1
|
Mueller JL, Leavitt AR, Rahman AA, Han CY, Ott LC, Mahdavian NS, Carbone SE, King SK, Burns AJ, Poole DP, Hotta R, Goldstein AM, Stavely R. Highly neurogenic glia from human and mouse myenteric ganglia generate functional neurons following culture and transplantation into the gut. Cell Rep 2024; 43:114919. [PMID: 39471175 PMCID: PMC11697211 DOI: 10.1016/j.celrep.2024.114919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/05/2024] [Accepted: 10/13/2024] [Indexed: 11/01/2024] Open
Abstract
Enteric neural stem cell (ENSC) therapy offers great promise for neurointestinal diseases; however, current isolation methods yield insufficient neurons for regenerative applications. Multiomic profiling of enteric glial cells (EGCs) suggests that subpopulations within myenteric ganglia (MyGa) are a reservoir of highly neurogenic ENSCs. Here, we describe protocols to enrich for intraganglionic EGCs by isolating intact fragments of MyGa, generating cultures with higher neuronal purity than traditional methodologies isolating intramuscular single cells (IM-SCs). MyGa-derived EGCs transdifferentiate into more neurons than IM-SC-derived EGCs do, confirming their neurogenic predisposition. Following transplantation to the mouse intestine, MyGa-derived neurons generate calcium transients and activate smooth muscle in response to optogenetic stimulation. In the human intestine, MyGa-derived cells are similarly highly neurogenic, are enriched for a distinct progenitor population identified by single-cell RNA sequencing (scRNA-seq), and exhibit neuromuscular connectivity following xenogeneic transplantation into mice. Highly neurogenic ENSCs are preferentially located within the MyGa, and their selective isolation offers considerable potential for therapy.
Collapse
Affiliation(s)
- Jessica L Mueller
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abigail R Leavitt
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ahmed A Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher Y Han
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leah C Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Narges S Mahdavian
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Simona E Carbone
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Sebastian K King
- Department of Paediatric Surgery, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Alan J Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel P Poole
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Fujiwara N, Lee D, Li B, Pierro A, Yamataka A. Enhancement of enteric neural stem cell neurogenesis by glial cell-derived neurotrophic factor in experimental Hirschsprung's disease. Pediatr Surg Int 2024; 40:274. [PMID: 39460767 DOI: 10.1007/s00383-024-05861-3] [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] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
PURPOSE Stem cell therapy offers a promising solution for congenital diseases like Hirschsprung's disease (HSCR). Optimizing stem cell efficacy by modifying the cells and their environment is crucial, but in vitro culture conditions need to be further improved. Glial cell-derived neurotrophic factor (GDNF) plays an important role in neuronal survival, proliferation, migration and differentiation during enteric nervous system (ENS) development. In this study, the effects of GDNF on neurites derived from an Ednrb knockout model were investigated with the aim of enhancing the neurogenic potential of enteric neural crest cells (ENCCs). METHODS Neurospheres were generated form Ednrb+/+ (control) and Ednrb-/- mice at embryonic day13.5 (E13.5) with Sox10-green fluorescent protein (Venus) transgenic expression. These neurospheres were cultured in control media and neurospheres from Ednrb-/- were cultured with either control media or media supplemented with GDNF. ENCCs differentiation was assessed using immunofluorescence staining after 18 days. RESULTS GDNF-treated Ednrb-/- neurospheres showed increased size and higher density of Sox10-positive ENCCs compared to untreated Ednrb-/- neurospheres. GDNF also enhanced the distribution of both TUJ1-positive neurons and S100-positive glial cells. CONCLUSION GDNF effectively enhanced the neurogenic potential of ENCCs from HSCR animal model. This finding is crucial for the development of cell therapy in HSCR.
Collapse
Affiliation(s)
- Naho Fujiwara
- Department of Pediatric General and Urogenital Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada.
| | - Dorothy Lee
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada
| | - Atsuyuki Yamataka
- Department of Pediatric General and Urogenital Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
3
|
Herath M, Speer AL. Bioengineering of Intestinal Grafts. Gastroenterol Clin North Am 2024; 53:461-472. [PMID: 39068007 PMCID: PMC11284275 DOI: 10.1016/j.gtc.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Intestinal failure manifests as an impaired capacity of the intestine to sufficiently absorb vital nutrients and electrolytes essential for growth and well-being in pediatric and adult populations. Although parenteral nutrition remains the mainstay therapeutic approach, the pursuit of a definitive and curative strategy, such as regenerative medicine, is imperative. Substantial advancements in the field of engineered intestinal tissues present a promising avenue for addressing intestinal failure; nevertheless, extensive research is still necessary for effective translation from experimental benchwork to clinical bedside applications.
Collapse
Affiliation(s)
- Madushani Herath
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), 6431 Fannin Street, Suite 5.254, Houston, TX 77030, USA
| | - Allison L Speer
- Program in Children's Regenerative Medicine, Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), 6431 Fannin Street, Suite 5.254, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Yoshimaru K, Matsuura T, Uchida Y, Sonoda S, Maeda S, Kajihara K, Kawano Y, Shirai T, Toriigahara Y, Kalim AS, Zhang XY, Takahashi Y, Kawakubo N, Nagata K, Yamaza H, Yamaza T, Taguchi T, Tajiri T. Cutting-edge regenerative therapy for Hirschsprung disease and its allied disorders. Surg Today 2024; 54:977-994. [PMID: 37668735 DOI: 10.1007/s00595-023-02741-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/06/2023] [Indexed: 09/06/2023]
Abstract
Hirschsprung disease (HSCR) and its associated disorders (AD-HSCR) often result in severe hypoperistalsis caused by enteric neuropathy, mesenchymopathy, and myopathy. Notably, HSCR involving the small intestine, isolated hypoganglionosis, chronic idiopathic intestinal pseudo-obstruction, and megacystis-microcolon-intestinal hypoperistalsis syndrome carry a poor prognosis. Ultimately, small-bowel transplantation (SBTx) is necessary for refractory cases, but it is highly invasive and outcomes are less than optimal, despite advances in surgical techniques and management. Thus, regenerative therapy has come to light as a potential form of treatment involving regeneration of the enteric nervous system, mesenchyme, and smooth muscle in affected areas. We review the cutting-edge regenerative therapeutic approaches for managing HSCR and AD-HSCR, including the use of enteric nervous system progenitor cells, embryonic stem cells, induced pluripotent stem cells, and mesenchymal stem cells as cell sources, the recipient intestine's microenvironment, and transplantation methods. Perspectives on the future of these treatments are also discussed.
Collapse
Affiliation(s)
- Koichiro Yoshimaru
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Toshiharu Matsuura
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Yasuyuki Uchida
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Soichiro Sonoda
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shohei Maeda
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keisuke Kajihara
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Kawano
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takeshi Shirai
- Department of Pediatric Surgery, Miyazaki Prefectural Miyazaki Hospital, 5-30 Kitatakamatsu-cho, Miyazaki, Miyazaki, 880-8510, Japan
| | - Yukihiro Toriigahara
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Alvin Santoso Kalim
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Xiu-Ying Zhang
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshiaki Takahashi
- Department of Pediatric Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757, Asahimachi-dori, Chuo-ku, Niigata, Japan
| | - Naonori Kawakubo
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kouji Nagata
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Department of Pediatric Dentistry, Kyushu University Graduate School of Dental Science, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoaki Taguchi
- Fukuoka College of Health Sciences, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
5
|
Elkrewi EZ, Al Abdulqader AA, Khasanov R, Maas-Omlor S, Boettcher M, Wessel LM, Schäfer KH, Tapia-Laliena MÁ. Role of Inflammation and the NF-κB Signaling Pathway in Hirschsprung's Disease. Biomolecules 2024; 14:992. [PMID: 39199380 PMCID: PMC11352745 DOI: 10.3390/biom14080992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Hirschsprung's disease (HSCR, incidence 1/5000 live births) is caused by the failure of neural crest-derived precursors to migrate, survive, proliferate, or differentiate during the embryonic development of the Enteric Nervous System (ENS), which could be disrupted by many factors, including inflammatory processes. The NF-κB family controls several biological processes, including inflammation, neurogenesis, and cell migration. With the aim of studying the potential role of NF-κB in HSCR, we have analyzed the expression of the NF-κB main subunits and other NF-κB-related genes by RT-qPCR in HSCR tissue samples (sub-divided into ganglionic and aganglionic segments). We found decreased gene expression of the NF-κB main subunit RELA but also of NFKBIA, TNFA, TFGBR2, and ERBB3 in the pathologic distal aganglionic segments compared to the proximal ganglionic segments. Moreover, we could also confirm the lower protein expression of RelA/p65 in the aganglionic distal segments by immunofluorescence staining. Further, we show that the expression of RelA/p65 protein in the proximal segments concurs with lymphocyte infiltration in the bowel tissue, indicating a pro-inflammatory activation of p65 in the proximal ganglionic HSCR tissue in the patients analyzed. All in all, our findings suggest that the modulation of NF-κB signaling in the neuro-enteric system does obviously contribute to the pathological effects of HSCR.
Collapse
Affiliation(s)
- Enas Zoheer Elkrewi
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany
| | - Ahmad A. Al Abdulqader
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany
- Department of Surgery, College of Medicine, King Faisal University, Al Hofuf 31982, Saudi Arabia
| | - Rasul Khasanov
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany
| | - Silke Maas-Omlor
- Working Group Enteric Nervous Systems (AGENS), University of Applied Sciences Kaiserslautern, Amerikastrasse 1,66482 Zweibrücken, Germany (K.-H.S.)
| | - Michael Boettcher
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany
| | - Lucas M. Wessel
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany
| | - Karl-Herbert Schäfer
- Working Group Enteric Nervous Systems (AGENS), University of Applied Sciences Kaiserslautern, Amerikastrasse 1,66482 Zweibrücken, Germany (K.-H.S.)
| | - María Ángeles Tapia-Laliena
- Department of Pediatric Surgery, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1–3, 68167 Mannheim, Germany
| |
Collapse
|
6
|
Rahman AA, Ohkura T, Bhave S, Pan W, Ohishi K, Ott L, Han C, Leavitt A, Stavely R, Burns AJ, Goldstein AM, Hotta R. Enteric neural stem cell transplant restores gut motility in mice with Hirschsprung disease. JCI Insight 2024; 9:e179755. [PMID: 39042470 PMCID: PMC11385093 DOI: 10.1172/jci.insight.179755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
The goal of this study was to determine if transplantation of enteric neural stem cells (ENSCs) can rescue the enteric nervous system, restore gut motility, reduce colonic inflammation, and improve survival in the Ednrb-KO mouse model of Hirschsprung disease (HSCR). ENSCs were isolated from mouse intestine, expanded to form neurospheres, and microinjected into the colons of recipient Ednrb-KO mice. Transplanted ENSCs were identified in recipient colons as cell clusters in "neo-ganglia." Immunohistochemical evaluation demonstrated extensive cell migration away from the sites of cell delivery and across the muscle layers. Electrical field stimulation and optogenetics showed significantly enhanced contractile activity of aganglionic colonic smooth muscle following ENSC transplantation and confirmed functional neuromuscular integration of the transplanted ENSC-derived neurons. ENSC injection also partially restored the colonic migrating motor complex. Histological examination revealed a significant reduction in inflammation in ENSC-transplanted aganglionic recipient colon compared with that of sham-operated mice. Interestingly, mice that received cell transplant also had prolonged survival compared with controls. This study demonstrates that ENSC transplantation can improve outcomes in HSCR by restoring gut motility and reducing the severity of Hirschsprung-associated enterocolitis, the leading cause of death in human HSCR.
Collapse
Affiliation(s)
- Ahmed A Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Weikang Pan
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Kensuke Ohishi
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan
| | - Leah Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher Han
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Abigail Leavitt
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan J Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Mandal A, Moneme C, Tewari BP, Goldstein AM, Sontheimer H, Cheng L, Moore SR, Levin D. A novel method for culturing enteric neurons generates neurospheres containing functional myenteric neuronal subtypes. J Neurosci Methods 2024; 407:110144. [PMID: 38670535 PMCID: PMC11144385 DOI: 10.1016/j.jneumeth.2024.110144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/04/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND The enteric nervous system (ENS) is comprised of neurons, glia, and neural progenitor cells that regulate essential gastrointestinal functions. Advances in high-efficiency enteric neuron culture would facilitate discoveries surrounding ENS regulatory processes, pathophysiology, and therapeutics. NEW METHOD Development of a simple, robust, one-step method to culture murine enteric neurospheres in a 3D matrix that supports neural growth and differentiation. RESULTS Myenteric plexus cells isolated from the entire length of adult murine small intestine formed ≥3000 neurospheres within 7 days. Matrigel-embedded neurospheres exhibited abundant neural stem and progenitor cells expressing Sox2, Sox10 and Msi1 by day 4. By day 5, neural progenitor cell marker Nestin appeared in the periphery of neurospheres prior to differentiation. Neurospheres produced extensive neurons and neurites, confirmed by Tubulin beta III, PGP9.5, HuD/C, and NeuN immunofluorescence, including neural subtypes Calretinin, ChAT, and nNOS following 8 days of differentiation. Individual neurons within and external to neurospheres generated depolarization induced action potentials which were inhibited in the presence of sodium channel blocker, Tetrodotoxin. Differentiated neurospheres also contained a limited number of glia and endothelial cells. COMPARISON WITH EXISTING METHODS This novel one-step neurosphere growth and differentiation culture system, in 3D format (in the presence of GDNF, EGF, and FGF2), allows for ∼2-fold increase in neurosphere count in the derivation of enteric neurons with measurable action potentials. CONCLUSION Our method describes a novel, robust 3D culture of electrophysiologically active enteric neurons from adult myenteric neural stem and progenitor cells.
Collapse
Affiliation(s)
- Arabinda Mandal
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Chioma Moneme
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Bhanu P Tewari
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Lily Cheng
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Sean R Moore
- Department of Pediatrics, Division of Pediatric Gastroenterology Hepatology, and Nutrition, University of Virginia, Charlottesville, VA, USA.
| | - Daniel Levin
- Department of Surgery, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
8
|
Pan W, Rahman AA, Ohkura T, Stavely R, Ohishi K, Han CY, Leavitt A, Kashiwagi A, Burns AJ, Goldstein AM, Hotta R. Autologous cell transplantation for treatment of colorectal aganglionosis in mice. Nat Commun 2024; 15:2479. [PMID: 38509106 PMCID: PMC10954649 DOI: 10.1038/s41467-024-46793-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/11/2024] [Indexed: 03/22/2024] Open
Abstract
Neurointestinal diseases cause significant morbidity and effective treatments are lacking. This study aimes to test the feasibility of transplanting autologous enteric neural stem cells (ENSCs) to rescue the enteric nervous system (ENS) in a model of colonic aganglionosis. ENSCs are isolated from a segment of small intestine from Wnt1::Cre;R26iDTR mice in which focal colonic aganglionosis is simultaneously created by diphtheria toxin injection. Autologous ENSCs are isolated, expanded, labeled with lentiviral-GFP, and transplanted into the aganglionic segment in vivo. ENSCs differentiate into neurons and glia, cluster to form neo-ganglia, and restore colonic contractile activity as shown by electrical field stimulation and optogenetics. Using a non-lethal model of colonic aganglionosis, our results demonstrate the potential of autologous ENSC therapy to improve functional outcomes in neurointestinal disease, laying the groundwork for clinical application of this regenerative cell-based approach.
Collapse
Affiliation(s)
- Weikang Pan
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Surgery, The second affiliated hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ahmed A Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kensuke Ohishi
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co., Ltd., Hiroshima, Japan
| | - Christopher Y Han
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abigail Leavitt
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aki Kashiwagi
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alan J Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Ohkura T, Burns AJ, Hotta R. Updates and Challenges in ENS Cell Therapy for the Treatment of Neurointestinal Diseases. Biomolecules 2024; 14:229. [PMID: 38397466 PMCID: PMC10887039 DOI: 10.3390/biom14020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Neurointestinal diseases represent a significant challenge in clinical management with current palliative approaches failing to overcome disease and treatment-related morbidity. The recent progress with cell therapy to restore missing or defective components of the gut neuromusculature offers new hope for potential cures. This review discusses the progress that has been made in the sourcing of putative stem cells and the studies into their biology and therapeutic potential. We also explore some of the practical challenges that must be overcome before cell-based therapies can be applied in the clinical setting. Although a number of obstacles remain, the rapid advances made in the enteric neural stem cell field suggest that such therapies are on the near horizon.
Collapse
Affiliation(s)
- Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (T.O.); (A.J.B.)
| | - Alan J. Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (T.O.); (A.J.B.)
- Stem Cells and Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (T.O.); (A.J.B.)
| |
Collapse
|
10
|
Scharr M, Hirt B, Neckel PH. Spatial gene expression profile of Wnt-signaling components in the murine enteric nervous system. Front Immunol 2024; 15:1302488. [PMID: 38322254 PMCID: PMC10846065 DOI: 10.3389/fimmu.2024.1302488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction Wnt-signaling is a key regulator of stem cell homeostasis, extensively studied in the intestinal crypt and other metazoan tissues. Yet, there is hardly any data available on the presence of Wnt-signaling components in the adult enteric nervous system (ENS) in vivo. Methods Therefore, we employed RNAscope HiPlex-assay, a novel and more sensitive in situ hybridization technology. By amplifying target specific signals, this technique enables the detection of low abundance, tightly regulated RNA content as is the case for Wnt-signaling components. Additionally, we compared our data to previously published physiological single cell RNA and RiboTag-based RNA sequencing analyses of enteric gliosis using data-mining approaches. Results Our descriptive analysis shows that several components of the multidi-mensional regulatory network of the Wnt-signaling pathway are present in the murine ENS. The transport and secretion protein for Wnt-ligands Wntless as well as canonical (Wnt3a and Wnt2b) and non-canonical Wnt-ligands (Wnt5a, Wnt7a, Wnt8b and Wnt11) are detectable within submucosal and myenteric plexus. Further, corresponding Frizzled receptors (Fzd1, Fzd3, Fzd6, and Fzd7) and regulatory signaling mediators like R-Spondin/DKK ligands are present in the ENS of the small and large intestine. Further, data mining approaches revealed, that several Wnt-related molecules are expressed by enteric glial cell clusters and are dynamically regulated during the inflammatory manifestation of enteric gliosis. Discussion Our results suggest, that canonical and non-canonical Wnt-signaling has a much broader impact on the mature ENS and its cellular homeostasis in health and inflammation, than previously anticipated.
Collapse
Affiliation(s)
| | | | - Peter H. Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Chen JC, Yang W, Tseng LY, Chang HL. Enteric neurospheres retain the capacity to assemble neural networks with motile and metamorphic gliocytes and ganglia. Stem Cell Res Ther 2023; 14:290. [PMID: 37798638 PMCID: PMC10557225 DOI: 10.1186/s13287-023-03517-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Neurosphere medium (NSM) and self-renewal medium (SRM) were widely used to isolate enteric neural stem cells (ENSCs) in the form of neurospheres. ENSCs or their neurosphere forms were neurogenic and gliogenic, but the compelling evidence for their capacity of assembling enteric neural networks remained lacking, raising the question of their aptitude for rebuilding the enteric nervous system (ENS) in ENSC therapeutics. It prompted us to explore an effective culture protocol or strategy for assembling ENS networks, which might also be employed as an in vitro model to simplify the biological complexity of ENS embedded in gut walls. METHODS NSM and SRM were examined for their capacity to generate neurospheres in mass culture of dispersed murine fetal enterocytes at serially diluted doses and assemble enteric neural networks in two- and three-dimensional cell culture systems and ex vivo on gut explants. Time-lapse microphotography was employed to capture cell activities of assembled neural networks. Neurosphere transplantation was performed via rectal submucosal injection. RESULTS In mass culture of dispersed enterocytes, NSM generated discrete units of neurospheres, whereas SRM promoted neural network assembly with neurospheres akin to enteric ganglia. Both were highly affected by seeding cell doses. SRM had similar ENSC mitosis-driving capacity to NSM, but was superior in driving ENSC differentiation in company with heightened ENSC apoptosis. Enteric neurospheres were motile, capable of merging together. It argued against their clonal entities. When nurtured in SRM, enteric neurospheres proved competent to assemble neural networks on two-dimensional coverslips, in three-dimensional hydrogels and on gut explants. In the course of neural network assembly from enteric neurospheres, neurite extension was preceded by migratory expansion of gliocytes. Assembled neural networks contained motile ganglia and gliocytes that constantly underwent shapeshift. Neurospheres transplanted into rectal submucosa might reconstitute myenteric plexuses of recipients' rectum. CONCLUSION Enteric neurospheres mass-produced in NSM might assemble neural networks in SRM-immersed two- or three-dimensional environments and on gut explants, and reconstitute myenteric plexuses of the colon after rectal submucosal transplantation. Our results also shed first light on the dynamic entity of ENS and open the experimental avenues to explore cellular activities of ENS and facilitate ENS demystification.
Collapse
Affiliation(s)
- Jeng-Chang Chen
- Department of Surgery, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, 5, Fu-Shin Street, Kweishan, Taoyuan, 333, Taiwan.
| | - Wendy Yang
- Department of Surgery, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, 5, Fu-Shin Street, Kweishan, Taoyuan, 333, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Li-Yun Tseng
- Pediatric Research Center, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Hsueh-Ling Chang
- Pediatric Research Center, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| |
Collapse
|
12
|
Hotta R, Rahman A, Bhave S, Stavely R, Pan W, Srinivasan S, de Couto G, Rodriguez-Borlado L, Myers R, Burns AJ, Goldstein AM. Transplanted ENSCs form functional connections with intestinal smooth muscle and restore colonic motility in nNOS-deficient mice. Stem Cell Res Ther 2023; 14:232. [PMID: 37667277 PMCID: PMC10478362 DOI: 10.1186/s13287-023-03469-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Enteric neuropathies, which result from abnormalities of the enteric nervous system, are associated with significant morbidity and high health-care costs, but current treatments are unsatisfactory. Cell-based therapy offers an innovative approach to replace the absent or abnormal enteric neurons and thereby restore gut function. METHODS Enteric neuronal stem cells (ENSCs) were isolated from the gastrointestinal tract of Wnt1-Cre;R26tdTomato mice and generated neurospheres (NS). NS transplants were performed via injection into the mid-colon mesenchyme of nNOS-/- mouse, a model of colonic dysmotility, using either 1 (n = 12) or 3 (n = 12) injections (30 NS per injection) targeted longitudinally 1-2 mm apart. Functional outcomes were assessed up to 6 weeks later using electromyography (EMG), electrical field stimulation (EFS), optogenetics, and by measuring colorectal motility. RESULTS Transplanted ENSCs formed nitrergic neurons in the nNOS-/- recipient colon. Multiple injections of ENSCs resulted in a significantly larger area of coverage compared to single injection alone and were associated with a marked improvement in colonic function, demonstrated by (1) increased colonic muscle activity by EMG recording, (2) faster rectal bead expulsion, and (3) increased fecal pellet output in vivo. Organ bath studies revealed direct neuromuscular communication by optogenetic stimulation of channelrhodopsin-expressing ENSCs and restoration of smooth muscle relaxation in response to EFS. CONCLUSIONS These results demonstrate that transplanted ENSCs can form effective neuromuscular connections and improve colonic motor function in a model of colonic dysmotility, and additionally reveal that multiple sites of cell delivery led to an improved response, paving the way for optimized clinical trial design.
Collapse
Affiliation(s)
- Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Ahmed Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Weikang Pan
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Shriya Srinivasan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Geoffrey de Couto
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Luis Rodriguez-Borlado
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Richard Myers
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Alan J Burns
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Sunardi M, Cirillo C. Mini-review: "Enteric glia functions in nervous tissue repair: Therapeutic target or tool?". Neurosci Lett 2023; 812:137360. [PMID: 37393007 DOI: 10.1016/j.neulet.2023.137360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
In the body, nerve tissue is not only present in the central nervous system, but also in the periphery. The enteric nervous system (ENS) is a highly organized intrinsic network of neurons and glial cells grouped to form interconnected ganglia. Glial cells in the ENS are a fascinating cell population: their neurotrophic role is well established, as well as their plasticity in specific circumstances. Gene expression profiling studies indicate that ENS glia retain neurogenic potential. The identification of neurogenic glial subtype(s) and the molecular basis of glia-derived neurogenesis may have profound biological and clinical implications. In this review, we discuss the potential of using gene-editing for ENS glia and cell transplantation as therapies for enteric neuropathies. Glia in the ENS: target or tool for nerve tissue repair?
Collapse
Affiliation(s)
- Mukhamad Sunardi
- Division of Neural Differentiation and Regeneration (NDR), Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan.
| | - Carla Cirillo
- Division of Neural Differentiation and Regeneration (NDR), Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan; Toulouse NeuroImaging Center (ToNIC), National Institute of Health and Medical Research (INSERM), Toulouse University Paul Sabatier, Toulouse, France.
| |
Collapse
|
14
|
Lefèvre MA, Soret R, Pilon N. Harnessing the Power of Enteric Glial Cells' Plasticity and Multipotency for Advancing Regenerative Medicine. Int J Mol Sci 2023; 24:12475. [PMID: 37569849 PMCID: PMC10419543 DOI: 10.3390/ijms241512475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
The enteric nervous system (ENS), known as the intrinsic nervous system of the gastrointestinal tract, is composed of a diverse array of neuronal and glial cell subtypes. Fascinating questions surrounding the generation of cellular diversity in the ENS have captivated ENS biologists for a considerable time, particularly with recent advancements in cell type-specific transcriptomics at both population and single-cell levels. However, the current focus of research in this field is predominantly restricted to the study of enteric neuron subtypes, while the investigation of enteric glia subtypes significantly lags behind. Despite this, enteric glial cells (EGCs) are increasingly recognized as equally important regulators of numerous bowel functions. Moreover, a subset of postnatal EGCs exhibits remarkable plasticity and multipotency, distinguishing them as critical entities in the context of advancing regenerative medicine. In this review, we aim to provide an updated overview of the current knowledge on this subject, while also identifying key questions that necessitate future exploration.
Collapse
Affiliation(s)
- Marie A. Lefèvre
- Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montreal, QC H3C 3P8, Canada;
- Centre D’excellence en Recherche Sur Les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Rodolphe Soret
- Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montreal, QC H3C 3P8, Canada;
- Centre D’excellence en Recherche Sur Les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Nicolas Pilon
- Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montreal, QC H3C 3P8, Canada;
- Centre D’excellence en Recherche Sur Les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Département de Pédiatrie, Université de Montréal, Montreal, QC H3T 1C5, Canada
| |
Collapse
|
15
|
Scharr M, Scherer S, Hirt B, Neckel PH. Dickkopf1 induces enteric neurogenesis and gliogenesis in vitro if apoptosis is evaded. Commun Biol 2023; 6:808. [PMID: 37532804 PMCID: PMC10397193 DOI: 10.1038/s42003-023-05072-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 06/25/2023] [Indexed: 08/04/2023] Open
Abstract
Neurogenesis in the postnatal enteric nervous system (ENS) is controversially discussed. Yet, deciphering the regenerative potential of the ENS is essential for our understanding and therapy of human enteric neuropathies. Dickkopf1 (DKK1) is a Wnt-antagonist and involved in the homeostasis of various tissues. We hypothesize that DKK1 could function as a negative regulator on the proliferation of ENS-progenitors in the postnatal gut of mice and human infants. Here, we provide evidence that DKK1 is expressed in the murine and human ENS. If applied to ENS-progenitors in vitro, DKK1 leads to an increased proliferation, however, followed by extensive apoptosis. Yet, once we block apoptosis, DKK1-stimulation markedly increases enteric neurogenesis in murine and human ENS-progenitors. Thus, DKK1 is a strong, ambivalent regulator of the ENS-progenitor cell pool in mice and humans. These results are fundamental steps to reshaping our understanding of the homeostasis of the ENS in health and disease.
Collapse
Affiliation(s)
- Melanie Scharr
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| | - Simon Scherer
- Department of Pediatric Surgery and Urology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Bernhard Hirt
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| | - Peter H Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
16
|
Heumüller-Klug S, Maurer K, Tapia-Laliena MÁ, Sticht C, Christmann A, Mörz H, Khasanov R, Wink E, Schulte S, Greffrath W, Treede RD, Wessel LM, Schäfer KH. Impact of cryopreservation on viability, gene expression and function of enteric nervous system derived neurospheres. Front Cell Dev Biol 2023; 11:1196472. [PMID: 37377739 PMCID: PMC10291272 DOI: 10.3389/fcell.2023.1196472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction: Impairment of both the central and peripheral nervous system is a major cause of mortality and disability. It varies from an affection of the brain to various types of enteric dysganglionosis. Congenital enteric dysganglionosis is characterized by the local absence of intrinsic innervation due to deficits in either migration, proliferation or differentiation of neural stem cells. Despite surgery, children's quality of life is reduced. Neural stem cell transplantation seems a promising therapeutic approach, requiring huge amounts of cells and multiple approaches to fully colonize the diseased areas completely. A combination of successful expansion and storage of neural stem cells is needed until a sufficient amount of cells is generated. This must be combined with suitable cell transplantation strategies, that cover all the area affected. Cryopreservation provides the possibility to store cells for long time, unfortunately with side effects, i.e., upon vitality. Methods: In this study we investigate the impact of different freezing and thawing protocols (M1-M4) upon enteric neural stem cell survival, protein and gene expression, and cell function. Results: Freezing enteric nervous system derived neurospheres (ENSdN) following slow-freezing protocols (M1-3) resulted in higher survival rates than flash-freezing (M4). RNA expression profiles were least affected by freezing protocols M1/2, whereas the protein expression of ENSdN remained unchanged after treatment with protocol M1 only. Cells treated with the most promising freezing protocol (M1, slow freezing in fetal calf serum plus 10% DMSO) were subsequently investigated using single-cell calcium imaging. Freezing of ENSdN did not alter the increase in intracellular calcium in response to a specific set of stimuli. Single cells could be assigned to functional subgroups according to response patterns and a significant shift towards cells responding to nicotine was observed after freezing. Discussion: The results demonstrate that cryopreservation of ENSdN is possible with reduced viability, only slight changes in protein/gene expression patterns and without an impact on the neuronal function of different enteric nervous system cell subtypes, with the exception of a subtle upregulation of cells expressing nicotinergic acetylcholine receptors. In summary, cryopreservation presents a good method to store sufficient amounts of enteric neural stem cells without neuronal impairment, in order to enable subsequent transplantation of cells into compromised tissues.
Collapse
Affiliation(s)
- Sabine Heumüller-Klug
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Kristina Maurer
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - María Á. Tapia-Laliena
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carsten Sticht
- Medical Research Centre Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anne Christmann
- AGENS, University of Applied Sciences Kaiserslautern Campus Zweibrücken, Kaiserslautern, Germany
| | - Handan Mörz
- Mannheim Center for Translational Neuroscience (MCTN), Department of Neurophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rasul Khasanov
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Elvira Wink
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Steven Schulte
- AGENS, University of Applied Sciences Kaiserslautern Campus Zweibrücken, Kaiserslautern, Germany
| | - Wolfgang Greffrath
- Mannheim Center for Translational Neuroscience (MCTN), Department of Neurophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rolf-Detlef Treede
- Mannheim Center for Translational Neuroscience (MCTN), Department of Neurophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lucas M. Wessel
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Karl-Herbert Schäfer
- AGENS, University of Applied Sciences Kaiserslautern Campus Zweibrücken, Kaiserslautern, Germany
| |
Collapse
|
17
|
Windster JD, Sacchetti A, Schaaf GJ, Bindels EM, Hofstra RM, Wijnen RM, Sloots CE, Alves MM. A combinatorial panel for flow cytometry-based isolation of enteric nervous system cells from human intestine. EMBO Rep 2023; 24:e55789. [PMID: 36852936 PMCID: PMC10074091 DOI: 10.15252/embr.202255789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 03/01/2023] Open
Abstract
Efficient isolation of neurons and glia from the human enteric nervous system (ENS) is challenging because of their rare and fragile nature. Here, we describe a staining panel to enrich ENS cells from the human intestine by fluorescence-activated cell sorting (FACS). We find that CD56/CD90/CD24 co-expression labels ENS cells with higher specificity and resolution than previous methods. Surprisingly, neuronal (CD24, TUBB3) and glial (SOX10) selective markers appear co-expressed by all ENS cells. We demonstrate that this contradictory staining pattern is mainly driven by neuronal fragments, either free or attached to glial cells, which are the most abundant cell types. Live neurons can be enriched by the highest CD24 and CD90 levels. By applying our protocol to isolate ENS cells for single-cell RNA sequencing, we show that these cells can be obtained with high quality, enabling interrogation of the human ENS transcriptome. Taken together, we present a selective FACS protocol that allows enrichment and discrimination of human ENS cells, opening up new avenues to study this complex system in health and disease.
Collapse
Affiliation(s)
- Jonathan D Windster
- Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Andrea Sacchetti
- Department of Pathology, Josephine Nefkens Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gerben J Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Eric Mj Bindels
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Robert Mw Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Rene Mh Wijnen
- Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Cornelius Ej Sloots
- Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Collier CA, Foncerrada S, Clevenger AJ, Shetty A, Raghavan SA. Acute Exposure to Pyridostigmine Bromide Disrupts Cholinergic Myenteric Neuroimmune Function in Mice. Adv Biol (Weinh) 2023; 7:e2200254. [PMID: 36802210 DOI: 10.1002/adbi.202200254] [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: 09/15/2022] [Revised: 11/30/2022] [Indexed: 02/21/2023]
Abstract
Gulf War Illness (GWI) results from chemical exposure during the Gulf War, with notable impacts on gastrointestinal motility. Due to the limited demographic impacted by this ailment, an in-depth investigation of the GWI has yielded little regarding the underlying pathophysiological mechanisms. Here, the hypothesis that exposure to pyridostigmine bromide (PB) results in severe enteric neuro-inflammation, that cascades to disruptions in colonic motility, is tested. The analyses are performed on male C57BL/6 mice that are treated with physiologically similar doses of PB given to GW veterans. When colonic motility is assessed, GWI colons have significantly reduced forces in response to acetylcholine or electrical field stimulation. GWI is also accompanied by high levels of pro-inflammatory cytokines and chemokines, associated with increased numbers of CD40+ pro-inflammatory macrophages within the myenteric plexus. Enteric neurons responsible for mediating colonic motility reside within the myenteric plexus, and PB exposure reduced their numbers. Significant smooth muscle hypertrophy is also observed due to increased inflammation. Together, the results show that PB exposure caused functional and anatomical dysfunction, promoting impaired motility within the colon. Achieving a greater understanding of the mechanisms of GWI will allow more refinement in therapeutic options that improve veterans' quality of life.
Collapse
Affiliation(s)
- Claudia A Collier
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 3120 TAMU, College Station, TX, 77843, USA
| | - Steven Foncerrada
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 3120 TAMU, College Station, TX, 77843, USA
| | - Abigail J Clevenger
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 3120 TAMU, College Station, TX, 77843, USA
| | - Ashok Shetty
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, 8447 Riverside Pkwy, Bryan, College Station, TX, 77807, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 3120 TAMU, College Station, TX, 77843, USA
| |
Collapse
|
19
|
Hotta R, Pan W, Bhave S, Nagy N, Stavely R, Ohkura T, Krishnan K, de Couto G, Myers R, Rodriguez-Borlado L, Burns AJ, Goldstein AM. Isolation, Expansion, and Endoscopic Delivery of Autologous Enteric Neuronal Stem Cells in Swine. Cell Transplant 2023; 32:9636897231215233. [PMID: 38049927 PMCID: PMC10697035 DOI: 10.1177/09636897231215233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/22/2023] [Accepted: 11/01/2023] [Indexed: 12/06/2023] Open
Abstract
The enteric nervous system (ENS) is an extensive network of neurons and glia within the wall of the gastrointestinal (GI) tract that regulates many essential GI functions. Consequently, disorders of the ENS due to developmental defects, inflammation, infection, or age-associated neurodegeneration lead to serious neurointestinal diseases. Despite the prevalence and severity of these diseases, effective treatments are lacking as they fail to directly address the underlying pathology. Neuronal stem cell therapy represents a promising approach to treating diseases of the ENS by replacing the absent or injured neurons, and an autologous source of stem cells would be optimal by obviating the need for immunosuppression. We utilized the swine model to address key questions concerning cell isolation, delivery, engraftment, and fate in a large animal relevant to human therapy. We successfully isolated neural stem cells from a segment of small intestine resected from 1-month-old swine. Enteric neuronal stem cells (ENSCs) were expanded as neurospheres that grew optimally in low-oxygen (5%) culture conditions. Enteric neuronal stem cells were labeled by lentiviral green fluorescent protein (GFP) transduction, then transplanted into the same swine from which they had been harvested. Endoscopic ultrasound was then utilized to deliver the ENSCs (10,000-30,000 neurospheres per animal) into the rectal wall. At 10 and 28 days following injection, autologously derived ENSCs were found to have engrafted within rectal wall, with neuroglial differentiation and no evidence of ectopic spreading. These findings strongly support the feasibility of autologous cell isolation and delivery using a clinically useful and minimally invasive technique, bringing us closer to first-in-human ENSC therapy for neurointestinal diseases.
Collapse
Affiliation(s)
- Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Weikang Pan
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Kumar Krishnan
- Division of Gastroenterology, Department of Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Geoffrey de Couto
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Richard Myers
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Luis Rodriguez-Borlado
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Alan J. Burns
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
20
|
Alhawaj AF. Stem cell-based therapy for hirschsprung disease, do we have the guts to treat? Gene Ther 2022; 29:578-587. [PMID: 34121091 PMCID: PMC9684071 DOI: 10.1038/s41434-021-00268-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023]
Abstract
Hirschsprung disease (HSCR) is a congenital anomaly of the colon that results from failure of enteric nervous system formation, leading to a constricted dysfunctional segment of the colon with variable lengths, and necessitating surgical intervention. The underlying pathophysiology includes a defect in neural crest cells migration, proliferation and differentiation, which are partially explained by identified genetic and epigenetic alterations. Despite the high success rate of the curative surgeries, they are associated with significant adverse outcomes such as enterocolitis, fecal soiling, and chronic constipation. In addition, some patients suffer from extensive lethal variants of the disease, all of which justify the need for an alternative cure. During the last 5 years, there has been considerable progress in HSCR stem cell-based therapy research. However, many major issues remain unsolved. This review will provide concise background information on HSCR, outline the future approaches of stem cell-based HSCR therapy, review recent key publications, discuss technical and ethical challenges the field faces prior to clinical translation, and tackle such challenges by proposing solutions and evaluating existing approaches to progress further.
Collapse
Affiliation(s)
- Ali Fouad Alhawaj
- Department of Haematology, UCL Cancer Institute, University College London, London, WC1E 6DD, United Kingdom.
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| |
Collapse
|
21
|
Enteric Neural Network Assembly Was Promoted by Basic Fibroblast Growth Factor and Vitamin A but Inhibited by Epidermal Growth Factor. Cells 2022; 11:cells11182841. [PMID: 36139415 PMCID: PMC9496868 DOI: 10.3390/cells11182841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/01/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Extending well beyond the original use of propagating neural precursors from the central nervous system and dorsal root ganglia, neurosphere medium (NSM) and self-renewal medium (SRM) are two distinct formulas with widespread popularity in enteric neural stem cell (ENSC) applications. However, it remains unknown what growth factors or nutrients are crucial to ENSC development, let alone whether the discrepancy in their components may affect the outcomes of ENSC culture. Dispersed enterocytes from murine fetal gut were nurtured in NSM, SRM or their modifications by selective component elimination or addition to assess their effects on ENSC development. NSM generated neuriteless neurospheres, whereas SRM, even deprived of chicken embryo extract, might wire ganglia together to assemble neural networks. The distinct outcomes came from epidermal growth factor, which inhibited enteric neuronal wiring in NSM. In contrast, basic fibroblast growth factor promoted enteric neurogenesis, gangliogenesis, and neuronal wiring. Moreover, vitamin A derivatives might facilitate neuronal maturation evidenced by p75 downregulation during ENSC differentiation toward enteric neurons to promote gangliogenesis and network assembly. Our results might help to better manipulate ENSC propagation and differentiation in vitro, and open a new avenue for the study of enteric neuronal neuritogenesis and synaptogenesis.
Collapse
|
22
|
Guo W, Zhang X, Zhai J, Xue J. The roles and applications of neural stem cells in spinal cord injury repair. Front Bioeng Biotechnol 2022; 10:966866. [PMID: 36105599 PMCID: PMC9465243 DOI: 10.3389/fbioe.2022.966866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/28/2022] [Indexed: 12/05/2022] Open
Abstract
Spinal cord injury (SCI), which has no current cure, places a severe burden on patients. Stem cell-based therapies are considered promising in attempts to repair injured spinal cords; such options include neural stem cells (NSCs). NSCs are multipotent stem cells that differentiate into neuronal and neuroglial lineages. This feature makes NSCs suitable candidates for regenerating injured spinal cords. Many studies have revealed the therapeutic potential of NSCs. In this review, we discuss from an integrated view how NSCs can help SCI repair. We will discuss the sources and therapeutic potential of NSCs, as well as representative pre-clinical studies and clinical trials of NSC-based therapies for SCI repair.
Collapse
Affiliation(s)
- Wen Guo
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xindan Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Jiliang Zhai
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| |
Collapse
|
23
|
Mueller JL, Stavely R, Hotta R, Goldstein AM. Peripheral nervous system: A promising source of neuronal progenitors for central nervous system repair. Front Neurosci 2022; 16:970350. [PMID: 35968387 PMCID: PMC9374275 DOI: 10.3389/fnins.2022.970350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
With a steadily aging population there is an increasing prevalence of neurological disorders. Given the lack of effective treatment strategies and a limited ability for the central nervous system (CNS) to regenerate endogenously, there is a critical need to better understand exogenous strategies for nervous system repair. Stem cell therapy offers a promising approach to promote the repair of neurologic tissue and function, however studies to date have been limited by various factors including challenges in harvesting donor cells from the CNS, ethical concerns regarding use of embryonic or fetal tissue, tumorigenic potential of induced pluripotent stem cells, and immune-mediated rejection of non-autologous cell sources. Here we review and propose two alternative sources of autologous cells derived from the peripheral nervous system (PNS) for CNS repair: enteric neuronal stem cells (ENSCs) and neural crest-derived Schwann cells found in subcutaneous adipose tissue (termed SAT-NSCs). ENSCs can be successfully isolated from the postnatal enteric nervous system, propagated in vitro, and transplanted successfully into models of CNS injury via both direct intracerebral injection and systemic tail vein injection. Similarly, SAT-NSCs can be readily isolated from both human and mouse adipose tissue and, although not yet utilized in models of CNS injury, have successfully been transplanted and restored function in models of colonic aganglionosis and gastroparesis. These unique sources of PNS-derived autologous cells offer an exciting option for stem cell therapies for the CNS as they have proven neurogenic potential and eliminate concerns around tumorigenic risk, ethical considerations, and immune-mediated rejection.
Collapse
|
24
|
Tullie L, Jones BC, De Coppi P, Li VSW. Building gut from scratch - progress and update of intestinal tissue engineering. Nat Rev Gastroenterol Hepatol 2022; 19:417-431. [PMID: 35241800 DOI: 10.1038/s41575-022-00586-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 12/18/2022]
Abstract
Short bowel syndrome (SBS), a condition defined by insufficient absorptive intestinal epithelium, is a rare disease, with an estimated prevalence up to 0.4 in 10,000 people. However, it has substantial morbidity and mortality for affected patients. The mainstay of treatment in SBS is supportive, in the form of intravenous parenteral nutrition, with the aim of achieving intestinal autonomy. The lack of a definitive curative therapy has led to attempts to harness innate developmental and regenerative mechanisms to engineer neo-intestine as an alternative approach to addressing this unmet clinical need. Exciting advances have been made in the field of intestinal tissue engineering (ITE) over the past decade, making a review in this field timely. In this Review, we discuss the latest advances in the components required to engineer intestinal grafts and summarize the progress of ITE. We also explore some key factors to consider and challenges to overcome when transitioning tissue-engineered intestine towards clinical translation, and provide the future outlook of ITE in therapeutic applications and beyond.
Collapse
Affiliation(s)
- Lucinda Tullie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK.,Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Brendan C Jones
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, DBC, Great Ormond Street Institute of Child Health, University College London, London, UK. .,Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, UK.
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
25
|
Hacene S, Le Friec A, Desmoulin F, Robert L, Colitti N, Fitremann J, Loubinoux I, Cirillo C. Present and future avenues of cell-based therapy for brain injury: The enteric nervous system as a potential cell source. Brain Pathol 2022; 32:e13105. [PMID: 35773942 PMCID: PMC9425017 DOI: 10.1111/bpa.13105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/09/2022] [Indexed: 01/01/2023] Open
Abstract
Cell therapy is a promising strategy in the field of regenerative medicine; however, several concerns limit the effective clinical use, namely a valid cell source. The gastrointestinal tract, which contains a highly organized network of nerves called the enteric nervous system (ENS), is a valuable reservoir of nerve cells. Together with neurons and neuronal precursor cells, it contains glial cells with a well described neurotrophic potential and a newly identified neurogenic one. Recently, enteric glia is looked at as a candidate for cell therapy in intestinal neuropathies. Here, we present the therapeutic potential of the ENS as cell source for brain repair, too. The example of stroke is introduced as a brain injury where cell therapy appears promising. This disease is the first cause of handicap in adults. The therapies developed in recent years allow a partial response to the consequences of the disease. The only prospect of recovery in the chronic phase is currently based on rehabilitation. The urgency to offer other treatments is therefore tangible. In the first part of the review, some elements of stroke pathophysiology are presented. An update on the available therapeutic strategies is provided, focusing on cell‐ and biomaterial‐based approaches. Following, the ENS is presented with its anatomical and functional characteristics, focusing on glial cells. The properties of these cells are depicted, with particular attention to their neurotrophic and, recently identified, neurogenic properties. Finally, preliminary data on a possible therapeutic approach combining ENS‐derived cells and a biomaterial are presented.
Collapse
Affiliation(s)
- Sirine Hacene
- National Veterinary School of Toulouse, University of Toulouse, Toulouse, France.,Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Alice Le Friec
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France.,Department of Biological and Chemical Engineering-Medical Biotechnology, Aarhus University, Aarhus, Denmark
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Lorenne Robert
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Nina Colitti
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Juliette Fitremann
- Laboratoire des IMRCP, CNRS UMR 5623, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Isabelle Loubinoux
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| | - Carla Cirillo
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse-Paul Sabatier, Toulouse, France
| |
Collapse
|
26
|
Gao Y, Zhang H, Wang Y, Han T, Jin J, Li J, Tang Y, Liu C. L-Cysteine Alleviates Myenteric Neuron Injury Induced by Intestinal Ischemia/Reperfusion via Inhibitin the Macrophage NLRP3-IL-1β Pathway. Front Pharmacol 2022; 13:899169. [PMID: 35754513 PMCID: PMC9213754 DOI: 10.3389/fphar.2022.899169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemia/reperfusion injury is a common pathophysiological process in the clinic. It causes various injuries, multiple organ dysfunction, and even death. There are several possible mechanisms about ischemia/reperfusion injury, but the influence on intestinal myenteric neurons and the underlying mechanism are still unclear. C57BL6/J mice were used to establish the ischemia/reperfusion model in vivo. Peritoneal macrophages were used for ATP depletion and hypoxia/reoxygenation experiment in vitro. L-cysteine, as the substrate of hydrogen sulfide, is involved in many physiological and pathological processes, including inflammation, metabolism, neuroprotection, and vasodilation. In the current study, we confirmed that intestinal ischemia/reperfusion led to the injury of myenteric neurons. From experiments in vitro and in vivo, we demonstrated that L-cysteine protected myenteric neurons from the injury. AOAA reversed the protective effect of L-cysteine. Also, L-cysteine played a protective role mainly by acting on intestinal macrophages via decreasing the expression of NLRP3, cleaved caspase-1, and mature IL-1β. L-cysteine increased cystathionine beta synthase and H2S produced by intestinal macrophages to protect myenteric mature neurons and enteric neural precursor cells from apoptosis. Moreover, the addition of IL-1β-neutralizing antibody alleviated the injury of myenteric neurons and enteric neural precursor cells caused by intestinal ischemia/reperfusion. Our study provided a new target for the protection of myenteric neurons in clinical intestinal ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Yifei Gao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Haojie Zhang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Yujin Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Ting Han
- Department of Gastroenterology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Jing Jin
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Yan Tang
- Department of Gastroenterology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Chuanyong Liu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China.,Provincial Key Lab of Mental Disorders, Shandong University, Jinan, China
| |
Collapse
|
27
|
Collier CA, Mendiondo C, Raghavan S. Tissue engineering of the gastrointestinal tract: the historic path to translation. J Biol Eng 2022; 16:9. [PMID: 35379299 PMCID: PMC8981633 DOI: 10.1186/s13036-022-00289-6] [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: 01/05/2022] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
The gastrointestinal (GI) tract is imperative for multiple functions including digestion, nutrient absorption, and timely waste disposal. The central feature of the gut is peristalsis, intestinal motility, which facilitates all of its functions. Disruptions in GI motility lead to sub-optimal GI function, resulting in a lower quality of life in many functional GI disorders. Over the last two decades, tissue engineering research directed towards the intestine has progressed rapidly due to advances in cell and stem-cell biology, integrative physiology, bioengineering and biomaterials. Newer biomedical tools (including optical tools, machine learning, and nuanced regenerative engineering approaches) have expanded our understanding of the complex cellular communication within the GI tract that lead to its orchestrated physiological function. Bioengineering therefore can be utilized towards several translational aspects: (i) regenerative medicine to remedy/restore GI physiological function; (ii) in vitro model building to mimic the complex physiology for drug and pharmacology testing; (iii) tool development to continue to unravel multi-cell communication networks to integrate cell and organ-level physiology. Despite the significant strides made historically in GI tissue engineering, fundamental challenges remain including the quest for identifying autologous human cell sources, enhanced scaffolding biomaterials to increase biocompatibility while matching viscoelastic properties of the underlying tissue, and overall biomanufacturing. This review provides historic perspectives for how bioengineering has advanced over time, highlights newer advances in bioengineering strategies, and provides a realistic perspective on the path to translation.
Collapse
Affiliation(s)
- Claudia A Collier
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA
| | - Christian Mendiondo
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA
| | - Shreya Raghavan
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA.
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
28
|
Mueller JL, Goldstein AM. The science of Hirschsprung disease: What we know and where we are headed. Semin Pediatr Surg 2022; 31:151157. [PMID: 35690468 DOI: 10.1016/j.sempedsurg.2022.151157] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The enteric nervous system (ENS) is a rich network of neurons and glial cells that comprise the gastrointestinal tract's intrinsic nervous system and are responsible for controlling numerous complex functions, including digestion, transit, secretion, barrier function, and maintenance of a healthy microbiome. Development of a functional ENS relies on the coordinated interaction between enteric neural crest-derived cells and their environment as the neural crest-derived cells migrate rostrocaudally along the embryonic gut mesenchyme. Congenital or acquired disruption of ENS development leads to various neurointestinal diseases. Hirschsprung disease is a congenital neurocristopathy, a disease of the neural crest. It is characterized by a variable length of distal colonic aganglionosis due to a failure in enteric neural crest-derived cell proliferation, migration, differentiation, and/or survival. In this review, we will review the science of Hirschsprung disease, targeting an audience of pediatric surgeons. We will discuss the basic biology of normal ENS development, as well as what goes awry in ENS development in Hirschsprung disease. We will review animal models that have been integral to studying this disease, as well as current hot topics and future research, including genetic risk profiling, stem cell therapy, non-invasive diagnostic techniques, single-cell sequencing techniques, and genotype-phenotype correlation.
Collapse
Affiliation(s)
- Jessica L Mueller
- Department of Pediatric Surgery, Massachusetts General Hospital, Massachusetts General Hospital for Children, Harvard Medical School, 55 Fruit St., WRN 1151, Boston, MA 02114, United States
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Massachusetts General Hospital for Children, Harvard Medical School, 55 Fruit St., WRN 1151, Boston, MA 02114, United States.
| |
Collapse
|
29
|
Jones BC, Shibuya S, Durkin N, De Coppi P. Regenerative medicine for childhood gastrointestinal diseases. Best Pract Res Clin Gastroenterol 2021; 56-57:101769. [PMID: 35331401 DOI: 10.1016/j.bpg.2021.101769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 01/31/2023]
Abstract
Several paediatric gastrointestinal diseases result in life-shortening organ failure. For many of these conditions, current therapeutic options are suboptimal and may not offer a cure. Regenerative medicine is an inter-disciplinary field involving biologists, engineers, and clinicians that aims to produce cell and tissue-based therapies to overcome organ failure. Exciting advances in stem cell biology, materials science, and bioengineering bring engineered gastrointestinal cell and tissue therapies to the verge of clinical trial. In this review, we summarise the requirements for bioengineered therapies, the possible sources of the various cellular and non-cellular components, and the progress towards clinical translation of oesophageal and intestinal tissue engineering to date.
Collapse
Affiliation(s)
- Brendan C Jones
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, United Kingdom
| | - Soichi Shibuya
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Natalie Durkin
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, United Kingdom
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, United Kingdom.
| |
Collapse
|
30
|
Mitxelena-Iribarren O, Olaizola C, Arana S, Mujika M. Versatile membrane-based microfluidic platform for in vitro drug diffusion testing mimicking in vivo environments. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 39:102462. [PMID: 34592426 DOI: 10.1016/j.nano.2021.102462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 07/22/2021] [Accepted: 08/16/2021] [Indexed: 01/18/2023]
Abstract
Mimicking the diffusion that drugs suffer through different body tissues before reaching their target is a challenge. In this work, a versatile membrane-based microfluidic platform was developed to allow for the identification of drugs that would keep their cytotoxic properties after diffusing through such a barrier. As an application case, this paper reports on a microfluidic device capable of mimicking the diffusion that free or encapsulated anticancer drugs would suffer in the intestine before reaching the bloodstream. It not only presents the successful fabrication results for the platform but also demonstrates the significant effect that the analyzed drugs have over the viability of osteosarcoma cells. This intestine-like microfluidic platform works as a tool to allow for the identification of drugs whose cytotoxic performance remains effective enough once they enter the bloodstream. Therefore, it allows for the prediction of the best treatment available for each patient in the battle against cancer.
Collapse
Affiliation(s)
- Oihane Mitxelena-Iribarren
- CEIT-Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain; Universidad de Navarra, Tecnun, Donostia-San Sebastián, Spain.
| | | | - Sergio Arana
- CEIT-Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain; Universidad de Navarra, Tecnun, Donostia-San Sebastián, Spain
| | - Maite Mujika
- CEIT-Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain; Universidad de Navarra, Tecnun, Donostia-San Sebastián, Spain
| |
Collapse
|
31
|
Mahadewa TGB, Mardhika PE, Awyono S, Putra MB, Saapang GS, Wiyanjana KDF, Putra KK, Natakusuma TISD, Ryalino C. Mesenteric Neural Stem Cell for Chronic Spinal Cord Injury: A Literature Review. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Spinal cord injury (SCI) is a common and potentially life-threatening condition with no established treatment to treat the primary injury. Mesenteric neural stem cell (NSC) therapy is a promising stem cell therapy to treat primary SCI in the chronic phase. We aimed to review the literature narratively to describe current evidence regarding mesenteric NSC in SCI. Primary SCI refers to tissue damage that occurs at the time of trauma that leads to the death of neuronal cells. In chronic SCI, the ability of neuronal regeneration is compromised by the development of gliotic scar. NSC is a stem cell therapy that targeted SCI in the chronic phase. Enteric NSC is one of the sources of NSC, and autologous gut harvesting in the appendix using endoscopic surgery provides a more straightforward and low-risk procedure. Intramedullary transplantation of stem cell with ultrasound guiding is administration technique which offers long-term regeneration. Mesenteric NSC is a promising stem cell therapy to treat chronic SCI with low risk and easier procedure to isolate cells compared to other NSC sources.
Collapse
|
32
|
Abstract
In the enteric nervous system, there exist a huge number of local intrinsic neurons, which control the gastrointestinal functions. Culture of enteric neurons provides a good model system for physiological, electrophysiological, and pharmacological studies. Here, we describe two methods to obtain sufficient enteric neurons from mouse myenteric plexuses by directly culturing primary neurons or inducing neuronal differentiation of enteric neural stem/progenitor cells.
Collapse
|
33
|
Yuan H, Hu H, Chen R, Mu W, Wang L, Li Y, Chen Y, Ding X, Xi Y, Mao S, Jiang M, Chen J, He Y, Wang L, Dong Y, Tou J, Chen W. Premigratory neural crest stem cells generate enteric neurons populating the mouse colon and regulating peristalsis in tissue-engineered intestine. Stem Cells Transl Med 2021; 10:922-938. [PMID: 33481357 PMCID: PMC8133337 DOI: 10.1002/sctm.20-0469] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/26/2020] [Accepted: 01/03/2021] [Indexed: 12/13/2022] Open
Abstract
Hirschsprung's disease (HSCR) is a common congenital defect. It occurs when bowel colonization by neural crest-derived enteric nervous system (ENS) precursors is incomplete during the first trimester of pregnancy. Several sources of candidate cells have been previously studied for their capacity to regenerate the ENS, including enteric neural crest stem cells (En-NCSCs) derived from native intestine or those simulated from human pluripotent stem cells (hPSCs). However, it is not yet known whether the native NCSCs other than En-NCSCs would have the potential of regenerating functional enteric neurons and producing neuron dependent motility under the intestinal environment. The present study was designed to determine whether premigratory NCSCs (pNCSCs), as a type of the nonenteric NCSCs, could form enteric neurons and mediate the motility. pNCSCs were firstly transplanted into the colon of adult mice, and were found to survive, migrate, differentiate into enteric neurons, and successfully integrate into the adult mouse colon. When the mixture of pNCSCs and human intestinal organoids was implanted into the subrenal capsule of nude mice and grown into the mature tissue-engineered intestine (TEI), the pNCSCs-derived neurons mediated neuron-dependent peristalsis of TEI. These results show that the pNCSCs that were previously assumed to not be induced by intestinal environment or cues can innervate the intestine and establish neuron-dependent motility. Future cell candidates for ENS regeneration may include nonenteric NCSCs.
Collapse
Affiliation(s)
- Huipu Yuan
- Institute of Translational Medicine, and Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouPeople's Republic of China
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Hui Hu
- Department of Laboratory MedicineHangzhou Medical CollegeHangzhouPeople's Republic of China
| | - Rui Chen
- Institute of Translational Medicine, and Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouPeople's Republic of China
- Department of Neonatal SurgeryChildren's Hospital, Zhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Wenbo Mu
- Institute of Translational Medicine, and Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouPeople's Republic of China
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Liangliang Wang
- Interdisciplinary Institutes of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang UniversityHangzhouPeople's Republic of China
| | - Ying Li
- Institute of Translational Medicine, and Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouPeople's Republic of China
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Yuelei Chen
- Cell Bank/Stem Cell BankInstitute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiPeople's Republic of China
| | - Xiaoyan Ding
- Cell Bank/Stem Cell BankInstitute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiPeople's Republic of China
| | - Yongmei Xi
- Institute of Genetics and Department of Genetics, Division of Human Reproduction and Developmental Genetics of the Women's HospitalZhejiang University School of MedicineHangzhouPeople's Republic of China
| | - ShanShan Mao
- Department of Internal NeurologyChildren's Hospital, Zhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Mizu Jiang
- Department of GastroenterologyZhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Jie Chen
- Department of Pediatric SurgeryXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems School of Mechanical EngineeringZhejiang UniversityHangzhouPeople's Republic of China
| | - Lang Wang
- Interdisciplinary Institutes of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang UniversityHangzhouPeople's Republic of China
| | - Yi Dong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education & Health CareEast China Normal UniversityShanghaiPeople's Republic of China
| | - Jinfa Tou
- Institute of Translational Medicine, and Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouPeople's Republic of China
- Department of Neonatal SurgeryChildren's Hospital, Zhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Wei Chen
- Institute of Translational Medicine, and Children's Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouPeople's Republic of China
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouPeople's Republic of China
- Department of NeurobiologyInstitute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of MedicineHangzhouPeople's Republic of China
| |
Collapse
|
34
|
Jevans B, James ND, Burnside E, McCann CJ, Thapar N, Bradbury EJ, Burns AJ. Combined treatment with enteric neural stem cells and chondroitinase ABC reduces spinal cord lesion pathology. Stem Cell Res Ther 2021; 12:10. [PMID: 33407795 PMCID: PMC7789480 DOI: 10.1186/s13287-020-02031-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) presents a significant challenge for the field of neurotherapeutics. Stem cells have shown promise in replenishing the cells lost to the injury process, but the release of axon growth-inhibitory molecules such as chondroitin sulfate proteoglycans (CSPGs) by activated cells within the injury site hinders the integration of transplanted cells. We hypothesised that simultaneous application of enteric neural stem cells (ENSCs) isolated from the gastrointestinal tract, with a lentivirus (LV) containing the enzyme chondroitinase ABC (ChABC), would enhance the regenerative potential of ENSCs after transplantation into the injured spinal cord. METHODS ENSCs were harvested from the GI tract of p7 rats, expanded in vitro and characterised. Adult rats bearing a contusion injury were randomly assigned to one of four groups: no treatment, LV-ChABC injection only, ENSC transplantation only or ENSC transplantation+LV-ChABC injection. After 16 weeks, rats were sacrificed and the harvested spinal cords examined for evidence of repair. RESULTS ENSC cultures contained a variety of neuronal subtypes suitable for replenishing cells lost through SCI. Following injury, transplanted ENSC-derived cells survived and ChABC successfully degraded CSPGs. We observed significant reductions in the injured tissue and cavity area, with the greatest improvements seen in the combined treatment group. ENSC-derived cells extended projections across the injury site into both the rostral and caudal host spinal cord, and ENSC transplantation significantly increased the number of cells extending axons across the injury site. Furthermore, the combined treatment resulted in a modest, but significant functional improvement by week 16, and we found no evidence of the spread of transplanted cells to ectopic locations or formation of tumours. CONCLUSIONS Regenerative effects of a combined treatment with ENSCs and ChABC surpassed either treatment alone, highlighting the importance of further research into combinatorial therapies for SCI. Our work provides evidence that stem cells taken from the adult gastrointestinal tract, an easily accessible source for autologous transplantation, could be strongly considered for the repair of central nervous system disorders.
Collapse
Affiliation(s)
- Benjamin Jevans
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
- Present Address: German Centre for Neurodegenerative diseases (DZNE), Bonn, Germany
| | - Nicholas D James
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Emily Burnside
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
- Neurogastroenterology and Motility Unit, Department of Gastroenterology, Great Ormond Street Hospital, London, UK
- Present Address: Department of Paediatric Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, Australia
| | - Elizabeth J Bradbury
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
- Present Address: Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Cambridge, USA.
| |
Collapse
|
35
|
Zhao Y, Ge X, Yu H, Kuil LE, Alves MM, Tian D, Huang Q, Chen X, Hofstra RMW, Gao Y. Inhibition of ROCK signaling pathway accelerates enteric neural crest cell-based therapy after transplantation in a rat hypoganglionic model. Neurogastroenterol Motil 2020; 32:e13895. [PMID: 32515097 DOI: 10.1111/nmo.13895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/23/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hirschsprung's disease (HSCR) is a congenital gastrointestinal disorder, characterized by enteric ganglia absence in part or entire of the colon, due to abnormal colonization and migration of enteric neural crest cells (ENCCs) during development. Currently, besides surgery which is the main therapy for HSCR, the potential of stem cell-based transplantation was investigated as an alternative option. Although promising, it has limitations, including poor survival, differentiation, and migration of the grafted cells. We hypothesized that modulation of extracellular factors during transplantation could promote ENCCs proliferation and migration, leading to increased transplantation efficiency. Considering that the RhoA/ROCK pathway is highly involved in cytoskeletal dynamics and neurite growth, our study explored the effect of inhibition of this pathway to improve the success of ENCCs transplantation. METHODS Enteric neural crest cells were isolated from rat embryos and labeled with a GFP-tag. Cell viability, apoptosis, differentiation, and migration assays were performed with and without RhoA/ROCK inhibition. Labeled ENCCs were transplanted into the muscle layer of an induced hypoganglionic rat model followed by intraperitoneal injections of ROCK inhibitor. The transplanted segments were collected 3 weeks after for histological analysis. KEY RESULTS Our results showed that inhibition of ROCK increased viable cell number, differentiation, and migration of ENCCs in vitro. Moreover, transplantation of labeled ENCCs into the hypoganglionic model showed enhanced distribution of grafted ENCCs, upon treatment with ROCK inhibitor. CONCLUSIONS AND INFERENCES ROCK inhibitors influence ENCCs growth and migration in vitro and in vivo, and should be considered to improve the efficiency of ENCCs transplantation.
Collapse
Affiliation(s)
- Yuying Zhao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Xin Ge
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hui Yu
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Donghao Tian
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qiang Huang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xinlin Chen
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ya Gao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
36
|
Dangerous Liaison: Helicobacter pylori, Ganglionitis, and Myenteric Gastric Neurons: A Histopathological Study. Anal Cell Pathol (Amst) 2019; 2019:3085181. [PMID: 32082967 PMCID: PMC7012220 DOI: 10.1155/2019/3085181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/17/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation induced by Helicobacter pylori (H. pylori) infection plays a major role in development of gastric cancer. However, recent findings suggested that progression of inflammation and neoplastic transformation in H. pylori infection are more complex than previously believed and could involve different factors that modulate gastric microenvironment and influence host-pathogen interaction. Among these factors, gastric myenteric plexus and its potential adaptive changes in H. pylori infection received little attention. This study is aimed at identifying the impact of H. pylori-associated gastritis on number and morphology of nerve cells in the stomach. The distribution of density, inflammation, and programmed cell death in neurons was immunohistochemically assessed in full-thickness archival tissue samples obtained from 40 patients with H. pylori infection who underwent surgery for gastric cancer and were compared with findings on samples collected from 40 age- and sex-matched subjects without bacteria. Overall, significant differences were noted between H. pylori-positive and H. pylori-negative patients. The analysis of tissue specimens obtained from those with infection revealed higher density and larger surface of the myenteric nervous plexus, as well as a significant increase in the number of gastric neuronal cell bodies and glial cells compared to controls. A predominant CD3-immunoreactive T cell infiltrate confined to the myenteric plexus was observed in infected subjects. The presence of mature B lymphocytes, plasma cells, and eosinophils was also noted, but to a lesser extent, within the ganglia. Myenteric ganglionitis was associated with degeneration and neuronal loss. Our results represent the first histopathological evidence supporting the hypothesis that H. pylori-induced gastric inflammation may induce morphological changes in myenteric gastric ganglia. These findings could help gain understanding of some still unclear aspects of pathogenesis of H. pylori infection, with the possibility of having broader implications for gastric cancer progression.
Collapse
|
37
|
Schmitteckert S, Mederer T, Röth R, Günther P, Holland-Cunz S, Metzger M, Samstag Y, Schröder-Braunstein J, Wabnitz G, Kurzhals S, Scheuerer J, Beretta CA, Lasitschka F, Rappold GA, Romero P, Niesler B. Postnatal human enteric neurospheres show a remarkable molecular complexity. Neurogastroenterol Motil 2019; 31:e13674. [PMID: 31318473 DOI: 10.1111/nmo.13674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/26/2019] [Accepted: 06/26/2019] [Indexed: 01/30/2023]
Abstract
BACKGROUND The enteric nervous system (ENS), a complex network of neurons and glial cells, coordinates major gastrointestinal functions. Impaired development or secondary aberrations cause severe enteric neuropathies. Neural crest-derived stem cells as well as enteric neuronal progenitor cells, which form enteric neurospheres, represent a promising tool to unravel molecular pathomechanisms and to develop novel therapy options. However, so far little is known about the detailed cellular composition and the proportional distribution of enteric neurospheres. Comprehensive knowledge will not only be essential for basic research but also for prospective cell replacement therapies to restore or to improve enteric neuronal dysfunction. METHODS Human enteric neurospheres were generated from three individuals with varying age. For detailed molecular characterization, nCounter target gene expression analyses focusing on stem, progenitor, neuronal, glial, muscular, and epithelial cell markers were performed. Corresponding archived paraffin-embedded individuals' specimens were analyzed accordingly. KEY RESULTS Our data revealed a remarkable molecular complexity of enteric neurospheres and archived specimens. Amongst the expression of multipotent stem cell, progenitor cell, neuronal, glial, muscle and epithelial cell markers, moderate levels for the pluripotency marker POU5F1 were observed. Furthermore, besides the interindividual variability, we identified highly distinct intraindividual expression profiles. CONCLUSIONS & INFERENCES Our results emphasize the assessment of molecular signatures to be essential for standardized use, optimization of experimental approaches, and elimination of potential risk factors, as the formation of tumors. Our study pipeline may serve as a blueprint implemented into the characterization procedure of enteric neurospheres for various future applications.
Collapse
Affiliation(s)
- Stefanie Schmitteckert
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Tanja Mederer
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ralph Röth
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,nCounter Core Facility, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Günther
- Division of Pediatric Surgery, Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Holland-Cunz
- Division of Pediatric Surgery, Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Pediatric Surgery, University Children's Hospital Basel, Basel, Switzerland
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research (ISC), Translational Centre Regenerative Therapies (TLC-RT) Wuerzburg, Wuerzburg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Guido Wabnitz
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Kurzhals
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jutta Scheuerer
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carlo A Beretta
- CellNetworks Math-Clinic Core Facility, Bioquant, Heidelberg University, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Philipp Romero
- Division of Pediatric Surgery, Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Niesler
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,nCounter Core Facility, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
38
|
Wnt Receptor Frizzled-4 as a Marker for Isolation of Enteric Neural Progenitors in Human Children. Cells 2019; 8:cells8080792. [PMID: 31366044 PMCID: PMC6721585 DOI: 10.3390/cells8080792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 11/20/2022] Open
Abstract
Identification and isolation of neural progenitor cells from the human enteric nervous system (ENS) is currently hampered by the lack of reliable, specific markers. Here, we define the Wnt-receptor frizzled-4 as a marker for the isolation of enteric neural progenitor cells derived from paediatric gut samples. We show that the Wnt-receptor frizzled-4 is expressed in the human colon and in Tunica muscularis-derived enterospheres. To obtain a purified culture, we carried out fluorescence-activated cell sorting (FACS) using PE-conjugated frizzled-4 antibodies. Frizzled-4positive cells gave rise to neurosphere-like bodies and ultimately differentiated into neurons as revealed by BrdU-proliferation assays and immunocytochemistry, whereas in frizzled-4negative cultures we did not detect any neuronal and glial cells. By using a patch-clamp approach, we also demonstrated the expression of functional sodium and potassium channels in frizzled-4positive cell cultures after differentiation in vitro.
Collapse
|
39
|
Grundmann D, Loris E, Maas-Omlor S, Schäfer KH. Enteric Neurogenesis During Life Span Under Physiological and Pathophysiological Conditions. Anat Rec (Hoboken) 2019; 302:1345-1353. [PMID: 30950581 DOI: 10.1002/ar.24124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/04/2019] [Accepted: 02/21/2019] [Indexed: 12/20/2022]
Abstract
The enteric nervous system (ENS) controls gastrointestinal key functions and is mainly characterized by two ganglionated plexus located in the gut wall: the myenteric plexus and the submucous plexus. The ENS harbors a high number and diversity of enteric neurons and glial cells, which generate neuronal circuitry to regulate intestinal physiology. In the past few years, the pivotal role of enteric neurons in the underlying mechanism of several intestinal diseases was revealed. Intestinal diseases are associated with neuronal death that could in turn compromise intestinal functionality. Enteric neurogenesis and regeneration is therefore a crucial aspect within the ENS and could be revealed not only during embryogenesis and early postnatal periods, but also in the adulthood. Enteric glia and/or enteric neural precursor/progenitor cells differentiate into enteric neurons, both under homeostatic and pathologic conditions beyond the perinatal period. The unique role of the intestinal microbiota and serotonin signaling in postnatal and adult neurogenesis has been shown by several studies in health and disease. In this review article, we will mainly focus on different recent studies, which advanced the concept of postnatal and adult ENS neurogenesis. Moreover, we will discuss the key factors and underlying mechanisms, which promote enteric neurogenesis. Finally, we will shortly describe neurogenesis of transplanted enteric neural progenitor cells. Anat Rec, 302:1345-1353, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David Grundmann
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrucken, Germany
| | - Eva Loris
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrucken, Germany
| | - Silke Maas-Omlor
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrucken, Germany
| | - Karl-Herbert Schäfer
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrucken, Germany.,Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
40
|
Zakhem E, Raghavan S, Suhar RA, Bitar KN. Bioengineering and regeneration of gastrointestinal tissue: where are we now and what comes next? Expert Opin Biol Ther 2019; 19:527-537. [PMID: 30880502 DOI: 10.1080/14712598.2019.1595579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The field of tissue engineering and regenerative medicine has been applied to the gastrointestinal (GI) tract for a couple decades. Several achievements have been accomplished that provide promising tools for treating diseases of the GI tract. AREAS COVERED The work described in this review covers the traditional aspect of using cells and scaffolds to replace parts of the tract. Several studies investigated different types of biomaterials and different types of cells. A more recent approach involved the use of gut-derived organoid units that can differentiate into all gut cell layers. The most recent approach introduced the use of organ-on-a-chip concept to understand the physiology and pathophysiology of the GI system. EXPERT OPINION The different approaches tackle the diseases of the GI tract from different perspectives. While all these different approaches provide a promising and encouraging future for this field, the translational aspect is yet to be studied.
Collapse
Affiliation(s)
- Elie Zakhem
- a Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine , Winston Salem , NC , USA.,b Section on Gastroenterology , Wake Forest School of Medicine , Winston Salem , NC , USA
| | - Shreya Raghavan
- c Department of Materials Science and Engineering , University of Michigan , Ann Arbor , MI , USA
| | - Riley A Suhar
- d Department of Materials Science and Engineering , Stanford University , Stanford , CA , USA
| | - Khalil N Bitar
- a Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine , Winston Salem , NC , USA.,b Section on Gastroenterology , Wake Forest School of Medicine , Winston Salem , NC , USA.,e Virginia Tech-Wake Forest School of Biomedical Engineering Sciences , Winston-Salem , NC , USA
| |
Collapse
|
41
|
Injury and stress responses of adult neural crest-derived cells. Dev Biol 2018; 444 Suppl 1:S356-S365. [DOI: 10.1016/j.ydbio.2018.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022]
|
42
|
Pesce M, Borrelli O, Saliakellis E, Thapar N. Gastrointestinal Neuropathies: New Insights and Emerging Therapies. Gastroenterol Clin North Am 2018; 47:877-894. [PMID: 30337038 DOI: 10.1016/j.gtc.2018.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The bewildering complexity of the enteric nervous system makes it susceptible to develop a wide array of motility disorders, collectively called enteric neuropathies. These gastrointestinal conditions are among the most challenging to manage, mainly given poor characterization of their etiopathophysiology and outcomes. Not surprisingly, therefore, targeted or curative therapies for enteric neuropathies are lacking and management is largely symptomatic. Nonetheless, recent advances in neurogastroenterology have witnessed improvements in established strategies, such as intestinal transplantation and the emergence of new treatments including novel drugs, electrical pacing, and manipulation of fecal microbiota, as well as stem cell and gene therapy.
Collapse
Affiliation(s)
- Marcella Pesce
- Neurogastroenterology and Motility Unit, Department of Pediatric Gastroenterology, Great Ormond Street Hospital, London WC1N 3JH, UK; Department of Clinical Medicine and Surgery, 'Federico II' University of Naples, Via Pansini 5, Naples 80131, Italy
| | - Osvaldo Borrelli
- Neurogastroenterology and Motility Unit, Department of Pediatric Gastroenterology, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Efstratios Saliakellis
- Neurogastroenterology and Motility Unit, Department of Pediatric Gastroenterology, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Nikhil Thapar
- Neurogastroenterology and Motility Unit, Department of Pediatric Gastroenterology, Great Ormond Street Hospital, London WC1N 3JH, UK; Stem Cells and Regenerative Medicine, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
43
|
McCann CJ, Thapar N. Enteric neural stem cell therapies for enteric neuropathies. Neurogastroenterol Motil 2018; 30:e13369. [PMID: 29707856 DOI: 10.1111/nmo.13369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Enteric neuropathies exist as a wide range of human disorders which impact on gastrointestinal motility. Current standard therapies for enteric neuropathies are limited to surgical resection or manipulation (eg, myotomy) of affected gut segments or medical management including both therapy (eg, prokinetic pharmacotherapy) and support such as parenteral nutrition. However, such treatments often result in poor prognosis and significant morbidity. The current limitations in treatment options for enteric neuropathies underline the need for alternative approaches to treat these devastating diseases. Recent advances have highlighted the potential of enteric neural stem cells as a possible treatment option for regenerative medicine, in such cases. PURPOSE The purpose of this review is to provide an up-to-date synopsis of the enteric neural stem cell research field. Here, we review in detail the initial characterization of enteric neural stem cells, early preclinical studies validating their use in murine models through to the most recent findings of therapeutic rescue of diseased gut tissue. We additionally pose a number of questions regarding these recent findings which will need to be addressed prior to clinical translation of this exciting cellular therapeutic.
Collapse
Affiliation(s)
- C J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - N Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
44
|
Jevans B, McCann CJ, Thapar N, Burns AJ. Transplanted enteric neural stem cells integrate within the developing chick spinal cord: implications for spinal cord repair. J Anat 2018; 233:592-606. [PMID: 30191559 DOI: 10.1111/joa.12880] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2018] [Indexed: 12/27/2022] Open
Abstract
Spinal cord injury (SCI) causes paralysis, multisystem impairment and reduced life expectancy, as yet with no cure. Stem cell therapy can potentially replace lost neurons, promote axonal regeneration and limit scar formation, but an optimal stem cell source has yet to be found. Enteric neural stem cells (ENSC) isolated from the enteric nervous system (ENS) of the gastrointestinal (GI) tract are an attractive source. Here, we used the chick embryo to assess the potential of ENSC to integrate within the developing spinal cord. In vitro, isolated ENSC formed extensive cell connections when co-cultured with spinal cord (SC)-derived cells. Further, qRT-PCR analysis revealed the presence of TuJ1+ neurons, S100+ glia and Sox10+ stem cells within ENSC neurospheres, as well as expression of key neuronal subtype genes, at levels comparable to SC tissue. Following ENSC transplantation to an ablated region of chick embryo SC, donor neurons were found up to 12 days later. These neurons formed bridging connections within the SC injury zone, aligned along the anterior/posterior axis, and were immunopositive for TuJ1. These data provide early proof of principle support for the use of ENSCs for SCI, and encourage further research into their potential for repair.
Collapse
Affiliation(s)
- Benjamin Jevans
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.,Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Cambridge, MA, USA
| |
Collapse
|
45
|
Liu W, Zhang L, Wu R. Enteric Neural Stem Cells Expressing Insulin-Like Growth Factor 1: A Novel Cellular Therapy for Hirschsprung's Disease in Mouse Model. DNA Cell Biol 2018; 37:642-648. [PMID: 29792527 DOI: 10.1089/dna.2017.4060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lijuan Zhang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Rongde Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
46
|
Obermayr F, Seitz G. Recent developments in cell-based ENS regeneration - a short review. Innov Surg Sci 2018; 3:93-99. [PMID: 31579772 PMCID: PMC6604576 DOI: 10.1515/iss-2018-0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 02/15/2018] [Indexed: 12/19/2022] Open
Abstract
Therapeutic options to treat neurogenic motility disorders of the gastrointestinal tract are usually limited to symptomatic treatment. The capacity of the enteric nervous system (ENS) to regenerate and the fact that progenitor cells of the enteric nervous system reside in the postnatal and adult gut led to the idea to develop cell-based strategies to treat ENS related disorders. This short review focuses on recent developments in cell-based ENS regeneration, discussing advantages and disadvantages of various cell sources, functional impact of transplanted cells and highlights the challenges of translation of small animal studies to human application.
Collapse
Affiliation(s)
- Florian Obermayr
- Department of Pediatric Surgery, University Hospital Marburg, UKGM, Baldingerstrasse, 35043 Marburg, Germany, Phone: +49-6421-5864117
| | - Guido Seitz
- Department of Pediatric Surgery, University Hospital Marburg, UKGM, Marburg, Germany
| |
Collapse
|
47
|
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: 86] [Impact Index Per Article: 10.8] [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
|
48
|
Zhang Y, Seid K, Obermayr F, Just L, Neckel PH. Activation of Wnt Signaling Increases Numbers of Enteric Neurons Derived From Neonatal Mouse and Human Progenitor Cells. Gastroenterology 2017; 153:154-165.e9. [PMID: 28359679 DOI: 10.1053/j.gastro.2017.03.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/17/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS Neural stem and progenitor cells from the enteric nervous system (ENS) might serve as a source of cells for treatment of neurogastrointestinal disorders. Before we can use these cells, we must increase our understanding of the signaling mechanisms that regulate proliferation and differentiation. We systematically evaluated the effects of canonical Wnt signaling on proliferation and differentiation of cultured ENS progenitor cells from neonatal mice and humans. METHODS We isolated ENS progenitors from tunica muscularis of the small intestine of newborn (postnatal day 0) wild-type C57BL/6 mice as well as from Wnt1-Cre2 reporter mice. We also obtained intestinal tissue samples from infants (2 and 7 months old) undergoing surgery for imperforate anus or focal intestinal perforation and isolated ENS cells. ENS cells were cultured under proliferation conditions leading to formation of 3-dimensional spheres, which we activated with Wnt3a and SB216763 in order to activate the β-catenin-dependent canonical Wnt pathway. We used immunoblot and quantitative polymerase chain reaction to evaluate the molecular response to Wnt stimuli and immunohistochemistry, proliferation, and cell death assays to identify new neurons. RESULTS In proliferating enterospheres derived from ENS progenitor cells, we verified the expression of Wnt receptors frizzled 1-10 and the co-receptors low-density lipoprotein receptor-related proteins 5 and 6. Pharmacologic stimulation with Wnt agonists led to intracellular accumulation of Wnt-dependent β-catenin and up-regulated expression of known Wnt target genes axin2, lef1, and lgr5. Activation of the canonical Wnt pathway promoted growth of ENS cell spheres during cell expansion and increased the number of newborn neurons derived from mouse and human progenitor cells. CONCLUSIONS In studies of human and mouse ENS progenitors, we found activation of the Wnt signaling pathway to promote neurogenesis of the ENS in vitro. The neurogenic effect of Wnt agonists on ENS progenitors supports their use in generation of cell pools for autologous cell replacement therapies.
Collapse
Affiliation(s)
- Ying Zhang
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| | - Karin Seid
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| | - Florian Obermayr
- Department of Pediatric Surgery, University Children's Hospital Tübingen, Germany
| | - Lothar Just
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| | - Peter H Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Neurointestinal diseases are increasingly recognized as causes of significant gastrointestinal morbidity in children. This review highlights the most common pediatric enteric neuropathies and their diagnosis and management, emphasizing insights and discoveries from the most recent literature available. RECENT FINDINGS The embryologic and histopathologic causes of enteric neuropathies are varied. They range from congenital aganglionosis in Hirschsprung disease, to autoimmune-mediated loss of neuronal subtypes in esophageal achalasia and Chagas disease, to degenerative neuropathies in some cases of chronic intestinal pseudo-obstruction and gastroparesis. Increased awareness of the clinical presentation and diagnostic evaluation of these conditions is essential as it allows for earlier initiation of treatment and improved outcomes. Most current therapies, which include medical management, neurostimulation, and operative intervention, aim to minimize the symptoms caused by these conditions. The evidence base for many of these treatments in children is poor, and multiinstitutional prospective studies are needed. An innovative therapy on the horizon involves using neuronal stem cell transplantation to treat the underlying disorder by replacing the missing or damaged neurons in these diseases. SUMMARY Although recent advances in basic and clinical neurogastroenterology have significantly improved our awareness and understanding of enteric neuropathies, the efficacy of current treatment approaches is limited. The development of novel therapies, including pharmacologic modulators of neurointestinal function, neurostimulation to enhance gut motility, and neuronal cell-based therapies, is essential to improve the long-term outcomes in children with these disorders.
Collapse
Affiliation(s)
- Maggie L. Westfal
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Neurointestinal Health, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
50
|
Kulkarni S, Micci MA, Leser J, Shin C, Tang SC, Fu YY, Liu L, Li Q, Saha M, Li C, Enikolopov G, Becker L, Rakhilin N, Anderson M, Shen X, Dong X, Butte MJ, Song H, Southard-Smith EM, Kapur RP, Bogunovic M, Pasricha PJ. Adult enteric nervous system in health is maintained by a dynamic balance between neuronal apoptosis and neurogenesis. Proc Natl Acad Sci U S A 2017; 114:E3709-E3718. [PMID: 28420791 PMCID: PMC5422809 DOI: 10.1073/pnas.1619406114] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
According to current dogma, there is little or no ongoing neurogenesis in the fully developed adult enteric nervous system. This lack of neurogenesis leaves unanswered the question of how enteric neuronal populations are maintained in adult guts, given previous reports of ongoing neuronal death. Here, we confirm that despite ongoing neuronal cell loss because of apoptosis in the myenteric ganglia of the adult small intestine, total myenteric neuronal numbers remain constant. This observed neuronal homeostasis is maintained by new neurons formed in vivo from dividing precursor cells that are located within myenteric ganglia and express both Nestin and p75NTR, but not the pan-glial marker Sox10. Mutation of the phosphatase and tensin homolog gene in this pool of adult precursors leads to an increase in enteric neuronal number, resulting in ganglioneuromatosis, modeling the corresponding disorder in humans. Taken together, our results show significant turnover and neurogenesis of adult enteric neurons and provide a paradigm for understanding the enteric nervous system in health and disease.
Collapse
Affiliation(s)
- Subhash Kulkarni
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555
| | - Jenna Leser
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Changsik Shin
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, PA 17033
| | | | - Ya-Yuan Fu
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Liansheng Liu
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Qian Li
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Monalee Saha
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Cuiping Li
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Grigori Enikolopov
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Center for Developmental Genetics, Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794
| | - Laren Becker
- Division of Gastroenterology, Stanford University School of Medicine, Stanford, CA 94305
| | - Nikolai Rakhilin
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853
| | - Michael Anderson
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Dermatology, Center for Sensory Biology, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205
- Howard Hughes Medical Institute, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205
| | - Xiling Shen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853
| | - Xinzhong Dong
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Department of Dermatology, Center for Sensory Biology, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205
- Howard Hughes Medical Institute, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205
| | - Manish J Butte
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Hongjun Song
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Institute for Cellular Engineering, Department of Neurology, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205
| | | | - Raj P Kapur
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA 98105
| | - Milena Bogunovic
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, PA 17033
| | - Pankaj J Pasricha
- Center for Neurogastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205;
| |
Collapse
|