North Carolina State University, College of Veterinary Medicine, Raleigh, NC 27607, USA
| | - Calvin S. Pohl
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Laura L. Edwards
- Department of Population Health and Pathobiology, North Carolina State University, College of Veterinary Medicine, Raleigh, NC 27607, USA
| | - Shellsea Frandsen
- Department of Population Health and Pathobiology, North Carolina State University, College of Veterinary Medicine, Raleigh, NC 27607, USA
| | - Kristen Bagley
- Department of Population Health and Pathobiology, North Carolina State University, College of Veterinary Medicine, Raleigh, NC 27607, USA
| | - Yihang Li
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Adam J. Moeser
- Gastrointestinal Stress Biology Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI 48824, USA,Neuroscience Program, Michigan State University
| |
Collapse
|
32
|
Enteric nervous system assembly: Functional integration within the developing gut. Dev Biol 2016; 417:168-81. [PMID: 27235816 DOI: 10.1016/j.ydbio.2016.05.030] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/26/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Co-ordinated gastrointestinal function is the result of integrated communication between the enteric nervous system (ENS) and "effector" cells in the gastrointestinal tract. Unlike smooth muscle cells, interstitial cells, and the vast majority of cell types residing in the mucosa, enteric neurons and glia are not generated within the gut. Instead, they arise from neural crest cells that migrate into and colonise the developing gastrointestinal tract. Although they are "later" arrivals into the developing gut, enteric neural crest-derived cells (ENCCs) respond to many of the same secreted signalling molecules as the "resident" epithelial and mesenchymal cells, and several factors that control the development of smooth muscle cells, interstitial cells and epithelial cells also regulate ENCCs. Much progress has been made towards understanding the migration of ENCCs along the gastrointestinal tract and their differentiation into neurons and glia. However, our understanding of how enteric neurons begin to communicate with each other and extend their neurites out of the developing plexus layers to innervate the various cell types lining the concentric layers of the gastrointestinal tract is only beginning. It is critical for postpartum survival that the gastrointestinal tract and its enteric circuitry are sufficiently mature to cope with the influx of nutrients and their absorption that occurs shortly after birth. Subsequently, colonisation of the gut by immune cells and microbiota during postnatal development has an important impact that determines the ultimate outline of the intrinsic neural networks of the gut. In this review, we describe the integrated development of the ENS and its target cells.
Collapse
|
33
|
Foong JPP. Postnatal Development of the Mouse Enteric Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 891:135-43. [DOI: 10.1007/978-3-319-27592-5_13] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Liang C, Xu B. Advances in understanding role of brain-derived neurotrophic factor in physiological and pathological processes in the intestinal tract. Shijie Huaren Xiaohua Zazhi 2015; 23:5649-5654. [DOI: 10.11569/wcjd.v23.i35.5649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Brain-derived neurotrophic factor is a kind of neurotrophic substance. In recent years, besides the central nervous system, brain-derived neurotrophic factor was also found to be expressed abundantly in the gastrointestinal tract, and it plays an important role in the development of the enteric nervous system and in regulating intestinal motility and visceral sensitivity. In this article, we review the role of brain-derived neurotrophic factor in the intestinal tract, and discuss its possible role in the pathogenesis of irritable bowel syndrome, with an aim to provide new ideas for clinical treatment of gastrointestinal diseases.
Collapse
|
35
|
Pohl CS, Medland JE, Moeser AJ. Early-life stress origins of gastrointestinal disease: animal models, intestinal pathophysiology, and translational implications. Am J Physiol Gastrointest Liver Physiol 2015; 309:G927-41. [PMID: 26451004 PMCID: PMC4683303 DOI: 10.1152/ajpgi.00206.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/01/2015] [Indexed: 01/31/2023]
Abstract
Early-life stress and adversity are major risk factors in the onset and severity of gastrointestinal (GI) disease in humans later in life. The mechanisms by which early-life stress leads to increased GI disease susceptibility in adult life remain poorly understood. Animal models of early-life stress have provided a foundation from which to gain a more fundamental understanding of this important GI disease paradigm. This review focuses on animal models of early-life stress-induced GI disease, with a specific emphasis on translational aspects of each model to specific human GI disease states. Early postnatal development of major GI systems and the consequences of stress on their development are discussed in detail. Relevant translational differences between species and models are highlighted.
Collapse
Affiliation(s)
- Calvin S. Pohl
- 1Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; ,2Gastrointestinal Stress Biology Laboratory, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan; and
| | - Julia E. Medland
- 3Comparative Biomedical Sciences Program, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Adam J. Moeser
- 1Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; ,2Gastrointestinal Stress Biology Laboratory, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan; and
| |
Collapse
|
36
|
Arab HA, Muhammadnejad S, Faghihi SM, Hassanpour H, Muhammadnejad A. Effects of nitric oxide modulating activities on development of enteric nervous system mediated gut motility in chick embryo model. J Biosci 2015; 39:835-48. [PMID: 25431412 DOI: 10.1007/s12038-014-9474-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The enteric nervous system (ENS) arises from the enteric neural crest-derived cells (ENCCs), and many molecules and biochemical processes may be involved in its development. This study examined the effects of modulating embryonic nitric oxide (NO) activity on the intestinal motility induced by ENS. One-hundred-and-twenty fertilized chicken eggs were assigned to three main groups and incubated at 37 degrees Centigrade and 60 percent humidity. The eggs were treated with NG-nitro-Larginine methyl ester (L-NAME), sodium nitroprusside (SNP), L-arginine (L-Arg) or vehicle from days 3 (1st group), 7 (2nd group) and 10 (3rd group) of incubation and continued up to day 18. On day 19, the embryos were sacrificed, the jejunal and colorectal segments were taken and the intestinal motility was assessed using isolated organ system. The intestinal motility was recorded normally and following cholinergic, adrenergic and non-adrenergic non-cholinergic (NANC) stimulations. The ENS structure was assessed by immunohistochemistry (IHC) using glial fibrillary acidic protein (GFAP). Rhythmic intestinal contractions were seen in all treatment groups, but inhibition of NO in the LNAME- treated embryos caused significant decrease (p less than 0.01) in the frequency and amplitude of the contraction. The responsiveness to adrenergic, cholinergic and NANC stimulations was also significantly decreased (p less than 0.05). The GFAP expression was significantly (p less than 0.05) reduced in the L-NAME-treated embryos. This study showed that the inhibition of NO caused a deficient development of the ENS, leading to a decrease in the frequency and amplitude of the intestinal contractions and reduced the responsiveness to adrenergic, cholinergic and NANC signalling.
Collapse
Affiliation(s)
- Hossein-Ali Arab
- Department of Pharmacology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran,
| | | | | | | | | |
Collapse
|
37
|
Hisamatsu C, Morioka I, Matsuo K, Iijima K, Nishijima E. Intraventricular hemorrhage as a comorbid factor for intestinal obstruction in very low-birthweight infants. Pediatr Int 2015; 57:427-30. [PMID: 25443271 DOI: 10.1111/ped.12548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 10/28/2014] [Accepted: 11/19/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND In very low-birthweight infants (VLBWI), intestinal obstruction (IO) tends to increase in severity due to the immaturity of the intestine and perinatal events. We examined the clinical comorbid factors related to IO in VLBWI. METHODS Clinical data of 160 VLBWI admitted to the neonatal intensive care unit in 2006-2011 were retrospectively reviewed. Patients were divided into two groups: IO group (n = 62) and non-IO group (n = 98). IO was defined as bile excretion via the mouth or nasogastric tube within 30 days after birth. The relationship between clinical factors and the incidence of IO was analyzed. RESULTS On univariate analysis, gestational age, birthweight, and the incidence of chronic lung disease, patent ductus arteriosus, intraventricular hemorrhage (IVH), retinopathy of prematurity, and the postnatal use of mechanical ventilation, catecholamines, steroids, and sedatives were associated with IO. On multivariate analysis, only IVH was strongly associated with IO (OR, 4.74; P < 0.01). CONCLUSIONS IVH is a significant comorbid factor of IO in VLBWI.
Collapse
Affiliation(s)
- Chieko Hisamatsu
- Division of Pediatric Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ichiro Morioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kiyomi Matsuo
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Eiji Nishijima
- Division of Pediatric Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
38
|
Rego SL, Raghavan S, Zakhem E, Bitar KN. Enteric neural differentiation in innervated, physiologically functional, smooth muscle constructs is modulated by bone morphogenic protein 2 secreted by sphincteric smooth muscle cells. J Tissue Eng Regen Med 2015; 11:1251-1261. [PMID: 25926098 DOI: 10.1002/term.2027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/09/2015] [Accepted: 03/19/2015] [Indexed: 01/01/2023]
Abstract
The enteric nervous system (ENS) controls gastrointestinal (GI) functions, including motility and digestion, which are impaired in ENS disorders. Differentiation of enteric neurons is mediated by factors released by the gut mesenchyme, including smooth muscle cells (SMCs). SMC-derived factors involved in adult enteric neural progenitor cells (NPCs) differentiation remain elusive. Furthermore, physiologically relevant in vitro models to investigate the innervations of various regions of the gut, such as the pylorus and lower oesophageal sphincter (LES), are not available. Here, neural differentiation in bioengineered innervated circular constructs composed of SMCs isolated from the internal anal sphincter (IAS), pylorus, LES and colon of rabbits was investigated. Additionally, SMC-derived factors that induce neural differentiation were identified to optimize bioengineered construct innervations. Sphincteric and non-sphincteric bioengineered constructs aligned circumferentially and SMCs maintained contractile phenotypes. Sphincteric constructs generated spontaneous basal tones. Higher levels of excitatory and inhibitory motor neuron differentiation and secretion of bone morphogenic protein 2 (BMP2) were observed in bioengineered, innervated, sphincteric constructs compared to non-sphincteric constructs. The addition of BMP2 to non-sphincteric colonic SMC constructs increased nitrergic innervations, and inhibition of BMP2 with noggin in sphincteric constructs decreased functional relaxation. These studies provide: (a) the first bioengineered innervated pylorus and LES constructs; (b) physiologically relevant models to investigate SMCs and adult NPCs interactions; and (c) evidence of the region-specific effects of SMCs on neural differentiation mediated by BMP2. Furthermore, this study paves the way for the development of innervated bioengineered GI tissue constructs tailored to specific disorders and locations within the gut. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Stephen L Rego
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shreya Raghavan
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Elie Zakhem
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Khalil N Bitar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
39
|
Enteric nervous system abnormalities are present in human necrotizing enterocolitis: potential neurotransplantation therapy. Stem Cell Res Ther 2014; 4:157. [PMID: 24423414 PMCID: PMC4054965 DOI: 10.1186/scrt387] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 10/15/2013] [Accepted: 11/11/2013] [Indexed: 02/07/2023] Open
Abstract
Introduction Intestinal dysmotility following human necrotizing enterocolitis suggests that the enteric nervous system is injured during the disease. We examined human intestinal specimens to characterize the enteric nervous system injury that occurs in necrotizing enterocolitis, and then used an animal model of experimental necrotizing enterocolitis to determine whether transplantation of neural stem cells can protect the enteric nervous system from injury. Methods Human intestinal specimens resected from patients with necrotizing enterocolitis (n = 18), from control patients with bowel atresia (n = 8), and from necrotizing enterocolitis and control patients undergoing stoma closure several months later (n = 14 and n = 6 respectively) were subjected to histologic examination, immunohistochemistry, and real-time reverse-transcription polymerase chain reaction to examine the myenteric plexus structure and neurotransmitter expression. In addition, experimental necrotizing enterocolitis was induced in newborn rat pups and neurotransplantation was performed by administration of fluorescently labeled neural stem cells, with subsequent visualization of transplanted cells and determination of intestinal integrity and intestinal motility. Results There was significant enteric nervous system damage with increased enteric nervous system apoptosis, and decreased neuronal nitric oxide synthase expression in myenteric ganglia from human intestine resected for necrotizing enterocolitis compared with control intestine. Structural and functional abnormalities persisted months later at the time of stoma closure. Similar abnormalities were identified in rat pups exposed to experimental necrotizing enterocolitis. Pups receiving neural stem cell transplantation had improved enteric nervous system and intestinal integrity, differentiation of transplanted neural stem cells into functional neurons, significantly improved intestinal transit, and significantly decreased mortality compared with control pups. Conclusions Significant injury to the enteric nervous system occurs in both human and experimental necrotizing enterocolitis. Neural stem cell transplantation may represent a novel future therapy for patients with necrotizing enterocolitis.
Collapse
|
40
|
Eifinger F, Lubomirov LT, Dercks E, Genchev B, Roth B, Neiss WF, Pfitzer G, Schroeter MM. Neonatal mouse ileum: functional properties and protein composition of the contractile machinery. Pediatr Res 2014; 76:252-60. [PMID: 24964231 DOI: 10.1038/pr.2014.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 03/21/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Immature motility of the ileum may contribute to life-threatening diseases. Little is known about the normal biomechanics of the neonatal ileum in relation to the protein composition of its contractile machinery. METHODS We analyzed the tissue architecture, the biomechanics in intact and β-escin-permeabilized preparations, and the protein composition in neonatal (P0) and adult murine ileum. RESULTS Muscle thickness of the P0 ileum was -50% of the adult ileum and passive compliance was higher. Carbachol- and KCl-elicited contractions were tonic rather than phasic as in the adult. Ca(2+) sensitivity was higher and relaxation rate was slower in β-escin-permeabilized P0 compared with adult ileum. The expression level of β-actin relative to α-actin was higher, and those of total actin, myosin, myosin light chain kinase, the catalytic subunit of myosin phosphatase and telokin were lower compared with the adult. The expression level of MYPT1 was similar, but P0 ileum expressed only the M133; the adult ileum also expressed the M130 isoform. CONCLUSION The mechanical features and protein composition of the P0 ileum are similar to those of adult tonic smooth muscles. We propose that this is highly adaptive during fetal life allowing the small intestine to act predominantly as a container.
Collapse
Affiliation(s)
- Frank Eifinger
- 1] Department of Neonatology, University Children's Hospital, Cologne, Germany [2] Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | | | - Elena Dercks
- 1] Department of Neonatology, University Children's Hospital, Cologne, Germany [2] Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Borislav Genchev
- Department of Anatomy I, University of Cologne, Cologne, Germany
| | - Bernhard Roth
- Department of Neonatology, University Children's Hospital, Cologne, Germany
| | - Wolfram F Neiss
- Department of Anatomy I, University of Cologne, Cologne, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
41
|
Gao J, Sathar S, O'Grady G, Han J, Cheng LK. Developmental changes in postnatal murine intestinal interstitial cell of Cajal network structure and function. Ann Biomed Eng 2014; 42:1729-39. [PMID: 24866568 DOI: 10.1007/s10439-014-1021-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 04/26/2014] [Indexed: 01/18/2023]
Abstract
The mammalian gastrointestinal (GI) tract undergoes rapid development during early postnatal life in order to transition from a milk to solid diet. Interstitial cells of Cajal (ICC) are the pacemaker cells that coordinate smooth muscle contractility within the GI tract, and hence we hypothesized that ICC networks undergo significant developmental changes during this early postnatal period. Numerical metrics for quantifying ICC network structural properties were applied on confocal ICC network imaging data obtained from the murine small intestine at various postnatal ages spanning birth to weaning. These imaging data were also coupled to a biophysically-based computational model to simulate pacemaker activity in the networks, to quantify how changes in structure may alter function. The results showed a pruning-like mechanism which occurs during postnatal development, and the temporal course of this phenomenon was defined. There was an initial ICC process overgrowth to optimize network efficiency and increase functional output volume. This was followed by a selective retaining and strengthening of processes, while others were discarded to further elevate functional output volume. Subsequently, new ICC processes were formed and the network was adjusted to its adult morphology. These postnatal ICC network developmental events may be critical in facilitating mature digestive function.
Collapse
Affiliation(s)
- Jerry Gao
- Auckland Bioengineering Institute, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | | | | | | | | |
Collapse
|
42
|
Abstract
The enteric nervous system is vulnerable to a range of congenital and acquired disorders that disrupt the function of its neurons or lead to their loss. The resulting enteric neuropathies are some of the most challenging clinical conditions to manage. Neural stem cells offer the prospect of a cure given their potential ability to replenish missing or dysfunctional neurons. This article discusses diseases that might be targets for stem cell therapies and the barriers that could limit treatment application. We explore various sources of stem cells and the proof of concept for their use. The critical steps that remain to be addressed before these therapies can be used in patients are also discussed. Key milestones include the harvesting of neural stem cells from the human gut and the latest in vivo transplantation studies in animals. The tremendous progress in the field has brought experimental studies exploring the potential of stem cell therapies for the management of enteric neuropathies to the cusp of clinical application.
Collapse
Affiliation(s)
- Alan J Burns
- Neural Development and Gastroenterology Units, Birth Defects Research Centre, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Nikhil Thapar
- 1] Neural Development and Gastroenterology Units, Birth Defects Research Centre, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK. [2] Division of Neurogastroenterology and Motility, Department of Paediatric Gastroenterology, Great Ormond Street Hospital, Great Ormond Street, London WC1N 3JH, UK
| |
Collapse
|
43
|
Mattos MP, da Silva AL. Comparative study of jejunal and colonic alterations between gastrojejunoduodenal and gastrocoloduodenal interposition in dogs. ABCD-ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA 2014; 26:260-7. [PMID: 24510032 DOI: 10.1590/s0102-67202013000400003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/19/2013] [Indexed: 11/22/2022]
Abstract
BACKGROUND The intestinal interpositions are important in various operations of the digestive tract. However, they are capable to produce serious immediate and late complications. AIM To compare clinical and surgical outcomes, as well as intestinal pathological findings, in dogs submitted to jejunum and colon interposition. METHODS Forty male mongreal dogs were immunized and dewormed, between 36 and 72 months old and weighting from 6 to 16 kg divided into two groups of 20 (G1=gastrojejunoduodenal and G2=gastrocoloduodenal). They were anesthetized and submitted to laparotomy with jejunum and colon interposition between the gastric antrum and the duodenum. On the 120th postoperative day, a relaparotomy was performed with removal of the jejunun/colon segments for evaluation. The segments were processed using Bouin and tissue were stained with H&E for histological analysis. Clinical and surgical outcomes analyzed were weight, diarrhea, fistula, infection, suture dehiscence and incisional hernia. The pathological analysis involved neutrophilic infiltration, increase in number of goblet cells and villous and crypts hypotrophy. Mann-Whitney and Fisher Exact tests were used for groups comparisons. RESULTS Dogs that underwent gastrocoloduodenal interposition had a higher weight loss (p<0.05) and more diarrhea (p<0.001). Clinically, controlled fistula rates were 20% in G1 and 100% in G2. Complications occurred in 20% in G1 and in all animals in G2 (p<0.001). Pathological alterations were also more prevalent in G2 (p<0.001). CONCLUSION There is a high morbidity and complication rates involving intestine interposition and higher in G2 where colon segments were used.
Collapse
|
44
|
Expression of neuropeptides and anoctamin 1 in the embryonic and adult zebrafish intestine, revealing neuronal subpopulations and ICC-like cells. Cell Tissue Res 2013; 354:355-70. [PMID: 23881406 DOI: 10.1007/s00441-013-1685-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 06/17/2013] [Indexed: 12/12/2022]
Abstract
This immunohistochemical study in zebrafish aims to extend the neurochemical characterization of enteric neuronal subpopulations and to validate a marker for identification of interstitial cells of Cajal (ICC). The expression of neuropeptides and anoctamin 1 (Ano1), a selective ICC marker in mammals, was analyzed in both embryonic and adult intestine. Neuropeptides were present from 3 days postfertilization (dpf). At 3 dpf, galanin-positive nerve fibers were found in the proximal intestine, while calcitonin gene-related peptide (CGRP)- and substance P-expressing fibers appeared in the distal intestine. At 5 dpf, immunoreactive fibers were present along the entire intestinal length, indicating a well-developed peptidergic innervation at the onset of feeding. In the adult intestine, vasoactive intestinal peptide (VIP), pituitary adenylate cyclase-activating peptide (PACAP), galanin, CGRP and substance P were detected in nerve fibers. Colchicine pretreatment enhanced only VIP and PACAP immunoreactivity. VIP and PACAP were coexpressed in enteric neurons. Colocalization stainings revealed three neuronal subpopulations expressing VIP and PACAP: a nitrergic noncholinergic subpopulation, a serotonergic subpopulation and a subpopulation expressing no other markers. Ano1-immunostaining revealed a 3-dimensional network in the adult intestine containing multipolar cells at the myenteric plexus and bipolar cells interspersed between circular smooth muscle cells. Ano1 immunoreactivity first appeared at 3 dpf, indicative of the onset of proliferation of ICC-like cells. It is shown that the Ano1 antiserum is a selective marker of ICC-like cells in the zebrafish intestine. Finally, it is hypothesized that ICC-like cells mediate the spontaneous regular activity of the embryonic intestine.
Collapse
|
45
|
Deoti B, Arantes RME, Campos CF, Hayck J, Silva ALD. Morphology and immunohistochemistry of the myenteric plexus of valves constructed in the colon of rats submitted to abdominoperineal amputation and perineal colostomy. Acta Cir Bras 2013; 28:272-81. [PMID: 23568235 DOI: 10.1590/s0102-86502013000400007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/18/2013] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To investigate immunohistochemical aspects of the myenteric plexus of valves constructed in the colon of rats to verify whether any denervation occurs both at the operative site and in those areas adjacent to the third valve. METHODS Thirty six male Wistar rats divided into the following three groups were used: Control Group (CG); Amputated Group (AG); Amputated Group with Valves (AGWV). In AG was held in the rectum amputation and the colon was sutured to the skin elaborating the perineal colostomy. In AGWV was held in the rectum amputation. A laparotomy was performed for the manufacture of valves (seromyotomy) in the colon. After this step, the colon was sutured to the skin elaborating the perineal colostomy. The density of the neural elements in the muscular wall as marked specifically using Protein Gene Product (PGP) 9.5 and utilising the proper tools of the KS300 software for measuring the area. From these measurements, a relation and three proportions were drawn and analysed according to the mean of the averages obtained from the measured images. RESULTS Immunoexpression of PGP 9.5 demonstrated a total absence of neural elements and myenteric plexus at the valve site. The density of the neural elements in the circular muscular layer at sites adjacent to the 3rd valve was lesser, however, was not significantly different. CONCLUSION The immunohistochemical study of valves constructed in the colon of rats submitted to abdominoperineal amputation and perineal colostomy revealed denervation at the seromyotomy site.
Collapse
Affiliation(s)
- Beatriz Deoti
- Department of Surgery, UFMG, Belo Horizonte, MG, Brazil.
| | | | | | | | | |
Collapse
|
46
|
CFTR-deficient pigs display peripheral nervous system defects at birth. Proc Natl Acad Sci U S A 2013; 110:3083-8. [PMID: 23382208 DOI: 10.1073/pnas.1222729110] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Peripheral nervous system abnormalities, including neuropathy, have been reported in people with cystic fibrosis. These abnormalities have largely been attributed to secondary manifestations of the disease. We tested the hypothesis that disruption of the cystic fibrosis transmembrane conductance regulator (CFTR) gene directly influences nervous system function by studying newborn CFTR(-/-) pigs. We discovered CFTR expression and activity in Schwann cells, and loss of CFTR caused ultrastructural myelin sheath abnormalities similar to those in known neuropathies. Consistent with neuropathic changes, we found increased transcripts for myelin protein zero, a gene that, when mutated, can cause axonal and/or demyelinating neuropathy. In addition, axon density was reduced and conduction velocities of the trigeminal and sciatic nerves were decreased. Moreover, in vivo auditory brainstem evoked potentials revealed delayed conduction of the vestibulocochlear nerve. Our data suggest that loss of CFTR directly alters Schwann cell function and that some nervous system defects in people with cystic fibrosis are likely primary.
Collapse
|
47
|
Theocharatos S, Wilkinson DJ, Darling S, Wilm B, Kenny SE, Edgar D. Regulation of progenitor cell proliferation and neuronal differentiation in enteric nervous system neurospheres. PLoS One 2013; 8:e54809. [PMID: 23372773 PMCID: PMC3553067 DOI: 10.1371/journal.pone.0054809] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 12/17/2012] [Indexed: 12/18/2022] Open
Abstract
Enteric nervous system (ENS) progenitor cells isolated from mouse and human bowel can be cultured in vitro as neurospheres which are aggregates of the proliferating progenitor cells, together with neurons and glial cells derived from them. To investigate the factors regulating progenitor cell proliferation and differentiation, we first characterised cell proliferation in mouse ENS neurospheres by pulse chase experiments using thymidine analogs. We demonstrate rapid and continuous cell proliferation near the neurosphere periphery, after which postmitotic cells move away from the periphery to become distributed throughout the neurosphere. While many proliferating cells expressed glial markers, expression of the neuronal markers β-tubulin III (Tuj1) and nitric oxide synthase was detected in increasing numbers of post-mitotic cells after a delay of several days. Treatment of both mouse and human neurospheres with the γ-secretase inhibitor N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) reduced expression of the transcription factors Hes1 and Hes5, demonstrating inhibition of Notch signaling. DAPT treatment also inhibited progenitor cell proliferation and increased the numbers of differentiating neurons expressing Tuj1 and nitric oxide synthase. To confirm that the cellular effects of DAPT treatment were due to inhibition of Notch signaling, siRNA knockdown of RBPjκ, a key component of the canonical Notch signaling pathway, was demonstrated both to reduce proliferation and to increase neuronal differentiation in neurosphere cells. These observations indicate that Notch signaling promotes progenitor cell proliferation and inhibits neuronal differentiation in ENS neurospheres.
Collapse
Affiliation(s)
- Sokratis Theocharatos
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - David J. Wilkinson
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
- Institute of Child Health, University of Liverpool, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Sarah Darling
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - Simon E. Kenny
- Institute of Child Health, University of Liverpool, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - David Edgar
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Rich A, Gordon S, Brown C, Gibbons SJ, Schaefer K, Hennig G, Farrugia G. Kit signaling is required for development of coordinated motility patterns in zebrafish gastrointestinal tract. Zebrafish 2013; 10:154-60. [PMID: 23297728 DOI: 10.1089/zeb.2012.0766] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Interstitial cells of Cajal (ICC) provide a pacemaker signal for coordinated motility patterns in the mammalian gastrointestinal (GI) tract. Kit signaling is required for development and maintenance of ICC, and these cells can be identified by Kit-like immunoreactivity. The zebrafish GI tract has two distinct ICC networks similar to mammals, suggesting a similar role in the generation of GI motility; however, a functional role for Kit-positive cells in zebrafish has not been determined. Analysis of GI motility in intact zebrafish larvae was performed during development and after disruption of Kit signaling. Development of coordinated motility patterns occurred after 5 days post-fertilization (dpf) and correlated with appearance of Kit-positive cells. Disruptions of Kit signaling using the Kit antagonist imatinib mesylate, and in Sparse, a null kita mutant, also disrupted development of coordinated motility patterns. These data suggest that Kit signaling is necessary for development of coordinated motility patterns and that Kit-positive cells in zebrafish are necessary for coordinated motility patterns.
Collapse
Affiliation(s)
- Adam Rich
- Department of Biology, The College at Brockport, State University of New York , Brockport, NY 14420, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Thomason RT, Bader DM, Winters NI. Comprehensive timeline of mesodermal development in the quail small intestine. Dev Dyn 2012; 241:1678-94. [PMID: 22930586 DOI: 10.1002/dvdy.23855] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND To generate the mature intestine, splanchnic mesoderm diversifies into six different tissue layers each with multiple cell types through concurrent and complex morphogenetic events. Hindering the progress of research in the field is the lack of a detailed description of the fundamental morphological changes that constitute development of the intestinal mesoderm. RESULTS We used immunofluorescence and morphometric analyses of wild-type and Tg(tie1:H2B-eYFP) quail embryos to establish a comprehensive timeline of mesodermal development in the avian intestine. The following landmark features were analyzed from appearance of the intestinal primordium through generation of the definitive structure: radial compartment formation, basement membrane dynamics, mesothelial differentiation, mesenchymal expansion and growth patterns, smooth muscle differentiation, and maturation of the vasculature. In this way, structural relationships between mesodermal components were identified over time. CONCLUSIONS This integrated analysis presents a roadmap for investigators and clinicians to evaluate diverse experimental data obtained at individual stages of intestinal development within the longitudinal context of intestinal morphogenesis.
Collapse
Affiliation(s)
- Rebecca T Thomason
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | | | | |
Collapse
|
50
|
Bagyánszki M, Bódi N. Diabetes-related alterations in the enteric nervous system and its microenvironment. World J Diabetes 2012; 3:80-93. [PMID: 22645637 PMCID: PMC3360223 DOI: 10.4239/wjd.v3.i5.80] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 04/06/2012] [Accepted: 05/11/2012] [Indexed: 02/05/2023] Open
Abstract
Gastric intestinal symptoms common among diabetic patients are often caused by intestinal motility abnormalities related to enteric neuropathy. It has recently been demonstrated that the nitrergic subpopulation of myenteric neurons are especially susceptible to the development of diabetic neuropathy. Additionally, different susceptibility of nitrergic neurons located in different intestinal segments to diabetic damage and their different levels of responsiveness to insulin treatment have been revealed. These findings indicate the importance of the neuronal microenvironment in the pathogenesis of diabetic nitrergic neuropathy. The main focus of this review therefore was to summarize recent advances related to the diabetes-related selective nitrergic neuropathy and associated motility disturbances. Special attention was given to the findings on capillary endothelium and enteric glial cells. Growing evidence indicates that capillary endothelium adjacent to the myenteric ganglia and enteric glial cells surrounding them are determinative in establishing the ganglionic microenvironment. Additionally, recent advances in the development of new strategies to improve glycemic control in type 1 and type 2 diabetes mellitus are also considered in this review. Finally, looking to the future, the recent and promising results of metagenomics for the characterization of the gut microbiome in health and disease such as diabetes are highlighted.
Collapse
Affiliation(s)
- Mária Bagyánszki
- Mária Bagyánszki, Nikolett Bódi, Department of Physiology, Anatomy and Neuroscience, Faculty of Science, University of Szeged, H-6726 Szeged, Hungary
| | | |
Collapse
|