1
|
Cao N, Merchant W, Gautron L. Limited evidence for anatomical contacts between intestinal GLP-1 cells and vagal neurons in male mice. Sci Rep 2024; 14:23666. [PMID: 39390033 PMCID: PMC11467209 DOI: 10.1038/s41598-024-74000-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
The communication between intestinal Glucagon like peptide 1 (GLP-1)-producing cells and the peripheral nervous system has garnered renewed interest considering the availability of anti-obesity and anti-diabetic approaches targeting GLP-1 signaling. While it is well-established that intestinal GLP-1 cells can exert influence through paracrine mechanisms, recent evidence suggests the possible existence of synaptic-like connections between GLP-1 cells and peripheral neurons, including those of the vagus nerve. In this study, using a reporter Phox2b-Cre-Tomato mouse model and super-resolution confocal microscopy, we demonstrated that vagal axons made apparent contacts with less than 0.5% of GLP-1 cells. Moreover, immunohistochemistry combined with super-resolution confocal microscopy revealed abundant post-synaptic density 95 (PSD-95) immunoreactivity within the enteric plexus of the lower intestines of C57/BL6 mice, with virtually none in its mucosa. Lastly, utilizing RNAScope in situ hybridization in the lower intestines of mice, we observed that GLP-1 cells expressed generic markers of secretory cells such as Snap25 and Nefm, but neither synaptic markers such as Syn1 and Nrxn2, nor glutamatergic markers such as Slc17a7. Through theoretical considerations and a critical review of the literature, we concluded that intestinal GLP-1 cells primarily communicate with vagal neurons through paracrine mechanisms, rather than synaptic-like contacts.
Collapse
Affiliation(s)
- Newton Cao
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Warda Merchant
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Laurent Gautron
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
2
|
Dopeso H, Rodrigues P, Cartón-García F, Macaya I, Bilic J, Anguita E, Jing L, Brotons B, Vivancos N, Beà L, Sánchez-Martín M, Landolfi S, Hernandez-Losa J, Ramon y Cajal S, Nieto R, Vicario M, Farre R, Schwartz S, van Ijzendoorn SC, Kobayashi K, Martinez-Barriocanal Á, Arango D. RhoA downregulation in the murine intestinal epithelium results in chronic Wnt activation and increased tumorigenesis. iScience 2024; 27:109400. [PMID: 38523777 PMCID: PMC10959657 DOI: 10.1016/j.isci.2024.109400] [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: 04/26/2023] [Revised: 12/23/2023] [Accepted: 02/28/2024] [Indexed: 03/26/2024] Open
Abstract
Rho GTPases are molecular switches regulating multiple cellular processes. To investigate the role of RhoA in normal intestinal physiology, we used a conditional mouse model overexpressing a dominant negative RhoA mutant (RhoAT19N) in the intestinal epithelium. Although RhoA inhibition did not cause an overt phenotype, increased levels of nuclear β-catenin were observed in the small intestinal epithelium of RhoAT19N mice, and the overexpression of multiple Wnt target genes revealed a chronic activation of Wnt signaling. Elevated Wnt signaling in RhoAT19N mice and intestinal organoids did not affect the proliferation of intestinal epithelial cells but significantly interfered with their differentiation. Importantly, 17-month-old RhoAT19N mice showed a significant increase in the number of spontaneous intestinal tumors. Altogether, our results indicate that RhoA regulates the differentiation of intestinal epithelial cells and inhibits tumor initiation, likely through the control of Wnt signaling, a key regulator of proliferation and differentiation in the intestine.
Collapse
Affiliation(s)
- Higinio Dopeso
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Paulo Rodrigues
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Fernando Cartón-García
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Irati Macaya
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Josipa Bilic
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Estefanía Anguita
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Li Jing
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Bruno Brotons
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Núria Vivancos
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Laia Beà
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Manuel Sánchez-Martín
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, 37007 Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Stefania Landolfi
- Translational Molecular Pathology, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Javier Hernandez-Losa
- Translational Molecular Pathology, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Santiago Ramon y Cajal
- Translational Molecular Pathology, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Rocío Nieto
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - María Vicario
- Digestive System Research Unit, Vall d’Hebron University Hospital Research Institute (VHIR), 08035 Barcelona, Spain
| | - Ricard Farre
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven 3000, Belgium
| | - Simo Schwartz
- Group of Drug Delivery and Targeting, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Clinical Biochemistry Department, Vall d'Hebron University Hospital, 08035 Barcelona, Spain
| | - Sven C.D. van Ijzendoorn
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Águeda Martinez-Barriocanal
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| |
Collapse
|
3
|
Singh PNP, Madha S, Leiter AB, Shivdasani RA. Cell and chromatin transitions in intestinal stem cell regeneration. Genes Dev 2022; 36:684-698. [PMID: 35738677 PMCID: PMC9296007 DOI: 10.1101/gad.349412.122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/06/2022] [Indexed: 11/25/2022]
Abstract
The progeny of intestinal stem cells (ISCs) dedifferentiate in response to ISC attrition. The precise cell sources, transitional states, and chromatin remodeling behind this activity remain unclear. In the skin, stem cell recovery after injury preserves an epigenetic memory of the damage response; whether similar memories arise and persist in regenerated ISCs is not known. We addressed these questions by examining gene activity and open chromatin at the resolution of single Neurog3-labeled mouse intestinal crypt cells, hence deconstructing forward and reverse differentiation of the intestinal secretory (Sec) lineage. We show that goblet, Paneth, and enteroendocrine cells arise by multilineage priming in common precursors, followed by selective access at thousands of cell-restricted cis-elements. Selective ablation of the ISC compartment elicits speedy reversal of chromatin and transcriptional features in large fractions of precursor and mature crypt Sec cells without obligate cell cycle re-entry. ISC programs decay and reappear along a cellular continuum lacking discernible discrete interim states. In the absence of gross tissue damage, Sec cells simply reverse their forward trajectories, without invoking developmental or other extrinsic programs, and starting chromatin identities are effectively erased. These findings identify strikingly plastic molecular frameworks in assembly and regeneration of a self-renewing tissue.
Collapse
Affiliation(s)
- Pratik N P Singh
- Department of Medical Oncology, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shariq Madha
- Department of Medical Oncology, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Andrew B Leiter
- Division of Gastroenterology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
4
|
Roberts A, Phuah P, Cheng S, Murphy KG. Targeting Enteroendocrine Cells to Treat Metabolic Disease. COMPREHENSIVE PHARMACOLOGY 2022:344-372. [DOI: 10.1016/b978-0-12-820472-6.00068-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Capdevila C, Trifas M, Miller J, Anderson T, Sims PA, Yan KS. Cellular origins and lineage relationships of the intestinal epithelium. Am J Physiol Gastrointest Liver Physiol 2021; 321:G413-G425. [PMID: 34431400 PMCID: PMC8560372 DOI: 10.1152/ajpgi.00188.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 01/31/2023]
Abstract
Knowledge of the development and hierarchical organization of tissues is key to understanding how they are perturbed in injury and disease, as well as how they may be therapeutically manipulated to restore homeostasis. The rapidly regenerating intestinal epithelium harbors diverse cell types and their lineage relationships have been studied using numerous approaches, from classical label-retaining and genetic lineage tracing methods to novel transcriptome-based annotations. Here, we describe the developmental trajectories that dictate differentiation and lineage specification in the intestinal epithelium. We focus on the most recent single-cell RNA-sequencing (scRNA-seq)-based strategies for understanding intestinal epithelial cell lineage relationships, underscoring how they have refined our view of the development of this tissue and highlighting their advantages and limitations. We emphasize how these technologies have been applied to understand the dynamics of intestinal epithelial cells in homeostatic and injury-induced regeneration models.
Collapse
Affiliation(s)
- Claudia Capdevila
- Columbia Stem Cell Initiative, Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
| | - Maria Trifas
- Columbia Stem Cell Initiative, Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
| | - Jonathan Miller
- Columbia Stem Cell Initiative, Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
| | - Troy Anderson
- Columbia Stem Cell Initiative, Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, New York
| | - Kelley S Yan
- Columbia Stem Cell Initiative, Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
6
|
Watkins JD, Koumanov F, Gonzalez JT. Protein- and Calcium-Mediated GLP-1 Secretion: A Narrative Review. Adv Nutr 2021; 12:2540-2552. [PMID: 34192748 PMCID: PMC8634310 DOI: 10.1093/advances/nmab078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Glucagon-like peptide 1 (GLP-1) is an incretin hormone produced in the intestine that is secreted in response to nutrient exposure. GLP-1 potentiates glucose-dependent insulin secretion from the pancreatic β cells and promotes satiety. These important actions on glucose metabolism and appetite have led to widespread interest in GLP-1 receptor agonism. Typically, this involves pharmacological GLP-1 mimetics or targeted inhibition of dipeptidyl peptidase-IV, the enzyme responsible for GLP-1 degradation. However, nutritional strategies provide a widely available, cost-effective alternative to pharmacological strategies for enhancing hormone release. Recent advances in nutritional research have implicated the combined ingestion of protein and calcium with enhanced endogenous GLP-1 release, which is likely due to activation of receptors with high affinity and/or sensitivity for amino acids and calcium. Specifically targeting these receptors could enhance gut hormone secretion, thus providing a new therapeutic option. This narrative review provides an overview of the latest research on protein- and calcium-mediated GLP-1 release with an emphasis on human data, and a perspective on potential mechanisms that link potent GLP-1 release to the co-ingestion of protein and calcium. In light of these recent findings, potential future research directions are also presented.
Collapse
Affiliation(s)
- Jonathan D Watkins
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, United Kingdom
| | - Françoise Koumanov
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, United Kingdom
| | | |
Collapse
|
7
|
Abstract
Glucagon like peptide-1 (GLP-1), a peptide hormone from the intestinal tract, plays a central role in the coordination of postprandial glucose homeostasis through actions on insulin secretion, food intake and gut motility. GLP-1 forms the basis for a variety of current drugs for the treatment of type 2 diabetes and obesity, as well as new agents currently being developed. Here, we provide a concise overview of the core physiology of GLP-1 secretion and action, and the role of the peptide in human health, disease and therapeutics.
Collapse
Affiliation(s)
- Fiona M Gribble
- Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - Frank Reimann
- Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
8
|
Zhang Y, Zeng F, Han X, Weng J, Gao Y. Lineage tracing: technology tool for exploring the development, regeneration, and disease of the digestive system. Stem Cell Res Ther 2020; 11:438. [PMID: 33059752 PMCID: PMC7559019 DOI: 10.1186/s13287-020-01941-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Lineage tracing is the most widely used technique to track the migration, proliferation, and differentiation of specific cells in vivo. The currently available gene-targeting technologies have been developing for decades to study organogenesis, tissue injury repairing, and tumor progression by tracing the fates of individual cells. Recently, lineage tracing has expanded the platforms available for disease model establishment, drug screening, cell plasticity research, and personalized medicine development in a molecular and cellular biology perspective. Lineage tracing provides new views for exploring digestive organ development and regeneration and techniques for digestive disease causes and progression. This review focuses on the lineage tracing technology and its application in digestive diseases.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Fanhong Zeng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Xu Han
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Jun Weng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China. .,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China. .,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Li Y, Chen M, Hu J, Sheng R, Lin Q, He X, Guo M. Volumetric Compression Induces Intracellular Crowding to Control Intestinal Organoid Growth via Wnt/β-Catenin Signaling. Cell Stem Cell 2020; 28:63-78.e7. [PMID: 33053374 DOI: 10.1016/j.stem.2020.09.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/26/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
Enormous amounts of essential intracellular events are crowdedly packed inside picoliter-sized cellular space. However, the significance of the physical properties of cells remains underappreciated because of a lack of evidence of how they affect cellular functionalities. Here, we show that volumetric compression regulates the growth of intestinal organoids by modifying intracellular crowding and elevating Wnt/β-catenin signaling. Intracellular crowding varies upon stimulation by different types of extracellular physical/mechanical cues and leads to significant enhancement of Wnt/β-catenin signaling by stabilizing the LRP6 signalosome. By enhancing intracellular crowding using osmotic and mechanical compression, we show that expansion of intestinal organoids was facilitated through elevated Wnt/β-catenin signaling and greater intestinal stem cell (ISC) self-renewal. Our results provide an entry point for understanding how intracellular crowdedness functions as a physical regulator linking extracellular physical cues with intracellular signaling and potentially facilitate the design of engineering approaches for expansion of stem cells and organoids.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maorong Chen
- F. M. Kirby Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jiliang Hu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ren Sheng
- F. M. Kirby Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; College of Life and Health Science, Northeastern University, Shenyang, Liaoning, 110004, China
| | - Qirong Lin
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xi He
- F. M. Kirby Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
10
|
Intestinal stem cells heterogeneity and clonal dominance during aging: two faces of the same coin? Mech Ageing Dev 2020; 189:111247. [PMID: 32505859 DOI: 10.1016/j.mad.2020.111247] [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] [Received: 01/23/2020] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 11/20/2022]
Abstract
Intestinal epithelium undergoes dysfunctions and diseases over time with an exponential increase in the elderly population. Recent studies reported that the intestinal stem cells (ISCs) show a functional decline during aging and a lack of an appropriate cell identity control. Increase of cell-to-cell heterogeneity is a hallmark of aging tissues and organs, however there is little experimental evidence with regard to the cell heterogeneity of the ISCs. On the other hand, the ISCs continuously experience a niche clonality process that diminishes the initial cell heterogeneity over time. In this review, we discuss the latest findings on these topics focusing on the potential mechanisms driving intestinal stem cell heterogeneity and clonality during aging.
Collapse
|
11
|
Abstract
The existence of "active" and "reserve" stem cell populations in the intestinal epithelium has been debated since 1977. Now in Cell Stem Cell, Murata et al. (2020) show that all intestinal regeneration arises from daughter cell dedifferentiation, marking the coming-of-age of the regenerative stem cell plasticity model.
Collapse
Affiliation(s)
- Simon J Leedham
- Intestinal Stem Cell Biology Laboratory, Wellcome Centre Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK.
| |
Collapse
|
12
|
Billing LJ, Larraufie P, Lewis J, Leiter A, Li J, Lam B, Yeo GS, Goldspink DA, Kay RG, Gribble FM, Reimann F. Single cell transcriptomic profiling of large intestinal enteroendocrine cells in mice - Identification of selective stimuli for insulin-like peptide-5 and glucagon-like peptide-1 co-expressing cells. Mol Metab 2019; 29:158-169. [PMID: 31668387 PMCID: PMC6812004 DOI: 10.1016/j.molmet.2019.09.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/21/2022] Open
Abstract
Objective Enteroendocrine cells (EECs) of the large intestine, found scattered in the epithelial layer, are known to express different hormones, with at least partial co-expression of different hormones in the same cell. Here we aimed to categorize colonic EECs and to identify possible targets for selective recruitment of hormones. Methods Single cell RNA-sequencing of sorted enteroendocrine cells, using NeuroD1-Cre x Rosa26-EYFP mice, was used to cluster EECs from the colon and rectum according to their transcriptome. G-protein coupled receptors differentially expressed across clusters were identified, and, as a proof of principle, agonists of Agtr1a and Avpr1b were tested as candidate EEC secretagogues in vitro and in vivo. Results EECs from the large intestine separated into 7 clear clusters, 4 expressing higher levels of Tph1 (enzyme required for serotonin (5-HT) synthesis; enterochromaffin cells), 2 enriched for Gcg (encoding glucagon-like peptide-1, GLP-1, L-cells), and the 7th expressing somatostatin (D-cells). Restricted analysis of L-cells identified 4 L-cell sub-clusters, exhibiting differential expression of Gcg, Pyy (Peptide YY), Nts (neurotensin), Insl5 (insulin-like peptide 5), Cck (cholecystokinin), and Sct (secretin). Expression profiles of L- and enterochromaffin cells revealed the clustering to represent gradients along the crypt-surface (cell maturation) and proximal-distal gut axes. Distal colonic/rectal L-cells differentially expressed Agtr1a and the ligand angiotensin II was shown to selectively increase GLP-1 and PYY release in vitro and GLP-1 in vivo. Conclusion EECs in the large intestine exhibit differential expression gradients along the crypt-surface and proximal-distal axes. Distal L-cells can be differentially stimulated by targeting receptors such as Agtr1a. Large intestinal enteroendocrine cells group into subclusters by single cell RNAseq. Enteroendocrine-cell subclusters differ along crypt-surface and longitudinal axes. L-cells differ longitudinally by production of NTS (proximal colon) or INSL5 (rectum). INSL5-positive cells express distinct GPCRs enabling cluster-specific stimulation. Targeted stimulation of INSL5-producing L-cells elevates plasma GLP-1 and PYY in vivo.
Collapse
Affiliation(s)
- Lawrence J Billing
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Pierre Larraufie
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Jo Lewis
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Andrew Leiter
- Division of Gastroenterology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Joyce Li
- Division of Gastroenterology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Brian Lam
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Giles Sh Yeo
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Deborah A Goldspink
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Richard G Kay
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Fiona M Gribble
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom.
| | - Frank Reimann
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science (IMS) & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom.
| |
Collapse
|
13
|
Gupta R, Bhatt LK, Johnston TP, Prabhavalkar KS. Colon cancer stem cells: Potential target for the treatment of colorectal cancer. Cancer Biol Ther 2019; 20:1068-1082. [PMID: 31050577 DOI: 10.1080/15384047.2019.1599660] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite incessant research, colon cancer still is one of the most common causes of fatalities in both men and women worldwide. Also, nearly 50% of patients with colorectal cancer show tumor recurrence. Recent investigations have highlighted the involvement of colon cancer stem cells (CCSCs) in cancer relapse and chemoresistance. CCSCs deliver a significant protumorigenic niche through persistent overexpression of self-renewal capabilities. Moreover, CSCs cross network with stromal cells, immune infiltrates, and cyotokine-chemokine, which potentiate their aggressive proliferative potential. Targeting CCSCs through small molecule inhibitors, miRNAs, and monoclonal antibodies (mAbs) in in vivo studies has generated compelling evidence for the effectiveness of these various treatments. This review effectively compiles the role of CCSC surface markers and dysregulated and/or upregulated pathways in the pathogenesis of colorectal cancer that can be used to target CCSCs for effective colorectal cancer treatment.
Collapse
Affiliation(s)
- Riya Gupta
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Lokesh Kumar Bhatt
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Thomas P Johnston
- b Division of Pharmacology and Pharmaceutical Sciences , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Kedar S Prabhavalkar
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| |
Collapse
|
14
|
Combination treatment of podophyllotoxin and rutin promotes mouse Lgr5+ ve intestinal stem cells survival against lethal radiation injury through Wnt signaling. Apoptosis 2019; 24:326-340. [DOI: 10.1007/s10495-019-01519-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Abstract
Enteroendocrine cells (EECs) are sensory cells of the gastrointestinal tract. Most EECs reside in the mucosal lining of the stomach or intestine and sense food in the gut lumen. Food signals stimulate the release of hormones into the paracellular space where they either act locally or are taken up into the blood and circulate to distant organs. It recently was recognized that many EECs possess basal processes known as neuropods that not only contain hormones but also connect to nerves. This review describes how neuropods contribute to EEC function beyond typical hormonal actions. For example, gastrointestinal hormones not only act on distant organs, but, through neuropods, some act locally to stimulate other mucosal cells such as intestinal stem cells, enterocytes, or other EECs. With the recent discovery that EECs communicate directly with enteric nerves, EECs not only have the ability to sense food and bacteria in the gastrointestinal tract, but can communicate these signals directly to the nervous system.
Collapse
|
16
|
Parham LR, Williams PA, Chatterji P, Whelan KA, Hamilton KE. RNA regulons are essential in intestinal homeostasis. Am J Physiol Gastrointest Liver Physiol 2019; 316:G197-G204. [PMID: 30520692 PMCID: PMC6383383 DOI: 10.1152/ajpgi.00403.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal epithelial cells are among the most rapidly proliferating cell types in the human body. There are several different subtypes of epithelial cells, each with unique functional roles in responding to the ever-changing environment. The epithelium's ability for rapid and customized responses to environmental changes requires multitiered levels of gene regulation. An emerging paradigm in gastrointestinal epithelial cells is the regulation of functionally related mRNA families, or regulons, via RNA-binding proteins (RBPs). RBPs represent a rapid and efficient mechanism to regulate gene expression and cell function. In this review, we will provide an overview of intestinal epithelial RBPs and how they contribute specifically to intestinal epithelial stem cell dynamics. In addition, we will highlight key gaps in knowledge in the global understanding of RBPs in gastrointestinal physiology as an opportunity for future studies.
Collapse
Affiliation(s)
- Louis R. Parham
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Patrick A. Williams
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Priya Chatterji
- 2Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kelly A. Whelan
- 3Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania,4Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kathryn E. Hamilton
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Miguel-Aliaga I, Jasper H, Lemaitre B. Anatomy and Physiology of the Digestive Tract of Drosophila melanogaster. Genetics 2018; 210:357-396. [PMID: 30287514 PMCID: PMC6216580 DOI: 10.1534/genetics.118.300224] [Citation(s) in RCA: 279] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal tract has recently come to the forefront of multiple research fields. It is now recognized as a major source of signals modulating food intake, insulin secretion and energy balance. It is also a key player in immunity and, through its interaction with microbiota, can shape our physiology and behavior in complex and sometimes unexpected ways. The insect intestine had remained, by comparison, relatively unexplored until the identification of adult somatic stem cells in the Drosophila intestine over a decade ago. Since then, a growing scientific community has exploited the genetic amenability of this insect organ in powerful and creative ways. By doing so, we have shed light on a broad range of biological questions revolving around stem cells and their niches, interorgan signaling and immunity. Despite their relatively recent discovery, some of the mechanisms active in the intestine of flies have already been shown to be more widely applicable to other gastrointestinal systems, and may therefore become relevant in the context of human pathologies such as gastrointestinal cancers, aging, or obesity. This review summarizes our current knowledge of both the formation and function of the Drosophila melanogaster digestive tract, with a major focus on its main digestive/absorptive portion: the strikingly adaptable adult midgut.
Collapse
Affiliation(s)
- Irene Miguel-Aliaga
- Medical Research Council London Institute of Medical Sciences, Imperial College London, W12 0NN, United Kingdom
| | - Heinrich Jasper
- Buck Institute for Research on Aging, Novato, California 94945-1400
- Immunology Discovery, Genentech, Inc., San Francisco, California 94080
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, École polytechnique fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
18
|
Maisonneuve C, Irrazabal T, Martin A, Girardin SE, Philpott DJ. The Impact of the Gut Microbiome on Colorectal Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050240] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Charles Maisonneuve
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada;,
| | - Thergiory Irrazabal
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada;,
| | - Alberto Martin
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada;,
| | - Stephen E. Girardin
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada;,
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Dana J. Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada;,
| |
Collapse
|
19
|
Fu DJ, Miller AD, Southard TL, Flesken-Nikitin A, Ellenson LH, Nikitin AY. Stem Cell Pathology. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:71-92. [PMID: 29059010 DOI: 10.1146/annurev-pathol-020117-043935] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rapid advances in stem cell biology and regenerative medicine have opened new opportunities for better understanding disease pathogenesis and the development of new diagnostic, prognostic, and treatment approaches. Many stem cell niches are well defined anatomically, thereby allowing their routine pathological evaluation during disease initiation and progression. Evaluation of the consequences of genetic manipulations in stem cells and investigation of the roles of stem cells in regenerative medicine and pathogenesis of various diseases such as cancer require significant expertise in pathology for accurate interpretation of novel findings. Therefore, there is an urgent need for developing stem cell pathology as a discipline to facilitate stem cell research and regenerative medicine. This review provides examples of anatomically defined niches suitable for evaluation by diagnostic pathologists, describes neoplastic lesions associated with them, and discusses further directions of stem cell pathology.
Collapse
Affiliation(s)
- Dah-Jiun Fu
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Andrew D Miller
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Teresa L Southard
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Andrea Flesken-Nikitin
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| | - Lora H Ellenson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Alexander Yu Nikitin
- Department of Biomedical Sciences and Cornell Stem Cell Program, Cornell University, Ithaca, New York 14853, USA;
| |
Collapse
|
20
|
Spatial organization of a model 15-member human gut microbiota established in gnotobiotic mice. Proc Natl Acad Sci U S A 2017; 114:E9105-E9114. [PMID: 29073107 DOI: 10.1073/pnas.1711596114] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Knowledge of the spatial organization of the gut microbiota is important for understanding the physical and molecular interactions among its members. These interactions are thought to influence microbial succession, community stability, syntrophic relationships, and resiliency in the face of perturbations. The complexity and dynamism of the gut microbiota pose considerable challenges for quantitative analysis of its spatial organization. Here, we illustrate an approach for addressing this challenge, using (i) a model, defined 15-member consortium of phylogenetically diverse, sequenced human gut bacterial strains introduced into adult gnotobiotic mice fed a polysaccharide-rich diet, and (ii) in situ hybridization and spectral imaging analysis methods that allow simultaneous detection of multiple bacterial strains at multiple spatial scales. Differences in the binding affinities of strains for substrates such as mucus or food particles, combined with more rapid replication in a preferred microhabitat, could, in principle, lead to localized clonally expanded aggregates composed of one or a few taxa. However, our results reveal a colonic community that is mixed at micrometer scales, with distinct spatial distributions of some taxa relative to one another, notably at the border between the mucosa and the lumen. Our data suggest that lumen and mucosa in the proximal colon should be conceptualized not as stratified compartments but as components of an incompletely mixed bioreactor. Employing the experimental approaches described should allow direct tests of whether and how specified host and microbial factors influence the nature and functional contributions of "microscale" mixing to the dynamic operations of the microbiota in health and disease.
Collapse
|
21
|
Chairatana P, Nolan EM. Defensins, lectins, mucins, and secretory immunoglobulin A: microbe-binding biomolecules that contribute to mucosal immunity in the human gut. Crit Rev Biochem Mol Biol 2017; 52:45-56. [PMID: 27841019 PMCID: PMC5233583 DOI: 10.1080/10409238.2016.1243654] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/19/2016] [Accepted: 09/28/2016] [Indexed: 02/08/2023]
Abstract
In the intestine, the mucosal immune system plays essential roles in maintaining homeostasis between the host and microorganisms, and protecting the host from pathogenic invaders. Epithelial cells produce and release a variety of biomolecules into the mucosa and lumen that contribute to immunity. In this review, we focus on a subset of these remarkable host-defense factors - enteric α-defensins, select lectins, mucins, and secretory immunoglobulin A - that have the capacity to bind microbes and thereby contribute to barrier function in the human gut. We provide an overview of the intestinal epithelium, describe specialized secretory cells named Paneth cells, and summarize our current understanding of the biophysical and functional properties of these select microbe-binding biomolecules. We intend for this compilation to complement prior reviews on intestinal host-defense factors, highlight recent advances in the field, and motivate investigations that further illuminate molecular mechanisms as well as the interplay between these molecules and microbes.
Collapse
Affiliation(s)
- Phoom Chairatana
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
22
|
Chairatana P, Nolan EM. Defensins, lectins, mucins, and secretory immunoglobulin A: microbe-binding biomolecules that contribute to mucosal immunity in the human gut. Crit Rev Biochem Mol Biol 2016. [PMID: 27841019 DOI: 10,1080/10409238.2016.124365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the intestine, the mucosal immune system plays essential roles in maintaining homeostasis between the host and microorganisms, and protecting the host from pathogenic invaders. Epithelial cells produce and release a variety of biomolecules into the mucosa and lumen that contribute to immunity. In this review, we focus on a subset of these remarkable host-defense factors - enteric α-defensins, select lectins, mucins, and secretory immunoglobulin A - that have the capacity to bind microbes and thereby contribute to barrier function in the human gut. We provide an overview of the intestinal epithelium, describe specialized secretory cells named Paneth cells, and summarize our current understanding of the biophysical and functional properties of these select microbe-binding biomolecules. We intend for this compilation to complement prior reviews on intestinal host-defense factors, highlight recent advances in the field, and motivate investigations that further illuminate molecular mechanisms as well as the interplay between these molecules and microbes.
Collapse
Affiliation(s)
- Phoom Chairatana
- a Department of Chemistry , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Elizabeth M Nolan
- a Department of Chemistry , Massachusetts Institute of Technology , Cambridge , MA , USA
| |
Collapse
|
23
|
Fu L, Shi YB. The Sox transcriptional factors: Functions during intestinal development in vertebrates. Semin Cell Dev Biol 2016; 63:58-67. [PMID: 27567710 DOI: 10.1016/j.semcdb.2016.08.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/19/2016] [Accepted: 08/24/2016] [Indexed: 12/28/2022]
Abstract
The intestine has long been studied as a model for adult stem cells due to the life-long self-renewal of the intestinal epithelium through the proliferation of the adult intestinal stem cells. Recent evidence suggests that the formation of adult intestinal stem cells in mammals takes place during the thyroid hormone-dependent neonatal period, also known as postembryonic development, which resembles intestinal remodeling during frog metamorphosis. Studies on the metamorphosis in Xenopus laevis have revealed that many members of the Sox family, a large family of DNA binding transcription factors, are upregulated in the intestinal epithelium during the formation and/or proliferation of the intestinal stem cells. Similarly, a number of Sox genes have been implicated in intestinal development and pathogenesis in mammals. Futures studies are needed to determine the expression and potential involvement of this important gene family in the development of the adult intestinal stem cells. These include the analyses of the expression and regulation of these and other Sox genes during postembryonic development in mammals as well as functional investigations in both mammals and amphibians by using the recently developed gene knockout technologies.
Collapse
Affiliation(s)
- Liezhen Fu
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., Bethesda, MD, 20892, United States
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., Bethesda, MD, 20892, United States.
| |
Collapse
|
24
|
Spherical nucleic acid targeting microRNA-99b enhances intestinal MFG-E8 gene expression and restores enterocyte migration in lipopolysaccharide-induced septic mice. Sci Rep 2016; 6:31687. [PMID: 27538453 PMCID: PMC4990839 DOI: 10.1038/srep31687] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/25/2016] [Indexed: 12/18/2022] Open
Abstract
Milk fat globule-EGF factor 8 (MFG-E8) maintains the intestinal homeostasis by enhancing enterocyte migration and attenuating inflammation. We previously reported that sepsis is associated with down-regulation of intestinal MFG-E8 and impairment of enterocyte migration. Here, we showed that impairment of intestinal epithelial cell migration occurred in lipopolysaccharide (LPS)-induced septic mice. Treatment of RAW264.7 cells (a murine macrophage-like cell line) with LPS increased expression of miR-99b, a microRNA that is predicted to target mouse MFG-E8 3′UTR. Using a luciferase assay, we showed that miR-99b mimic suppressed the activity of a reporter containing MFG-E8 3′UTR. This suggests the role of miR-99b in inhibition of MFG-E8 gene expression. In addition, we developed an anti-miR99b spherical nucleic acid nanoparticle conjugate (SNA-NCanti-miR99b). Treatment of both naïve and LPS-challenged cells with SNA-NCanti-miR99b enhanced MFG-E8 expression in the cells. Administration of SNA-NCanti-miR99b rescued intestinal MFG-E8 expression in LPS-induced septic mice and attenuated LPS inhibitory effects on intestinal epithelial cell migration along the crypt-villus axis. Collectively, our study suggests that LPS represses MFG-E8 expression and disrupts enterocyte migration via a miR-99b dependent mechanism. Furthermore, this work shows that SNA-NCanti-miR99b is a novel nanoparticle-conjugate capable of rescuing MFG-E8 gene expression and maintaining intestinal epithelial homeostasis in sepsis.
Collapse
|
25
|
Smith NR, Gallagher AC, Wong MH. Defining a stem cell hierarchy in the intestine: markers, caveats and controversies. J Physiol 2016; 594:4781-90. [PMID: 26864260 DOI: 10.1113/jp271651] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/06/2016] [Indexed: 12/22/2022] Open
Abstract
The past decade has appreciated rapid advance in identifying the once elusive intestinal stem cell (ISC) populations that fuel the continual renewal of the epithelial layer. This advance was largely driven by identification of novel stem cell marker genes, revealing the existence of quiescent, slowly- and active-cycling ISC populations. However, a critical barrier for translating this knowledge to human health and disease remains elucidating the functional interplay between diverse stem cell populations. Currently, the precise hierarchical and regulatory relationships between these ISC populations are under intense scrutiny. The classical theory of a linear hierarchy, where quiescent and slowly-cycling stem cells self-renew but replenish an active-cycling population, is well established in other rapidly renewing tissues such as the haematopoietic system. Efforts to definitively establish a similar stem cell hierarchy within the intestinal epithelium have yielded conflicting results, been difficult to interpret, and suggest non-conventional alternatives to a linear hierarchy. While these new and potentially paradigm-shifting discoveries are intriguing, the field will require development of a number of critical tools, including highly specific stem cell marker genes along with more rigorous experimental methodologies, to delineate the complex cellular relationships within this dynamic organ system.
Collapse
Affiliation(s)
- Nicholas R Smith
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Alexandra C Gallagher
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Melissa H Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, 97239, USA.,OHSU Stem Cell Center, Oregon Health and Science University, Portland, OR, 97239, USA
| |
Collapse
|
26
|
Hansen CF, Thymann T, Andersen AD, Holst JJ, Hartmann B, Hilsted L, Langhorn L, Jelsing J, Sangild PT. Rapid gut growth but persistent delay in digestive function in the postnatal period of preterm pigs. Am J Physiol Gastrointest Liver Physiol 2016; 310:G550-60. [PMID: 26822913 PMCID: PMC4836131 DOI: 10.1152/ajpgi.00221.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/22/2016] [Indexed: 01/31/2023]
Abstract
Preterm infants often tolerate full enteral nutrition a few weeks after birth but it is not known how this is related to gut maturation. Using pigs as models, we hypothesized that intestinal structure and digestive function are similar in preterm and term individuals at 3-4 wk after birth and that early enteral nutrition promotes maturation. Preterm or term cesarean-delivered pigs were fed total parenteral nutrition, or partial enteral nutrition [Enteral (Ent), 16-64 ml·kg(-1)·day(-1) of bovine colostrum] for 5 days, followed by full enteral milk feeding until day 26 The intestine was collected for histological and biochemical analyses at days 0, 5, and 26 (n = 8-12 in each of 10 treatment groups). Intestinal weight (relative to body weight) was reduced in preterm pigs at 0-5 days but ENT feeding stimulated the mucosal volume and peptidase activities. Relative to term pigs, mucosal volume remained reduced in preterm pigs until 26 days although plasma glucagon-like peptide 2 (GLP-2) and glucose-dependent insulin-trophic peptide (GIP) levels were increased. Preterm pigs also showed reduced hexose absorptive capacity and brush-border enzyme (sucrase, maltase) activities at 26 days, relative to term pigs. Intestinal structure shows a remarkable growth adaptation in the first week after preterm birth, especially with enteral nutrition, whereas some digestive functions remain immature until at least 3-4 wk. It is important to identify feeding regimens that stimulate intestinal maturation in the postnatal period of preterm infants because some intestinal functions may show long-term developmental delay.
Collapse
Affiliation(s)
- Carl Frederik Hansen
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | - Thomas Thymann
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | | | - Jens Juul Holst
- 2Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Bolette Hartmann
- 2Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Linda Hilsted
- 4Department of Clinical Biochemistry, Copenhagen University Hospital, Cophenhagen Denmark; and
| | - Louise Langhorn
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | | | - Per Torp Sangild
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark; ,5Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Denmark
| |
Collapse
|
27
|
Reimann F, Gribble FM. Mechanisms underlying glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 secretion. J Diabetes Investig 2016; 7 Suppl 1:13-9. [PMID: 27186350 PMCID: PMC4854499 DOI: 10.1111/jdi.12478] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/17/2015] [Accepted: 01/06/2016] [Indexed: 01/10/2023] Open
Abstract
The incretin hormones, glucose‐dependent insulinotropic peptide and glucagon‐like peptide‐1, are secreted from intestinal K‐ and L cells, respectively, with the former being most abundant in the proximal small intestine, whereas the latter increase in number towards the distal gut. Although an overlap between K‐ and L cells can be observed immunohistochemically or in murine models expressing fluorescent markers under the control of the two hormone promoters, the majority (>80%) of labeled cells seems to produce only one of these hormones. Transcriptomic analysis showed a close relationship between small intestinal K‐ and L cells, and glucose sensing mechanisms appear similar in both cell types with a predominant role of electrogenic glucose uptake through sodium‐coupled glucose transporter 1. Similarly, both cell types produce the long‐chain fatty acid sensing G‐protein‐coupled receptors, FFAR1 (GPR40) and FFAR4 (GPR120), but differ in the expression/functionality of other lipid sensing receptors. GPR119 and FFAR2/3, for example, have clearly documented roles in glucagon‐like peptide‐1 secretion, whereas agonists for the endocannabinoid receptor type 1 have been found to show largely selective inhibition of glucose‐dependent insulinotropic peptide secretion. In conclusion, although K‐ and L cell populations overlap and share key molecular nutrient‐sensing mechanisms, subtle differences between the responsiveness of the different cell types might be exploited to differentially modulate glucose‐dependent insulinotropic peptide or glucagon‐like peptide‐1 secretion.
Collapse
Affiliation(s)
- Frank Reimann
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit WT-MRC Institute of Metabolic Science Addenbrooke's Hospital Cambridge UK
| | - Fiona M Gribble
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit WT-MRC Institute of Metabolic Science Addenbrooke's Hospital Cambridge UK
| |
Collapse
|
28
|
Bloemendaal ALA, Buchs NC, George BD, Guy RJ. Intestinal stem cells and intestinal homeostasis in health and in inflammation: A review. Surgery 2016; 159:1237-48. [PMID: 26936524 DOI: 10.1016/j.surg.2016.01.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/06/2016] [Accepted: 01/23/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The human intestine is a complex group of organs, highly specialized in processing food and providing nutrients to the body. It is under constant threat from microbials and toxins and has therefore developed a number of protective mechanisms. One important mechanism is the constant shedding of epithelial cells into the lumen; another is the production and maintenance of a double-layered mucous boundary in which there is continuous sampling of the luminal microbiota and a persistent presence of antimicrobial enzymes. However, the gut needs commensal bacteria to effectively break down food into absorbable nutrients, which necessitates constant communication between the luminal bacteria and the intestinal immune cells in homeostasis. Disruption of homeostasis, for whatever reason, will give rise to (chronic) inflammation. DISCUSSION Both medical and surgical management of this disruption is discussed.
Collapse
Affiliation(s)
- Alexander L A Bloemendaal
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom.
| | - Nicolas C Buchs
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| | - Bruce D George
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| | - Richard J Guy
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
29
|
Okada M, Wen L, Miller TC, Su D, Shi YB. Molecular and cytological analyses reveal distinct transformations of intestinal epithelial cells during Xenopus metamorphosis. Cell Biosci 2015; 5:74. [PMID: 26719790 PMCID: PMC4696227 DOI: 10.1186/s13578-015-0065-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/22/2015] [Indexed: 12/15/2022] Open
Abstract
Background The thyroid hormone (T3)-induced formation of adult intestine during amphibian metamorphosis resembles the maturation of the mammalian intestine during postembryonic development, the period around birth when plasma T3 level peaks. This process involves de novo formation of adult intestinal stem cells as well as the removal of the larval epithelial cells through apoptosis. Earlier studies have revealed a number of cytological and molecular markers for the epithelial cells undergoing different changes during metamorphosis. However, the lack of established double labeling has made it difficult to ascertain the identities of the metamorphosing epithelial cells. Results Here, we carried out different double-staining with a number of cytological and molecular markers during T3-induced and natural metamorphosis in Xenopus laevis. Our studies demonstrated conclusively that the clusters of proliferating cells in the epithelium at the climax of metamorphosis are undifferentiated epithelial cells and express the well-known adult intestinal stem cell marker gene Lgr5. We further show that the adult stem cells and apoptotic larval epithelial cells are distinct epithelial cells during metamorphosis. Conclusions Our findings suggest that morphologically identical larval epithelial cells choose two alternative paths: programmed cell death or dedifferentiation to form adult stem cells, in response to T3 during metamorphosis with apoptosis occurring prior to the formation of the proliferating adult stem cell clusters (islets).
Collapse
Affiliation(s)
- Morihiro Okada
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., Bethesda, MD 20892 USA
| | - Luan Wen
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., Bethesda, MD 20892 USA
| | - Thomas C Miller
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., Bethesda, MD 20892 USA.,Meso-Scale Discovery, Rockville, MD USA
| | - Dan Su
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., Bethesda, MD 20892 USA.,Oncology Department, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., Bethesda, MD 20892 USA
| |
Collapse
|
30
|
Abstract
The enteroendocrine system orchestrates how the body responds to the ingestion of foods, employing a diversity of hormones to fine-tune a wide range of physiological responses both within and outside the gut. Recent interest in gut hormones has surged with the realization that they modulate glucose tolerance and food intake through a variety of mechanisms, and such hormones are therefore excellent therapeutic candidates for the treatment of diabetes and obesity. Characterizing the roles and functions of different enteroendocrine cells is an essential step in understanding the physiology, pathophysiology, and therapeutics of the gut-brain-pancreas axis.
Collapse
Affiliation(s)
- Fiona M Gribble
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, and Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; ,
| | - Frank Reimann
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, and Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; ,
| |
Collapse
|
31
|
Okada M, Miller TC, Fu L, Shi YB. Direct Activation of Amidohydrolase Domain-Containing 1 Gene by Thyroid Hormone Implicates a Role in the Formation of Adult Intestinal Stem Cells During Xenopus Metamorphosis. Endocrinology 2015; 156:3381-93. [PMID: 26086244 PMCID: PMC4541628 DOI: 10.1210/en.2015-1190] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The T3-dependent anuran metamorphosis resembles postembryonic development in mammals, the period around birth when plasma T3 levels peak. In particular, the remodeling of the intestine during metamorphosis mimics neonatal intestinal maturation in mammals when the adult intestinal epithelial self-renewing system is established. We have been using intestinal metamorphosis to investigate how the organ-specific adult stem cells are formed during vertebrate development. Early studies in Xenopus laevis have shown that this process involves complete degeneration of the larval epithelium and de novo formation of adult stem cells. A tissue-specific microarray analysis of intestinal gene expression during Xenopus laevis metamorphosis has identified a number of candidate stem cell genes. Here we have carried out detailed analyses of one such gene, amidohydrolase domain containing 1 (AMDHD1) gene, which encodes an enzyme in the histidine catabolic pathway. We show that AMDHD1 is exclusively expressed in the proliferating adult epithelial stem cells during metamorphosis with little expression in other intestinal tissues. We further provide evidence that T3 activates AMDHD1 gene expression directly at the transcription level through T3 receptor binding to the AMDHD1 gene in the intestine. In addition, we have reported earlier that histidine ammonia-lyase gene, another gene in histidine catabolic pathway, is similarly regulated by T3 in the intestine. These results together suggest that histidine catabolism plays a critical role in the formation and/or proliferation of adult intestinal stem cells during metamorphosis.
Collapse
Affiliation(s)
- Morihiro Okada
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Thomas C Miller
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Liezhen Fu
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
32
|
Malaterre J, Pereira L, Putoczki T, Millen R, Paquet-Fifield S, Germann M, Liu J, Cheasley D, Sampurno S, Stacker SA, Achen MG, Ward RL, Waring P, Mantamadiotis T, Ernst M, Ramsay RG. Intestinal-specific activatable Myb initiates colon tumorigenesis in mice. Oncogene 2015; 35:2475-84. [PMID: 26300002 PMCID: PMC4867492 DOI: 10.1038/onc.2015.305] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 05/31/2015] [Accepted: 07/13/2015] [Indexed: 02/07/2023]
Abstract
Transcription factor Myb is overexpressed in most colorectal cancers (CRC). Patients with CRC expressing the highest Myb are more likely to relapse. We previously showed that mono-allelic loss of Myb in an Adenomatous polyposis coli (APC)-driven CRC mouse model (ApcMin/+) significantly improves survival. Here we directly investigated the association of Myb with poor prognosis and how Myb co-operates with tumor suppressor genes (TSGs) (Apc) and cell cycle regulator, p27. Here we generated the first intestinal-specific, inducible transgenic model; a MybER transgene encoding a tamoxifen-inducible fusion protein between Myb and the estrogen receptor-α ligand-binding domain driven by the intestinal-specific promoter, Gpa33. This was to mimic human CRC with constitutive Myb activity in a highly tractable mouse model. We confirmed that the transgene was faithfully expressed and inducible in intestinal stem cells (ISCs) before embarking on carcinogenesis studies. Activation of the MybER did not change colon homeostasis unless one p27 allele was lost. We then established that MybER activation during CRC initiation using a pro-carcinogen treatment, azoxymethane (AOM), augmented most measured aspects of ISC gene expression and function and accelerated tumorigenesis in mice. CRC-associated symptoms of patients including intestinal bleeding and anaemia were faithfully mimicked in AOM-treated MybER transgenic mice and implicated hypoxia and vessel leakage identifying an additional pathogenic role for Myb. Collectively, the results suggest that Myb expands the ISC pool within which CRC is initiated while co-operating with TSG loss. Myb further exacerbates CRC pathology partly explaining why high MYB is a predictor of worse patient outcome.
Collapse
Affiliation(s)
- J Malaterre
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - L Pereira
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - T Putoczki
- Walter and Elisa Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - R Millen
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - S Paquet-Fifield
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - M Germann
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - J Liu
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - D Cheasley
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Walter and Elisa Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - S Sampurno
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - S A Stacker
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - M G Achen
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - R L Ward
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - P Waring
- Prince of Wales Clinical School and Lowy Cancer Research Centre, UNSW Medicine, Sydney, New South Wales, Australia
| | - T Mantamadiotis
- Prince of Wales Clinical School and Lowy Cancer Research Centre, UNSW Medicine, Sydney, New South Wales, Australia
| | - M Ernst
- Walter and Elisa Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - R G Ramsay
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Prince of Wales Clinical School and Lowy Cancer Research Centre, UNSW Medicine, Sydney, New South Wales, Australia
| |
Collapse
|
33
|
Tetteh PW, Farin HF, Clevers H. Plasticity within stem cell hierarchies in mammalian epithelia. Trends Cell Biol 2015; 25:100-8. [DOI: 10.1016/j.tcb.2014.09.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/08/2014] [Accepted: 09/12/2014] [Indexed: 12/20/2022]
|
34
|
Sun G, Fu L, Wen L, Shi YB. Activation of Sox3 gene by thyroid hormone in the developing adult intestinal stem cell during Xenopus metamorphosis. Endocrinology 2014; 155:5024-32. [PMID: 25211587 PMCID: PMC4239430 DOI: 10.1210/en.2014-1316] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The maturation of the intestine into the adult form involves the formation of adult stem cells in a thyroid hormone (T3)-dependent process in vertebrates. In mammals, this takes place during postembryonic development, a period around birth when the T3 level peaks. Due to the difficulty of manipulating late-stage, uterus-enclosed embryos, very little is known about the development of the adult intestinal stem cells. Interestingly, the remodeling of the intestine during the T3-dependent amphibian metamorphosis mimics the maturation of mammalian intestine. Our earlier microarray studies in Xenopus laevis revealed that the transcription factor SRY (sex-determining region Y)-box 3 (Sox3), well known for its involvement in neural development, was upregulated in the intestinal epithelium during metamorphosis. Here, we show that Sox3 is highly and specifically expressed in the developing adult intestinal progenitor/stem cells. We further show that its induction by T3 is independent of new protein synthesis, suggesting that Sox3 is directly activated by liganded T3 receptor. Thus, T3 activates Sox3 as one of the earliest changes in the epithelium, and Sox3 in turn may facilitate the dedifferentiation of the larval epithelial cells into adult stem cells.
Collapse
Affiliation(s)
- Guihong Sun
- School of Basic Medical Sciences (G.S.), Wuhan University, Wuhan 430072, People's Republic of China; and Section on Molecular Morphogenesis (L.F., L.W., Y.-B.S.), Program in Cellular Regulation and Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | | | | | | |
Collapse
|
35
|
Gracz AD, Magness ST. Defining hierarchies of stemness in the intestine: evidence from biomarkers and regulatory pathways. Am J Physiol Gastrointest Liver Physiol 2014; 307:G260-73. [PMID: 24924746 PMCID: PMC4121637 DOI: 10.1152/ajpgi.00066.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
For decades, the rapid proliferation and well-defined cellular lineages of the small intestinal epithelium have driven an interest in the biology of the intestinal stem cells (ISCs) and progenitors that produce the functional cells of the epithelium. Recent and significant advances in ISC biomarker discovery have established the small intestinal epithelium as a powerful model system for studying general paradigms in somatic stem cell biology and facilitated elegant genetic and functional studies of stemness in the intestine. However, this newfound wealth of ISC biomarkers raises important questions of marker specificity. Furthermore, the ISC field must now begin to reconcile biomarker status with functional stemness, a challenge that is made more complex by emerging evidence that cellular hierarchies in the intestinal epithelium are more plastic than previously imagined, with some progenitor populations capable of dedifferentiating and functioning as ISCs following damage. In this review, we discuss the state of the ISC field in terms of biomarkers, tissue dynamics, and cellular hierarchies, and how these processes might be informed by earlier studies into signaling networks in the small intestine.
Collapse
Affiliation(s)
- A. D. Gracz
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; ,2Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - S. T. Magness
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; ,2Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and ,3Joint Department of Biomedical Engineering, University of North Carolina/North Carolina State University, Chapel Hill, North Carolina
| |
Collapse
|
36
|
IL-1Ra selectively protects intestinal crypt epithelial cells, but not tumor cells, from chemotoxicity via p53-mediated upregulation of p21WAF1 and p27KIP1. Pharmacol Res 2014; 82:21-33. [DOI: 10.1016/j.phrs.2014.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 12/27/2022]
|
37
|
Yang Z, Tang LH, Klimstra DS. Gastroenteropancreatic neuroendocrine neoplasms: historical context and current issues. Semin Diagn Pathol 2013; 30:186-96. [PMID: 24144288 DOI: 10.1053/j.semdp.2013.06.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The digestive organs contain a large number of neuroendocrine cells as part of the diffuse neuroendocrine system. Neuroendocrine tumors can occur in every digestive organ. It has long been recognized that this is a diverse group of tumors with very different clinical outcomes; however, well-recognized prognostic parameters had been elusive until recently. Over the years, there have been several different classification schemes, each with different strengths and weaknesses. In an effort to standardize the classification and grading criteria for gastroenteropancreatic neuroendocrine tumors, the current World Health Organization classification includes a histologic grade based on proliferative rate (mitotic rate and Ki67 index) and a TNM stage that varies from organ to organ. The prognostic value of both the grade and stage has been validated in multiple studies. However, several issues remain, including the lack of standardized methods to assess proliferative rate, potential discrepancies between the mitotic count and the Ki67 index; intratumoral heterogeneity in proliferative rate; and the need for refinement in proliferative cut-points to define the grades. More studies are needed to further improve the classification of neuroendocrine tumors, thus guiding optimal treatment for these tumors.
Collapse
Affiliation(s)
- Zhaohai Yang
- Department of Pathology, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | | | | |
Collapse
|
38
|
Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat Rev Mol Cell Biol 2013; 15:19-33. [PMID: 24326621 DOI: 10.1038/nrm3721] [Citation(s) in RCA: 889] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Small populations of adult stem cells are responsible for the remarkable ability of the epithelial lining of the intestine to be efficiently renewed and repaired throughout life. The recent discovery of specific markers for these stem cells, together with the development of new technologies to track endogenous stem cell activity in vivo and to exploit their ability to generate new epithelia ex vivo, has greatly improved our understanding of stem cell-driven homeostasis, regeneration and cancer in the intestine. These exciting new insights into the biology of intestinal stem cells have the potential to accelerate the development of stem cell-based therapies and ameliorate cancer treatments.
Collapse
|
39
|
Abstract
The intestine has become a prime model system to study stem cell biology. Intestinal stem cells can be identified based on the expression of a unique marker gene, namely Lgr5. A transgenic mouse model expressing green fluorescent protein in intestinal stem cells has allowed their visualization, isolation, molecular characterization and use in generating organoids: small mini-guts that contain all cell types of the intestine. Detailing the behavior of intestinal stem cells has also led to new insights concerning the mechanism of self-renewal versus differentiation. Genes and pathways directing daughter cells of stem cells towards the differentiated lineages of the intestine are getting better defined. Of all differentiated cells, Paneth cells play a distinguished role: they emerged from pure bystanders to the guardians of the stem cell. Taken together, a detailed molecular picture emerges that describes the mechanisms of intestinal homeostatic self-renewal and outlines new therapeutic avenues.
Collapse
Affiliation(s)
- Daniel E Stange
- Department of General, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, University Dresden, Dresden, Germany
| |
Collapse
|
40
|
Luu N, Wen L, Fu L, Fujimoto K, Shi YB, Sun G. Differential regulation of two histidine ammonia-lyase genes during Xenopus development implicates distinct functions during thyroid hormone-induced formation of adult stem cells. Cell Biosci 2013; 3:43. [PMID: 24499573 PMCID: PMC3874607 DOI: 10.1186/2045-3701-3-43] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/30/2013] [Indexed: 11/16/2022] Open
Abstract
Background Organ-specific, adult stem cells are essential for organ-homeostasis and tissue repair and regeneration. The formation of such stem cells during vertebrate development remains to be investigated. Frog metamorphosis offers an excellent opportunity to study the formation of adult stem cells as this process involves essentially the transformations of all larval tissues/organs into the adult form. Of particular interest is the remodeling of the intestine. Early studies in Xenopus laevis have shown that this process involves complete degeneration of the larval epithelium and de novo formation of adult stem cells through dedifferentiation of some larval epithelial cells. A major advantage of this metamorphosis model is its total dependence on thyroid hormone (T3). In an effort to identify genes that are important for stem cell development, we have previously carried out tissue-specific microarray analysis of intestinal gene expression during Xenopus laevis metamorphosis. Results We report the detailed characterization of one of the genes thus identified, the histidine ammonia-lyase (HAL) gene, which encodes an enzyme known as histidase or histidinase. We show that there are two duplicated HAL genes, HAL1 and HAL2, in both Xenopus laevis and Xenopus tropicalis, a highly related but diploid species. Interestingly, only HAL2 is highly upregulated by T3 and appears to be specifically expressed in the adult intestinal progenitor/stem cells while HAL1 is not expressed in the intestine during metamorphosis. Furthermore, when analyzed in whole animals, HAL1 appears to be expressed only during embryogenesis but not metamorphosis while the opposite appears to be true for HAL2. Conclusions Our results suggest that the duplicated HAL genes have distinct functions with HAL2 likely involved in the formation and/or proliferation of the adult stem cells during metamorphosis.
Collapse
Affiliation(s)
- Nga Luu
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., 20892 Bethesda, Maryland, USA
| | - Luan Wen
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., 20892 Bethesda, Maryland, USA
| | - Liezhen Fu
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., 20892 Bethesda, Maryland, USA
| | - Kenta Fujimoto
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., 20892 Bethesda, Maryland, USA.,Present address: Division of Gene Structure and Function, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, 350-1241 Hidaka-shi, Saitama, Japan
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 18 Library Dr., 20892 Bethesda, Maryland, USA
| | - Guihong Sun
- School of Basic Medical Sciences, Wuhan University, 430072 Wuhan, P.R. China
| |
Collapse
|
41
|
Anderson EC, Hessman C, Levin TG, Monroe MM, Wong MH. The role of colorectal cancer stem cells in metastatic disease and therapeutic response. Cancers (Basel) 2013; 3:319-39. [PMID: 21318087 PMCID: PMC3036171 DOI: 10.3390/cancers3010319] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is the third-leading cause of cancer related mortality in the United States. The intricate molecular mechanisms involved in the regenerative process of the normal intestine and the identity of putative somatic intestinal stem cells have become clear. In parallel with this, experiment evidence has emerged supporting the century old hypothesis that solid tumor initiation, progression, chemoresistance and recurrence is the result of a small population of cancer cells with self-renewal and pluripotency capabilities. These "cancer stem cells" (CSCs) present a unique opportunity to better understand the biology of solid tumors in general, as well as targets for future therapeutics. In this review, we will summarize the current understanding of intestinal stem cell biology and translate it to colorectal CSCs to provide a basis for understanding chemoresistance, cancer recurrence and metastasis. A more complete understanding of the biology of colorectal CSCs will translate into the development of better chemotherapeutic and biological agents for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Eric C. Anderson
- Division of Hematology/Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; E-Mail:
| | - Crystal Hessman
- Department of Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; E-Mail:
| | - Trevor G. Levin
- Department of Cell and Developmental Biology, Knight Cancer Institute, Oregon Stem Cell Center, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; E-Mail:
| | - Marcus M. Monroe
- Department of Otolaryngology—Head and Neck Surgery, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; E-Mail:
| | - Melissa H. Wong
- Department of Dermatology, Department of Cell and Developmental Biology, Knight Cancer Institute, Oregon Stem Cell Center, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1 (503) 494-8749; Fax: +1 (503) 418-4266
| |
Collapse
|
42
|
Sun G, Heimeier RA, Fu L, Hasebe T, Das B, Ishizuya-Oka A, Shi YB. Expression profiling of intestinal tissues implicates tissue-specific genes and pathways essential for thyroid hormone-induced adult stem cell development. Endocrinology 2013; 154:4396-407. [PMID: 23970787 PMCID: PMC3800751 DOI: 10.1210/en.2013-1432] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The study of the epithelium during development in the vertebrate intestine touches upon many contemporary aspects of biology: to name a few, the formation of the adult stem cells (ASCs) essential for the life-long self-renewal and the balance of stem cell activity for renewal vs cancer development. Although extensive analyses have been carried out on the property and functions of the adult intestinal stem cells in mammals, little is known about their formation during development due to the difficulty of manipulating late-stage, uterus-enclosed embryos. The gastrointestinal tract of the amphibian Xenopus laevis is an excellent model system for the study of mammalian ASC formation, cell proliferation, and differentiation. During T3-dependent amphibian metamorphosis, the digestive tract is extensively remodeled from the larval to the adult form for the adaptation of the amphibian from its aquatic herbivorous lifestyle to that of a terrestrial carnivorous frog. This involves de novo formation of ASCs that requires T3 signaling in both the larval epithelium and nonepithelial tissues. To understand the underlying molecular mechanisms, we have characterized the gene expression profiles in the epithelium and nonepithelial tissues by using cDNA microarrays. Our results revealed that T3 induces distinct tissue-specific gene regulation programs associated with the remodeling of the intestine, particularly the formation of the ASCs, and further suggested the existence of potentially many novel stem cell-associated genes, at least in the intestine during development.
Collapse
Affiliation(s)
- Guihong Sun
- National Institutes of Health, National Institute of Child Health and Human Development, Laboratory of Gene Regulation and Development, Building 18T, Room 106, 18 Library Drive, MSC 5431, Bethesda, Maryland 20892; Rachel A. Heimeier, Section on Molecular Morphogenesis, Program in Cellular Regulation and Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; or Atsuko Ishizuya-Oka, Department of Biology, Nippon Medical School, Kawasaki, Kanagawa 211-0063, Japan. , , or
| | | | | | | | | | | | | |
Collapse
|
43
|
Fatehullah A, Appleton PL, Näthke IS. Cell and tissue polarity in the intestinal tract during tumourigenesis: cells still know the right way up, but tissue organization is lost. Philos Trans R Soc Lond B Biol Sci 2013; 368:20130014. [PMID: 24062584 PMCID: PMC3785964 DOI: 10.1098/rstb.2013.0014] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cell and tissue polarity are tightly coupled and are vital for normal tissue homeostasis. Changes in cellular and tissue organization are common to even early stages of disease, particularly cancer. The digestive tract is the site of the second most common cause of cancer deaths in the developed world. Tumours in this tissue arise in an epithelium that has a number of axes of cell and tissue polarity. Changes in cell and tissue polarity in response to genetic changes that are known to underpin disease progression provide clues about the link between molecular-, cellular- and tissue-based mechanisms that accompany cancer. Mutations in adenomatous polyposis coli (APC) are common to most colorectal cancers in humans and are sufficient to cause tumours in mouse intestine. Tissue organoids mimic many features of whole tissue and permit identifying changes at different times after inactivation of APC. Using gut organoids, we show that tissue polarity is lost very early during cancer progression, whereas cell polarity, at least apical-basal polarity, is maintained and changes only at later stages. These observations reflect the situation in tumours and validate tissue organoids as a useful system to investigate the relationship between cell polarity and tissue organization.
Collapse
Affiliation(s)
- Aliya Fatehullah
- Cell and Developmental Biology, University of Dundee, , Dundee DD1 5EH, UK
| | | | | |
Collapse
|
44
|
Yamamoto S, Nakase H, Matsuura M, Honzawa Y, Matsumura K, Uza N, Yamaguchi Y, Mizoguchi E, Chiba T. Heparan sulfate on intestinal epithelial cells plays a critical role in intestinal crypt homeostasis via Wnt/β-catenin signaling. Am J Physiol Gastrointest Liver Physiol 2013; 305:G241-9. [PMID: 23744737 PMCID: PMC3742857 DOI: 10.1152/ajpgi.00480.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Heparan sulfate (HS), a constituent of HS proteoglycans (HSPGs), is a linear polysaccharide present on the cell surface. HSPGs modulate functions of several growth factors and signaling molecules. We examined whether small intestinal epithelial HS plays some roles in crypt homeostasis using intestinal epithelium cell (IEC)-specific HS-deficient C57Bl/6 mice. Survival rate after total body irradiation was significantly reduced in HS-deficient mice due to profound intestinal injury. HS-deficient IECs exhibited Wnt/β-catenin pathway disruption, decreased levels of β-catenin nuclear localization, and reduced expression of Wnt target genes, including Lgr5 during crypt regeneration. Moreover, epithelial HS increased Wnt binding affinity of IECs, promoted phosphorylation of Wnt coreceptor LRP6, and enhanced Wnt/β-catenin signaling following ex vivo stimulation with Wnt3a, whereas activation of canonical Wnt signaling following direct inhibition of glycogen synthase kinase-3β by lithium chloride was similar between HS-deficient and wild-type mice. Thus HS influences the binding affinity of IECs to Wnt, thereby promoting activation of canonical Wnt signaling and facilitating regeneration of small intestinal crypts after epithelial injury.
Collapse
Affiliation(s)
- Shuji Yamamoto
- 1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto; ,2Japan Society for the Promotion of Science, Tokyo, Japan;
| | - Hiroshi Nakase
- 1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto;
| | - Minoru Matsuura
- 1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto;
| | - Yusuke Honzawa
- 1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto;
| | - Kayoko Matsumura
- 1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto;
| | - Norimitsu Uza
- 1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto;
| | - Yu Yamaguchi
- 3Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California;
| | - Emiko Mizoguchi
- 4Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tsutomu Chiba
- 1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto;
| |
Collapse
|
45
|
Gonzalez LM, Williamson I, Piedrahita JA, Blikslager AT, Magness ST. Cell lineage identification and stem cell culture in a porcine model for the study of intestinal epithelial regeneration. PLoS One 2013; 8:e66465. [PMID: 23840480 PMCID: PMC3696067 DOI: 10.1371/journal.pone.0066465] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 05/05/2013] [Indexed: 01/22/2023] Open
Abstract
Significant advances in intestinal stem cell biology have been made in murine models; however, anatomical and physiological differences between mice and humans limit mice as a translational model for stem cell based research. The pig has been an effective translational model, and represents a candidate species to study intestinal epithelial stem cell (IESC) driven regeneration. The lack of validated reagents and epithelial culture methods is an obstacle to investigating IESC driven regeneration in a pig model. In this study, antibodies against Epithelial Adhesion Molecule 1 (EpCAM) and Villin marked cells of epithelial origin. Antibodies against Proliferative Cell Nuclear Antigen (PCNA), Minichromosome Maintenance Complex 2 (MCM2), Bromodeoxyuridine (BrdU) and phosphorylated Histone H3 (pH3) distinguished proliferating cells at various stages of the cell cycle. SOX9, localized to the stem/progenitor cells zone, while HOPX was restricted to the +4/‘reserve’ stem cell zone. Immunostaining also identified major differentiated lineages. Goblet cells were identified by Mucin 2 (MUC2); enteroendocrine cells by Chromogranin A (CGA), Gastrin and Somatostatin; and absorptive enterocytes by carbonic anhydrase II (CAII) and sucrase isomaltase (SIM). Transmission electron microscopy demonstrated morphologic and sub-cellular characteristics of stem cell and differentiated intestinal epithelial cell types. Quantitative PCR gene expression analysis enabled identification of stem/progenitor cells, post mitotic cell lineages, and important growth and differentiation pathways. Additionally, a method for long-term culture of porcine crypts was developed. Biomarker characterization and development of IESC culture in the porcine model represents a foundation for translational studies of IESC-driven regeneration of the intestinal epithelium in physiology and disease.
Collapse
Affiliation(s)
- Liara M. Gonzalez
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Ian Williamson
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- UNC/NCSU Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Jorge A. Piedrahita
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Anthony T. Blikslager
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Scott T. Magness
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- UNC/NCSU Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
46
|
Paquette CE, Kent ML, Buchner C, Tanguay RL, Guillemin K, Mason TJ, Peterson TS. A retrospective study of the prevalence and classification of intestinal neoplasia in zebrafish (Danio rerio). Zebrafish 2013; 10:228-36. [PMID: 23544991 DOI: 10.1089/zeb.2012.0828] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
For over a decade, spontaneous intestinal neoplasia has been observed in zebrafish (Danio rerio) submitted to the ZIRC (Zebrafish International Resource Center) diagnostic service. In addition, zebrafish displayed preneoplastic intestinal changes including hyperplasia, dysplasia, and enteritis. A total of 195 zebrafish, representing 2% of the total fish submitted to the service, were diagnosed with these lesions. Neoplastic changes were classified either as adenocarcinoma or small cell carcinoma, with a few exceptions (carcinoma not otherwise specified, tubular adenoma, and tubulovillous adenoma). Tumor prevalence appeared similarly distributed between sexes and generally occurred in zebrafish greater than 1 year of age, although neoplastic changes were observed in fish 6 months of age. Eleven lines displayed these preneoplastic and neoplastic changes, including wild-types and mutants. Affected zebrafish originated from 18 facilities, but the majority of fish were from a single zebrafish research facility (hereafter referred to as the primary facility) that has submitted numerous samples to the ZIRC diagnostic service. Zebrafish from the primary facility submitted as normal sentinel fish demonstrate that these lesions are most often subclinical. Fish fed the diet from the primary facility and held at another location did not develop intestinal lesions, indicating that diet is not the etiologic agent.
Collapse
Affiliation(s)
- Colleen E Paquette
- Department of Microbiology, Oregon State University, Corvallis, OR 97331-3804, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Huynh D, Akçora D, Malaterre J, Chan CK, Dai XM, Bertoncello I, Stanley ER, Ramsay RG. CSF-1 receptor-dependent colon development, homeostasis and inflammatory stress response. PLoS One 2013; 8:e56951. [PMID: 23451116 PMCID: PMC3579891 DOI: 10.1371/journal.pone.0056951] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/16/2013] [Indexed: 01/09/2023] Open
Abstract
The colony stimulating factor-1 (CSF-1) receptor (CSF-1R) directly regulates the development of Paneth cells (PC) and influences proliferation and cell fate in the small intestine (SI). In the present study, we have examined the role of CSF-1 and the CSF-1R in the large intestine, which lacks PC, in the steady state and in response to acute inflammation induced by dextran sulfate sodium (DSS). As previously shown in mouse, immunohistochemical (IHC) analysis of CSF-1R expression showed that the receptor is baso-laterally expressed on epithelial cells of human colonic crypts, indicating that this expression pattern is shared between species. Colons from Csf1r null and Csf1(op/op) mice were isolated and sectioned for IHC identification of enterocytes, enteroendocrine cells, goblet cells and proliferating cells. Both Csf1r(-/-) and Csf1(op/op) mice were found to have colon defects in enterocytes and enteroendocrine cell fate, with excessive goblet cell staining and reduced cell proliferation. In addition, the gene expression profiles of the cell cycle genes, cyclinD1, c-myc, c-fos, and c-myb were suppressed in Csf1r(-/-) colonic crypt, compared with those of WT mice and the expression of the stem cell marker gene Lgr5 was markedly reduced. However, analysis of the proliferative responses of immortalized mouse colon epithelial cells (lines; Immorto-5 and YAMC) indicated that CSF-1R is not a major regulator of colonocyte proliferation and that its effects on proliferation are indirect. In an examination of the acute inflammatory response, Csf1r(+/-) male mice were protected from the adverse affects of DSS-induced colitis compared with WT mice, while Csf1r(+/-) female mice were significantly less protected. These data indicate that CSF-1R signaling plays an important role in colon homeostasis and stem cell gene expression but that the receptor exacerbates the response to inflammatory challenge in male mice.
Collapse
Affiliation(s)
- Duy Huynh
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Department of Genetics, Latrobe University, Victoria, Australia
| | - Dilara Akçora
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jordane Malaterre
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Chee Kai Chan
- Department of Genetics, Latrobe University, Victoria, Australia
| | - Xu-Ming Dai
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Ivan Bertoncello
- Department of Pharmacology the University of Melbourne, Parkville, Victoria, Australia
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Robert G. Ramsay
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
48
|
Sasikala M, Rao GV, Tandan M, Reddy DN. Gastro Intestinal Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
49
|
Natividad JMM, Verdu EF. Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications. Pharmacol Res 2012; 69:42-51. [PMID: 23089410 DOI: 10.1016/j.phrs.2012.10.007] [Citation(s) in RCA: 320] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 10/11/2012] [Accepted: 10/13/2012] [Indexed: 02/07/2023]
Abstract
Mammals and their intestinal microbiota peacefully coexist in a mutualistic relationship. Commensal bacteria play an active role in shaping and modulating physiological processes in the host, which include, but are not restricted to, the immune system and the intestinal barrier. Both play a crucial role in containing intestinal bacteria and other potentially noxious luminal antigens within the lumen and mucosal compartment. Although mutualism defines the relationship between the host and the intestinal microbiota, disruptions in this equilibrium may promote disease. Thus, alterations in gut microbiota (dysbiosis) have been linked to the recent increased expression of obesity, allergy, autoimmunity, functional and inflammatory disorders such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). In this article, we review the evidence supporting a role of gut microbiota in regulating intestinal barrier function. We discuss the hypothesis that microbial factors can modulate the barrier in ways that can prevent or promote gastrointestinal disease. A better understanding of the role of the intestinal microbiota in maintaining a functional intestinal barrier may help develop targeted strategies to prevent and treat disease.
Collapse
Affiliation(s)
- Jane M M Natividad
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | | |
Collapse
|
50
|
Grigoryan M, Kedees MH, Guz Y, Teitelman G. Phenotype of entero-endocrine L cells becomes restricted during development. Dev Dyn 2012; 241:1986-92. [PMID: 23027401 DOI: 10.1002/dvdy.23875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2012] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP) are hormones secreted by L and K cells, respectively, and by LK cells. To characterize L and K cells during development, we examined ileum from embryonic (e)- 12 to e-17. RESULTS GLP-1 cells were first seen at e-15 and their number increased at e-17. At e-17, most GLP-1 cells co-expressed GIP. The transcription factors Pax6 and Pdx-1 are required for GIP expression, while Pax6 activates the expression of GLP-1. At e-17, the mucosa has GIP+ Pax6+, GIP+ Pdx-1+, GLP-1+ Pax6+, and GLP-1+ Pdx-1+ cells. Unlike ileal L cells of postnatal and adult mice, a subset of ileal L cells of e-17 embryos co-expressed GLP-1 and glucagon (Glu). Glu-positive cells contain proprotein-convertase 2 (PC2) and PC3/1, the enzymes responsible for Glu and GLP-1 synthesis, respectively. CONCLUSIONS Our findings indicate that most GLP-1+ cells of ileum of e-17 embryos co-express GIP and, therefore, are LK cells. In addition, a subset of GLP-1+ cells of embryos but not of neonates co-express glucagon, indicating that the expression of Glu in GLP-1+ cells disappears after birth.
Collapse
Affiliation(s)
- Marine Grigoryan
- Department of Cell Biology, SUNY-Downstate Medical Center, Brooklyn, New York 11203, USA
| | | | | | | |
Collapse
|