1
|
Arcuri S, Pennarossa G, Pasquariello R, Prasadani M, Gandolfi F, Brevini TAL. Generation of Porcine and Rainbow Trout 3D Intestinal Models and Their Use to Investigate Astaxanthin Effects In Vitro. Int J Mol Sci 2024; 25:5966. [PMID: 38892151 PMCID: PMC11172962 DOI: 10.3390/ijms25115966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Astaxanthin (AST) is a natural compound derived from shellfish, microorganisms, and algae, with several healthy properties. For this reason, it is widely used in the diet of humans and animals, such as pigs, broilers, and fish, where its addition is related to its pigmenting properties. Moreover, AST's ability to reduce free radicals and protect cells from oxidative damage finds application during the weaning period, when piglets are exposed to several stressors. To better elucidate the mechanisms involved, here we generate ad hoc pig and rainbow trout in vitro platforms able to mimic the intestinal mucosa. The morphology is validated through histological and molecular analysis, while functional properties of the newly generated intestinal barriers, both in porcine and rainbow trout models, are demonstrated by measuring trans-epithelial electrical resistance and analyzing permeability with fluorescein isothiocyanate-dextran. Exposure to AST induced a significant upregulation of antioxidative stress markers and a reduction in the transcription of inflammation-related interleukins. Altogether, the present findings demonstrate AST's ability to interact with the molecular pathways controlling oxidative stress and inflammation both in the porcine and rainbow trout species and suggest AST's positive role in prevention and health.
Collapse
Affiliation(s)
- Sharon Arcuri
- Laboratory of Biomedical Embryology, Department of Veterinary Medicine and Animal Science and Center for Stem Cell Research, Università degli Studi di Milano, 26900 Lodi, Italy; (S.A.); (G.P.)
| | - Georgia Pennarossa
- Laboratory of Biomedical Embryology, Department of Veterinary Medicine and Animal Science and Center for Stem Cell Research, Università degli Studi di Milano, 26900 Lodi, Italy; (S.A.); (G.P.)
| | - Rolando Pasquariello
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy, Università degli Studi di Milano, 20133 Milan, Italy; (R.P.); (F.G.)
| | - Madhusha Prasadani
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia;
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy, Università degli Studi di Milano, 20133 Milan, Italy; (R.P.); (F.G.)
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology, Department of Veterinary Medicine and Animal Science and Center for Stem Cell Research, Università degli Studi di Milano, 26900 Lodi, Italy; (S.A.); (G.P.)
| |
Collapse
|
2
|
Mulero-Russe A, García AJ. Engineered Synthetic Matrices for Human Intestinal Organoid Culture and Therapeutic Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307678. [PMID: 37987171 PMCID: PMC10922691 DOI: 10.1002/adma.202307678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/02/2023] [Indexed: 11/22/2023]
Abstract
Human intestinal organoids (HIOs) derived from pluripotent stem cells or adult stem cell biopsies represent a powerful platform to study human development, drug testing, and disease modeling in vitro, and serve as a cell source for tissue regeneration and therapeutic advances in vivo. Synthetic hydrogels can be engineered to serve as analogs of the extracellular matrix to support HIO growth and differentiation. These hydrogels allow for tuning the mechanical and biochemical properties of the matrix, offering an advantage over biologically derived hydrogels such as Matrigel. Human intestinal organoids have been used for repopulating transplantable intestinal grafts and for in vivo delivery to an injured intestinal site. The use of synthetic hydrogels for in vitro culture and for in vivo delivery is expected to significantly increase the relevance of human intestinal organoids for drug screening, disease modeling, and therapeutic applications.
Collapse
Affiliation(s)
- Adriana Mulero-Russe
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
3
|
Pennarossa G, Arcuri S, Pasquariello R, Gandolfi F, Maranesi M, Brevini TAL. Cruciferous vegetable-derived indole-3-carbinol prevents coronavirus cell egression mechanisms in tracheal and intestinal 3D in vitro models. PHYTOCHEMISTRY 2023; 212:113713. [PMID: 37169138 PMCID: PMC10168192 DOI: 10.1016/j.phytochem.2023.113713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
The potential antiviral effects of indole-3-carbinol (I3C), a phytochemical found in Cruciferous vegetables, were investigated. Fibroblasts and epithelial cells were co-cultured on Alvetex® scaffolds, to obtain ad hoc 3D in vitro platforms able to mimic the trachea and intestinal mucosae, which represent the primary structures involved in the coronavirus pathogenesis. The two barriers generated in vitro were treated with various concentrations of I3C for different incubation periods. A protective effect of I3C on both intestinal and trachea models was demonstrated. A significant reduction in the transcription of the two main genes belonging to the Homologous to E6AP C-terminus (HECT)-E3 ligase family members, namely NEDD4 E3 Ubiquitin Protein Ligase (NEDD4) and WW Domain Containing E3 Ubiquitin Protein Ligase 1 (WWP1), which promote virus matrix protein ubiquitination and inhibit viral egression, were detected. These findings indicate I3C potential effect in preventing coronavirus cell egression processes that inhibit viral production. Although further studies are needed to clarify the molecular mechanisms whereby HECT family members control virus life cycle, this work paves the way to the possible therapeutic use of new natural compounds that may reduce the clinical severity of future pandemics.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Via Dell'Università 6, 26900, Lodi, Italy
| | - Sharon Arcuri
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Via Dell'Università 6, 26900, Lodi, Italy
| | - Rolando Pasquariello
- Department of Agricultural and Environmental Sciences - Production, Landscape, Agroenergy, Università Degli Studi di Milano, Via Celoria 10, 20133, Milan, Italy
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences - Production, Landscape, Agroenergy, Università Degli Studi di Milano, Via Celoria 10, 20133, Milan, Italy
| | - Margherita Maranesi
- Department of Veterinary Medicine, University of Perugia, Via S. Costanzo 4, 06126, Perugia, Italy.
| | - Tiziana A L Brevini
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Via Dell'Università 6, 26900, Lodi, Italy.
| |
Collapse
|
4
|
Temporal and spatial development of intestinal smooth muscle layers of human embryos and fetuses. J Dev Orig Health Dis 2023; 14:24-32. [PMID: 35924440 DOI: 10.1017/s2040174422000253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The sequential occurrence of three layers of smooth muscle layers (SML) in human embryos and fetus is not known. Here, we investigated the process of gut SML development in human embryos and fetuses and compared the morphology of SML in fetuses and neonates. The H&E, Masson trichrome staining, and Immunohistochemistry were conducted on 6-12 gestation week human embryos and fetuses and on normal neonatal intestine. We showed that no lumen was seen in 6-7th gestation week embryonic gut, neither gut wall nor SML was developed in this period. In 8-9th gestation week embryonic and fetal gut, primitive inner circular SML (IC-SML) was identified in a narrow and discontinuous gut lumen with some vacuoles. In 10th gestation week fetal gut, the outer longitudinal SML (OL-SML) in gut wall was clearly identifiable, both the inner and outer SML expressed α-SMA. In 11-12th gestation week fetal gut, in addition to the IC-SML and OL-SML, the muscularis mucosae started to develop as revealed by α-SMA immune-reactivity beneath the developing mucosal epithelial layer. Comparing with the gut of fetuses of 11-12th week of gestation, the muscularis mucosae, IC-SML, and OL-SML of neonatal intestine displayed different morphology, including branching into glands of lamina propria in mucosa and increased thickness. In conclusions, in the human developing gut between week-8 to week-12 of gestation, the IC-SML develops and forms at week-8, followed by the formation of OL-SML at week-10, and the muscularis mucosae develops and forms last at week-12.
Collapse
|
5
|
Ghannam SF, Rutland CS, Allegrucci C, Mongan NP, Rakha E. Defining invasion in breast cancer: the role of basement membrane. J Clin Pathol 2023; 76:11-18. [PMID: 36253088 DOI: 10.1136/jcp-2022-208584] [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: 09/08/2022] [Accepted: 10/01/2022] [Indexed: 12/27/2022]
Abstract
Basement membrane (BM) is an amorphous, sheet-like structure separating the epithelium from the stroma. BM is characterised by a complex structure comprising collagenous and non-collagenous proteoglycans and glycoproteins. In the breast, the thickness, density and composition of the BM around the ductal lobular system vary during differing development stages. In pathological conditions, the BM provides a physical barrier that separates proliferating intraductal epithelial cells from the surrounding stroma, and its absence or breach in malignant lesions is a hallmark of invasion and metastases. Currently, diagnostic services often use special stains and immunohistochemistry (IHC) to identify the BM in order to distinguish in situ from invasive lesions. However, distinguishing BM on stained sections, and differentiating the native BM from the reactive capsule or BM-like material surrounding some invasive malignant breast tumours is challenging. Although diagnostic use of the BM is being replaced by myoepithelial cell IHC markers, BM is considered by many to be a useful marker to distinguish in situ from invasive lesions in ambiguous cases. In this review, the structure, function and biological and clinical significance of the BM are discussed in relation to the various breast lesions with emphasis on how to distinguish the native BM from alternative pathological tissue mimicking its histology.
Collapse
Affiliation(s)
- Suzan F Ghannam
- Division of cancer and stem cells, school of Medicine, University of Nottingham, Nottingham, UK
- Histology and Cell Biology, Suez Canal University Faculty of Medicine, Ismailia, Egypt
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Catrin Sian Rutland
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, UK
| | - Cinzia Allegrucci
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, UK
| | - Nigel P Mongan
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, UK
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA
| | - Emad Rakha
- Division of cancer and stem cells, school of Medicine, University of Nottingham, Nottingham, UK
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Histopathology,school of Medicine, University of Nottingham School of Medicine, Nottingham, UK
| |
Collapse
|
6
|
Choo J, Glisovic N, Matic Vignjevic D. Gut homeostasis at a glance. J Cell Sci 2022; 135:281168. [DOI: 10.1242/jcs.260248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
ABSTRACT
The intestine, a rapidly self-renewing organ, is part of the gastrointestinal system. Its major roles are to absorb food-derived nutrients and water, process waste and act as a barrier against potentially harmful substances. Here, we will give a brief overview of the primary functions of the intestine, its structure and the luminal gradients along its length. We will discuss the dynamics of the intestinal epithelium, its turnover, and the maintenance of homeostasis. Finally, we will focus on the characteristics and functions of intestinal mesenchymal and immune cells. In this Cell Science at a Glance article and the accompanying poster, we aim to present the most recent information about gut cell biology and physiology, providing a resource for further exploration.
Collapse
Affiliation(s)
- Jieun Choo
- Institut Curie, PSL Research University, CNRS UMR 144 , F-75005 Paris , France
| | - Neda Glisovic
- Institut Curie, PSL Research University, CNRS UMR 144 , F-75005 Paris , France
| | | |
Collapse
|
7
|
Hautefort I, Poletti M, Papp D, Korcsmaros T. Everything You Always Wanted to Know About Organoid-Based Models (and Never Dared to Ask). Cell Mol Gastroenterol Hepatol 2022; 14:311-331. [PMID: 35643188 PMCID: PMC9233279 DOI: 10.1016/j.jcmgh.2022.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/12/2022]
Abstract
Homeostatic functions of a living tissue, such as the gastrointestinal tract, rely on highly sophisticated and finely tuned cell-to-cell interactions. These crosstalks evolve and continuously are refined as the tissue develops and give rise to specialized cells performing general and tissue-specific functions. To study these systems, stem cell-based in vitro models, often called organoids, and non-stem cell-based primary cell aggregates (called spheroids) appeared just over a decade ago. These models still are evolving and gaining complexity, making them the state-of-the-art models for studying cellular crosstalk in the gastrointestinal tract, and to investigate digestive pathologies, such as inflammatory bowel disease, colorectal cancer, and liver diseases. However, the use of organoid- or spheroid-based models to recapitulate in vitro the highly complex structure of in vivo tissue remains challenging, and mainly restricted to expert developmental cell biologists. Here, we condense the founding knowledge and key literature information that scientists adopting the organoid technology for the first time need to consider when using these models for novel biological questions. We also include information that current organoid/spheroid users could use to add to increase the complexity to their existing models. We highlight the current and prospective evolution of these models through bridging stem cell biology with biomaterial and scaffold engineering research areas. Linking these complementary fields will increase the in vitro mimicry of in vivo tissue, and potentially lead to more successful translational biomedical applications. Deepening our understanding of the nature and dynamic fine-tuning of intercellular crosstalks will enable identifying novel signaling targets for new or repurposed therapeutics used in many multifactorial diseases.
Collapse
Affiliation(s)
- Isabelle Hautefort
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom
| | - Martina Poletti
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom; Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom
| | - Diana Papp
- Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom
| | - Tamas Korcsmaros
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom; Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom; Imperial College London, Department of Metabolism, Digestion and Reproduction, London, United Kingdom.
| |
Collapse
|
8
|
Darling NJ, Mobbs CL, González-Hau AL, Freer M, Przyborski S. Bioengineering Novel in vitro Co-culture Models That Represent the Human Intestinal Mucosa With Improved Caco-2 Structure and Barrier Function. Front Bioeng Biotechnol 2020; 8:992. [PMID: 32984279 PMCID: PMC7487342 DOI: 10.3389/fbioe.2020.00992] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/29/2020] [Indexed: 12/17/2022] Open
Abstract
The Caco-2 monolayer is the most widely used in vitro model of the human intestinal mucosa to study absorption. However, models lack communication from other cells present in the native intestine, such as signals from fibroblasts in the lamina propria. In this study, we have investigated the effects of fibroblasts upon the Caco-2 epithelium through two mechanisms: indirect signaling from fibroblasts and direct contact with fibroblasts. Culture of Caco-2 cells with paracrine signals from fibroblasts, through the use of conditioned media, did not induce a significant change in epithelial cell morphology or function. To examine the effects of direct contact between the epithelium and fibroblasts, we developed novel, humanized three-dimensional (3D) co-culture models whereby Caco-2 cells are grown on the surface of a subepithelial-like tissue construct containing intestinal or dermal fibroblasts. In our models, we observed endogenous extracellular matrix production from the fibroblasts that provides support to the above epithelium. The Caco-2 epithelium displayed morphological changes in 3D co-culture including enhanced polarization and the formation of a basement membrane-like attachment to the underlying stromal compartment. An important structural alteration was the significantly straightened lateral membrane that closely mimics the structure of the in vivo intestinal mucosa. This enhanced lateral membrane phenotype, in correlation with an reduction in TEER to levels more similar to the human intestine, is thought to be responsible for the increased paracellular permeability observed in 3D co-cultures. Our results demonstrate that direct contact between epithelial and mesenchymal cells results in an enhanced epithelial barrier. The in vitro models described herein have the potential to be used for studying intestinal epithelial-fibroblast interactions and could provide more accurate tools for drug permeability studies.
Collapse
Affiliation(s)
- Nicole J Darling
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Claire L Mobbs
- Department of Biosciences, Durham University, Durham, United Kingdom.,Reprocell Europe Ltd, Sedgefield, United Kingdom
| | | | - Matthew Freer
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, United Kingdom.,Reprocell Europe Ltd, Sedgefield, United Kingdom
| |
Collapse
|
9
|
Abstract
The adult gastrointestinal tract (GI) is a series of connected organs (esophagus, stomach, small intestine, colon) that develop via progressive regional specification of a continuous tubular embryonic organ anlage. This chapter focuses on organogenesis of the small intestine. The intestine arises by folding of a flat sheet of endodermal cells into a tube of highly proliferative pseudostratified cells. Dramatic elongation of this tube is driven by rapid epithelial proliferation. Then, epithelial-mesenchymal crosstalk and physical forces drive a stepwise cascade that results in convolution of the tubular surface into finger-like projections called villi. Concomitant with villus formation, a sharp epithelial transcriptional boundary is defined between stomach and intestine. Finally, flask-like depressions called crypts are established to house the intestinal stem cells needed throughout life for epithelial renewal. New insights into these events are being provided by in vitro organoid systems, which hold promise for future regenerative engineering of the small intestine.
Collapse
Affiliation(s)
- Sha Wang
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| | - Katherine D Walton
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States.
| | - Deborah L Gumucio
- University of Michigan, Cell and Developmental Biology Department, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Cancer-associated fibroblasts induce metalloprotease-independent cancer cell invasion of the basement membrane. Nat Commun 2017; 8:924. [PMID: 29030636 PMCID: PMC5640679 DOI: 10.1038/s41467-017-00985-8] [Citation(s) in RCA: 252] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/08/2017] [Indexed: 12/24/2022] Open
Abstract
At the stage of carcinoma in situ, the basement membrane (BM) segregates tumor cells from the stroma. This barrier must be breached to allow dissemination of the tumor cells to adjacent tissues. Cancer cells can perforate the BM using proteolysis; however, whether stromal cells play a role in this process remains unknown. Here we show that an abundant stromal cell population, cancer-associated fibroblasts (CAFs), promote cancer cell invasion through the BM. CAFs facilitate the breaching of the BM in a matrix metalloproteinase-independent manner. Instead, CAFs pull, stretch, and soften the BM leading to the formation of gaps through which cancer cells can migrate. By exerting contractile forces, CAFs alter the organization and the physical properties of the BM, making it permissive for cancer cell invasion. Blocking the ability of stromal cells to exert mechanical forces on the BM could therefore represent a new therapeutic strategy against aggressive tumors.Stromal cells play various roles in tumor establishment and metastasis. Here the authors, using an ex-vivo model, show that cancer-associated fibroblasts facilitate colon cancer cells invasion in a matrix metalloproteinase-independent manner, likely by pulling and stretching the basement membrane to form gaps.
Collapse
|
11
|
YAP/TAZ and Hedgehog Coordinate Growth and Patterning in Gastrointestinal Mesenchyme. Dev Cell 2017; 43:35-47.e4. [PMID: 28943241 DOI: 10.1016/j.devcel.2017.08.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 05/04/2017] [Accepted: 08/25/2017] [Indexed: 12/11/2022]
Abstract
YAP/TAZ are the major mediators of mammalian Hippo signaling; however, their precise function in the gastrointestinal tract remains poorly understood. Here we dissect the distinct roles of YAP/TAZ in endodermal epithelium and mesenchyme and find that, although dispensable for gastrointestinal epithelial development and homeostasis, YAP/TAZ function as the critical molecular switch to coordinate growth and patterning in gut mesenchyme. Our genetic analyses reveal that Lats1/2 kinases suppress expansion of the primitive mesenchymal progenitors, where YAP activation also prevents induction of the smooth muscle lineage through transcriptional repression of Myocardin. During later development, zone-restricted downregulation of YAP/TAZ provides the positional cue and allows smooth muscle cell differentiation induced by Hedgehog signaling. Taken together, our studies identify the mesenchymal requirement of YAP/TAZ in the gastrointestinal tract and highlight the functional interplays between Hippo and Hedgehog signaling underlying temporal and spatial control of tissue growth and specification in developing gut.
Collapse
|
12
|
Interaction of prostate carcinoma-associated fibroblasts with human epithelial cell lines in vivo. Differentiation 2017; 96:40-48. [PMID: 28779656 DOI: 10.1016/j.diff.2017.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
Stromal-epithelial interactions play a crucial and poorly understood role in carcinogenesis and tumor progression. Mesenchymal-epithelial interactions have a long history of research in relation to the development of organs. Models designed to study development are often also applicable to studies of benign and malignant disease. Tumor stroma is a complex mixture of cells that includes a fibroblastic component often referred to as cancer-associated fibroblasts (CAF), desmoplasia or "reactive" stroma. Here we discuss the history of, and approaches to, understanding these interactions with particular reference to prostate cancer and to in vivo modeling using human cells and tissues. A series of studies have revealed a complex mixture of signaling molecules acting both within the stromal tissue and between the stromal and epithelial tissues. We are starting to understand the interactions of some of these pathways, however the work is still ongoing. This area of research provide a basis for new medical approaches aimed at stabilizing early stage cancers rendering them chronic rather than acute problems. Such work is especially relevant to slow growing tumors found in older patients, a class that would include many prostate cancers.
Collapse
|
13
|
Nakamura T, Watanabe M. Intestinal stem cell transplantation. J Gastroenterol 2017; 52:151-157. [PMID: 27888356 DOI: 10.1007/s00535-016-1288-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 02/04/2023]
Abstract
Organoid technologies to expand intestinal epithelial cells are gaining increasing attention as a useful tool to investigate many aspects of intestinal epithelial biology and pathology. One important application of organoid systems would be to use intestinal epithelial cells expanded in culture for following transplantation experiments. In this article, we present a brief overview of the studies that have succeeded in generating new epithelial tissues in the surface of native intestines in mice by organoid transplantation. We also discuss possible applications of this experimental approach in basic research on the intestinal epithelium as well as in regenerative medicine for various types of intestinal diseases in humans.
Collapse
Affiliation(s)
- Tetsuya Nakamura
- Department of Advanced Therapeutics for GI Diseases, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan.
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| |
Collapse
|
14
|
Burrin DG. Trophic Factors and Regulation of Gastrointestinal Tract and Liver Development. FETAL AND NEONATAL PHYSIOLOGY 2017:855-860.e1. [DOI: 10.1016/b978-0-323-35214-7.00086-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
15
|
Tsai YH, Nattiv R, Dedhia PH, Nagy MS, Chin AM, Thomson M, Klein OD, Spence JR. In vitro patterning of pluripotent stem cell-derived intestine recapitulates in vivo human development. Development 2016; 144:1045-1055. [PMID: 27927684 PMCID: PMC5358103 DOI: 10.1242/dev.138453] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/23/2016] [Indexed: 12/16/2022]
Abstract
The intestine plays a central role in digestion, nutrient absorption and metabolism, with individual regions of the intestine having distinct functional roles. Many examples of region-specific gene expression in the adult intestine are known, but how intestinal regional identity is established during development is a largely unresolved issue. Here, we have identified several genes that are expressed in a region-specific manner in the developing human intestine. Using human embryonic stem cell-derived intestinal organoids, we demonstrate that the duration of exposure to active FGF and WNT signaling controls regional identity. Short-term exposure to FGF4 and CHIR99021 (a GSK3β inhibitor that stabilizes β-catenin) resulted in organoids with gene expression patterns similar to developing human duodenum, whereas longer exposure resulted in organoids similar to ileum. When region-specific organoids were transplanted into immunocompromised mice, duodenum-like organoids and ileum-like organoids retained their regional identity, demonstrating that regional identity of organoids is stable after initial patterning occurs. This work provides insights into the mechanisms that control regional specification of the developing human intestine and provides new tools for basic and translational research. Summary: Human embryonic stem cell-derived intestinal organoids can be patterned into duodenum-like or ileum-like tissue, recapitulating in vivo human development.
Collapse
Affiliation(s)
- Yu-Hwai Tsai
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Roy Nattiv
- Institute for Human Genetics and Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA.,Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Priya H Dedhia
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Melinda S Nagy
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alana M Chin
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthew Thomson
- Center for Systems and Synthetic Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ophir D Klein
- Institute for Human Genetics and Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA .,Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason R Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA .,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Goi M, Childs SJ. Patterning mechanisms of the sub-intestinal venous plexus in zebrafish. Dev Biol 2015; 409:114-128. [PMID: 26477558 DOI: 10.1016/j.ydbio.2015.10.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 10/05/2015] [Accepted: 10/12/2015] [Indexed: 12/31/2022]
Abstract
Despite considerable interest in angiogenesis, organ-specific angiogenesis remains less well characterized. The vessels that absorb nutrients from the yolk and later provide blood supply to the developing digestive system are primarily venous in origin. In zebrafish, these are the vessels of the Sub-intestinal venous plexus (SIVP) and they represent a new candidate model to gain an insight into the mechanisms of venous angiogenesis. Unlike other vessel beds in zebrafish, the SIVP is not stereotypically patterned and lacks obvious sources of patterning information. However, by examining the area of vessel coverage, number of compartments, proliferation and migration speed we have identified common developmental steps in SIVP formation. We applied our analysis of SIVP development to obd mutants that have a mutation in the guidance receptor PlexinD1. obd mutants show dysregulation of nearly all parameters of SIVP formation. We show that the SIVP responds to a unique combination of pathways that control both arterial and venous growth in other systems. Blocking Shh, Notch and Pdgf signaling has no effect on SIVP growth. However Vegf promotes sprouting of the predominantly venous plexus and Bmp promotes outgrowth of the structure. We propose that the SIVP is a unique model to understand novel mechanisms utilized in organ-specific angiogenesis.
Collapse
Affiliation(s)
- Michela Goi
- Department of Biochemistry and Molecular Biology and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - Sarah J Childs
- Department of Biochemistry and Molecular Biology and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
17
|
Sirakov M, Boussouar A, Kress E, Frau C, Lone IN, Nadjar J, Angelov D, Plateroti M. The thyroid hormone nuclear receptor TRα1 controls the Notch signaling pathway and cell fate in murine intestine. Development 2015; 142:2764-74. [PMID: 26286942 DOI: 10.1242/dev.121962] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Thyroid hormones control various aspects of gut development and homeostasis. The best-known example is in gastrointestinal tract remodeling during amphibian metamorphosis. It is well documented that these hormones act via the TR nuclear receptors, which are hormone-modulated transcription factors. Several studies have shown that thyroid hormones regulate the expression of several genes in the Notch signaling pathway, indicating a possible means by which they participate in the control of gut physiology. However, the mechanisms and biological significance of this control have remained unexplored. Using multiple in vivo and in vitro approaches, we show that thyroid hormones positively regulate Notch activity through the TRα1 receptor. From a molecular point of view, TRα1 indirectly controls Notch1, Dll1, Dll4 and Hes1 expression but acts as a direct transcriptional regulator of the Jag1 gene by binding to a responsive element in the Jag1 promoter. Our findings show that the TRα1 nuclear receptor plays a key role in intestinal crypt progenitor/stem cell biology by controlling the Notch pathway and hence the balance between cell proliferation and cell differentiation.
Collapse
Affiliation(s)
- Maria Sirakov
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Amina Boussouar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Elsa Kress
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Carla Frau
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Imtiaz Nisar Lone
- Laboratoire de Biologie Moléculaire de la cellule, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon 69007, France
| | - Julien Nadjar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| | - Dimitar Angelov
- Laboratoire de Biologie Moléculaire de la cellule, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, Lyon 69007, France
| | - Michelina Plateroti
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, 16 Rue Raphael Dubois, Villeurbanne 69622, France
| |
Collapse
|
18
|
Glentis A, Gurchenkov V, Matic Vignjevic D. Assembly, heterogeneity, and breaching of the basement membranes. Cell Adh Migr 2015; 8:236-45. [PMID: 24727304 DOI: 10.4161/cam.28733] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Basement membranes are thin sheets of self-assembled extracellular matrices that are essential for embryonic development and for the homeostasis of adult tissues. They play a role in structuring, protecting, polarizing, and compartmentalizing cells, as well as in supplying them with growth factors. All basement membranes are built from laminin and collagen IV networks stabilized by nidogen/perlecan bridges. The precise composition of basement membranes, however, varies between different tissues. Even though basement membranes represent physical barriers that delimit different tissues, they are breached in many physiological or pathological processes, including development, the immune response, and tumor invasion. Here, we provide a brief overview of the molecular composition of basement membranes and the process of their assembly. We will then illustrate the heterogeneity of basement membranes using two examples, the epithelial basement membrane in the gut and the vascular basement membrane. Finally, we examine the different strategies cells use to breach the basement membrane.
Collapse
|
19
|
Jerman UD, Kreft ME, Veranič P. Epithelial-Mesenchymal Interactions in Urinary Bladder and Small Intestine and How to Apply Them in Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:521-30. [PMID: 26066408 DOI: 10.1089/ten.teb.2014.0678] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Reciprocal interactions between the epithelium and mesenchyme are essential for the establishment of proper tissue morphology during organogenesis and tissue regeneration as well as for the maintenance of cell differentiation. With this review, we highlight the importance of epithelial-mesenchymal cross talk in healthy tissue and further discuss its significance in engineering functional tissues in vitro. We focus on the urinary bladder and small intestine, organs that are often compromised by disease and are as such in need of research that would advance effective treatment or tissue replacement. To date, the understanding of epithelial-mesenchymal reciprocal interactions has enabled the development of in vitro biomimetic tissue equivalents that have provided many possibilities in treating defective, damaged, or even cancerous tissues. Although research of the past several years has advanced the field of bladder and small intestine tissue engineering, one must be aware of its current limitations in successfully and above all safely introducing tissue-engineered constructs into clinical practice. Special attention is in particular needed when treating cancerous tissues, as initially successful tumor excision and tissue reconstruction may later on result in cancer recurrence due to oncogenic signals originating from an altered stroma. Recent rather poor outcomes in pioneering clinical trials of bladder reconstructions should serve as a reminder that recreating a functional organ to replace a dysfunctional one is an objective far more difficult to reach than initially foreseen. When considering effective tissue engineering approaches for diseased tissues in humans, it is imperative to introduce animal models with dysfunctional or, even more importantly, cancerous organs, which would greatly contribute to predicting possible complications and, hence, reducing risks when translating to the clinic.
Collapse
Affiliation(s)
- Urška Dragin Jerman
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana , Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana , Ljubljana, Slovenia
| | - Peter Veranič
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana , Ljubljana, Slovenia
| |
Collapse
|
20
|
Postnatal development, maturation and aging in the mouse cochlea and their effects on hair cell regeneration. Hear Res 2012; 297:68-83. [PMID: 23164734 DOI: 10.1016/j.heares.2012.11.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/22/2012] [Accepted: 11/07/2012] [Indexed: 12/23/2022]
Abstract
The organ of Corti in the mammalian inner ear is comprised of mechanosensory hair cells (HCs) and nonsensory supporting cells (SCs), both of which are believed to be terminally post-mitotic beyond late embryonic ages. Consequently, regeneration of HCs and SCs does not occur naturally in the adult mammalian cochlea, though recent evidence suggests that these cells may not be completely or irreversibly quiescent at earlier postnatal ages. Furthermore, regenerative processes can be induced by genetic and pharmacological manipulations, but, more and more reports suggest that regenerative potential declines as the organ of Corti continues to age. In numerous mammalian systems, such effects of aging on regenerative potential are well established. However, in the cochlea, the problem of regeneration has not been traditionally viewed as one of aging. This is an important consideration as current models are unable to elicit widespread regeneration or full recovery of function at adult ages yet regenerative therapies will need to be developed specifically for adult populations. Still, the advent of gene targeting and other genetic manipulations has established mice as critically important models for the study of cochlear development and HC regeneration and suggests that auditory HC regeneration in adult mammals may indeed be possible. Thus, this review will focus on the pursuit of regeneration in the postnatal and adult mouse cochlea and highlight processes that occur during postnatal development, maturation, and aging that could contribute to an age-related decline in regenerative potential. Second, we will draw upon the wealth of knowledge pertaining to age related senescence in tissues outside of the ear to synthesize new insights and potentially guide future research aimed at promoting HC regeneration in the adult cochlea.
Collapse
|
21
|
Gomceli I, Demiriz B, Tez M. Gastric carcinogenesis. World J Gastroenterol 2012; 18:5164-70. [PMID: 23066309 PMCID: PMC3468847 DOI: 10.3748/wjg.v18.i37.5164] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 04/30/2012] [Accepted: 05/05/2012] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the second most common cancer worldwide and the second most common cause of cancer-related deaths. Despite complete resection of gastric cancer and lymph node dissection, as well as improvements in chemotherapy and radiotherapy, there are still 700 000 gastric cancer-related deaths per year worldwide and more than 80% of patients with advanced gastric cancer die of the disease or recurrent disease within 1 year after diagnosis. None of the treatment modalities we have been applying today can influence the overall survival rates: at present, the overall 5-year relative survival rate for gastric cancer is about 28%. Cellular metaplasia due to chronic inflammation, injury and repair are the most documented processes for neoplasia. It appears that chronic inflammation stimulates tumor development and plays a critical role in initiating, sustaining and advancing tumor growth. It is also evident that not all inflammation is tumorigenic. Additional mutations can be acquired, and this leads to the cancer cell gaining a further growth advantage and acquiring a more malignant phenotype. Intestinalization of gastric units, which is called “intestinal metaplasia”; phenotypic antralization of fundic units, which is called “spasmolytic polypeptide-expressing metaplasia”; and the development directly from the stem/progenitor cell zone are three pathways that have been described for gastric carcinogenesis. Also, an important factor for the development of gastrointestinal cancers is peritumoral stroma. However, the initiating cellular event in gastric metaplasia is still controversial. Understanding gastric carcinogenesis and its precursor lesions has been under intense investigation, and our paper attempts to highlight recent progress in this field of cancer research.
Collapse
|
22
|
Zeng L, Childs SJ. The smooth muscle microRNA miR-145 regulates gut epithelial development via a paracrine mechanism. Dev Biol 2012; 367:178-86. [PMID: 22609551 DOI: 10.1016/j.ydbio.2012.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 04/27/2012] [Accepted: 05/03/2012] [Indexed: 01/25/2023]
Abstract
MicroRNAs are potent modulators of cellular differentiation. miR-145 is expressed in, and promotes the differentiation of vascular and visceral smooth muscle cells (SMCs). Interestingly, we have observed that miR-145 also promotes differentiation of the gut epithelium in the developing zebrafish, a cell type where it is not expressed. Here we identify that a paracrine pathway involving the morphogens Sonic hedgehog (Shh) in epithelium and bone morphogenic protein 4 (Bmp4) in SMCs is modulated by miR-145. We show that expression of miR-145 in visceral SMCs normally represses the expression of the morphogen bmp4, as loss of miR-145 leads to upregulation of bmp4 in SMCs. We show that bmp4 in turn controls expression of Shh in the visceral epithelium. Conversely, in miR-145 morphants where bmp4 expression is increased, expression of sonic hedgehog a (shha) is strongly increased in gut epithelium. We show that expression of bmp4 is modulated by the miR-145 direct target gata6 but not a second potential direct target, klf5a. Thus although miR-145 is a tissue-restricted microRNA, it plays an essential role in promoting the patterning of both gut layers during gut development via a paracrine mechanism.
Collapse
Affiliation(s)
- Lei Zeng
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada T2N 4N1
| | | |
Collapse
|
23
|
Beaulieu JF, Ménard D. Isolation, characterization, and culture of normal human intestinal crypt and villus cells. Methods Mol Biol 2012; 806:157-173. [PMID: 22057451 DOI: 10.1007/978-1-61779-367-7_11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The intestinal epithelium is a highly dynamic tissue undergoing constant and rapid renewal. It consists of a functional villus compartment responsible for terminal digestion and nutrient absorption and a progenitor cell compartment located in the crypts that produce new cells. The mechanisms regulating cell proliferation in the crypt, their migration, and differentiation are still incompletely understood. Until recently, normal human intestinal cell models allowing the study of these mechanisms have been lacking. In our laboratory, using fetal human intestines obtained at mid-gestation, we have generated the first normal human intestinal epithelial crypt-like (HIEC) cell line and villus-like primary cultures of differentiated enterocytes (PCDE). In this chapter, we provide a detailed description of the methodologies used to generate and characterize these normal intestinal crypt and villus cell models.
Collapse
Affiliation(s)
- Jean-François Beaulieu
- CIHR Team on Digestive Epithelium, Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | | |
Collapse
|
24
|
Kim TH, Kim BM, Mao J, Rowan S, Shivdasani RA. Endodermal Hedgehog signals modulate Notch pathway activity in the developing digestive tract mesenchyme. Development 2011; 138:3225-33. [PMID: 21750033 DOI: 10.1242/dev.066233] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The digestive tract epithelium and its adjoining mesenchyme undergo coordinated patterning and growth during development. The signals they exchange in the process are not fully characterized but include ligands of the Hedgehog (Hh) family, which originate in the epithelium and are necessary for mesenchymal cells to expand in number and drive elongation of the developing gut tube. The Notch signaling pathway has known requirements in fetal and adult intestinal epithelial progenitors. We detected Notch pathway activity in the embryonic gut mesenchyme and used conditional knockout mice to study its function. Selective disruption of the Notch effector gene RBP-Jκ (Rbpj) in the mesenchyme caused progressive loss of subepithelial fibroblasts and abbreviated gut length, revealing an unexpected requirement in this compartment. Surprisingly, constitutive Notch activity also induced rapid mesenchymal cell loss and impaired organogenesis, probably resulting from increased cell death and suggesting the need for a delicate balance in Notch signaling. Because digestive tract anomalies in mouse embryos with excess Notch activity phenocopy the absence of Hh signaling, we postulated that endodermal Hh restrains mesenchymal Notch pathway activity. Indeed, Hh-deficient embryos showed Notch overactivity in their defective gut mesenchyme and exposure to recombinant sonic hedgehog could override Notch-induced death of cultured fetal gut mesenchymal cells. These results reveal unexpected interactions between prominent signals in gastrointestinal development and provide a coherent explanation for Hh requirements in mesenchymal cell survival and organ growth.
Collapse
Affiliation(s)
- Tae-Hee Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
25
|
Wulkersdorfer B, Kao KK, Agopian VG, Dunn JC, Wu BM, Stelzner M. Growth Factors Adsorbed on Polyglycolic Acid Mesh Augment Growth of Bioengineered Intestinal Neomucosa. J Surg Res 2011; 169:169-78. [DOI: 10.1016/j.jss.2009.11.719] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 10/07/2009] [Accepted: 11/18/2009] [Indexed: 01/18/2023]
|
26
|
Yu T, Lan SY, Wu B, Pan QH, Shi L, Huang KH, Lin Y, Chen QK. Musashi1 and hairy and enhancer of split 1 high expression cells derived from embryonic stem cells enhance the repair of small-intestinal injury in the mouse. Dig Dis Sci 2011; 56:1354-1368. [PMID: 21221806 DOI: 10.1007/s10620-010-1441-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Accepted: 09/19/2010] [Indexed: 12/13/2022]
Abstract
BACKGROUND Embryonic stem cells have great plasticity. In this study, we repaired impaired small intestine by transplanting putative intestinal epithelial stem cells (Musashi1 and hairy and enhancer of split 1 high expression cells) derived from embryonic stem cells. METHODS The differentiation of definitive endoderm in embryoid bodies, derived from male ES-E14TG2a cells by the hanging-drop method, was monitored to define a time point for maximal induction of putative intestinal epithelial stem cells by epidermal growth factor. Furthermore, to evaluate the regenerative potential of intestinal epithelium, these putative stem cells were engrafted into NOD/SCID mice and female mice with enteritis. Donor cells were located by SRY DNA in situ hybridization. RESULTS The results revealed that definitive endodermal markers were highly expressed in 5-day embryoid bodies. These embryoid body cells were induced into putative intestinal epithelial stem cells on the 5th day of epidermal growth factor administration. Grafts from these cells consisted of adenoid structures and nonspecific structural cells with strong expression of small-intestinal epithelial cell markers. In situ hybridization revealed that the donor cells could specifically locate in damaged intestinal epithelium, contribute to epithelial structures, and enhance regeneration. CONCLUSIONS In conclusion, the Musashi1 and hairy and enhancer of split 1 high expression cells, derived from mouse embryonic stem cells, locate predominantly in impaired small-intestinal epithelium after transplantation and contribute to epithelial regeneration.
Collapse
Affiliation(s)
- Tao Yu
- Department of Gastroenterology, The Second Affiliated Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, 510120, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Kim BM, Woo J, Kanellopoulou C, Shivdasani RA. Regulation of mouse stomach development and Barx1 expression by specific microRNAs. Development 2011; 138:1081-6. [PMID: 21307095 DOI: 10.1242/dev.056317] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although microRNAs (miRNAs) are postulated to fine-tune many developmental processes, their relationships with specific targets and tissues remain largely undefined. The mesenchymal transcription factor Barx1 controls spleen and stomach morphogenesis and is required to specify stomach-specific epithelium in adjacent endoderm. Barx1 expression is precisely regulated in space and time, with a sharp drop in stomach levels after epithelial specification. We tested the hypothesis that specific miRNAs mediate this marked decline in Barx1 levels. Depletion of the miRNA-processing enzyme Dicer in cultured stomach mesenchyme and conditional Dicer gene deletion in mice significantly increased Barx1 levels, disrupted stomach and intestine development and caused spleen agenesis. Computational and experimental studies identified miR-7a and miR-203 as candidate miRNAs that regulate Barx1 and are expressed in inverse proportion to it in the fetal mouse stomach. Through specific interactions with cognate sequences in the Barx1 3' untranslated region, miR-7a and miR-203 repress Barx1 expression in stomach mesenchymal cells and its function in inducing gastric epithelium. These results indicate that miRNAs are required for proper digestive tract organogenesis and that miR-7a and miR-203 control expression of the stomach homeotic regulator Barx1.
Collapse
Affiliation(s)
- Byeong-Moo Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
28
|
Mao J, Kim BM, Rajurkar M, Shivdasani RA, McMahon AP. Hedgehog signaling controls mesenchymal growth in the developing mammalian digestive tract. Development 2010; 137:1721-9. [PMID: 20430747 DOI: 10.1242/dev.044586] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Homeostasis of the vertebrate digestive tract requires interactions between an endodermal epithelium and mesenchymal cells derived from the splanchnic mesoderm. Signaling between these two tissue layers is also crucial for patterning and growth of the developing gut. From early developmental stages, sonic hedgehog (Shh) and indian hedgehog (Ihh) are secreted by the endoderm of the mammalian gut, indicative of a developmental role. Further, misregulated hedgehog (Hh) signaling is implicated in both congenital defects and cancers arising from the gastrointestinal tract. In the mouse, only limited gastrointestinal anomalies arise following removal of either Shh or Ihh. However, given the considerable overlap in their endodermal expression domains, a functional redundancy between these signals might mask a more extensive role for Hh signaling in development of the mammalian gut. To address this possibility, we adopted a conditional approach to remove both Shh and Ihh functions from early mouse gut endoderm. Analysis of compound mutants indicates that continuous Hh signaling is dispensable for regional patterning of the gut tube, but is essential for growth of the underlying mesenchyme. Additional in vitro analysis, together with genetic gain-of-function studies, further demonstrate that Hh proteins act as paracrine mitogens to promote the expansion of adjacent mesenchymal progenitors, including those of the smooth muscle compartment. Together, these studies provide new insights into tissue interactions underlying mammalian gastrointestinal organogenesis and disease.
Collapse
Affiliation(s)
- Junhao Mao
- Department of Cancer Biology, University of Massachusetts Medical School, LRB 405, Worcester, MA 01605, USA.
| | | | | | | | | |
Collapse
|
29
|
Shaker A, Swietlicki EA, Wang L, Jiang S, Onal B, Bala S, DeSchryver K, Newberry R, Levin MS, Rubin DC. Epimorphin deletion protects mice from inflammation-induced colon carcinogenesis and alters stem cell niche myofibroblast secretion. J Clin Invest 2010; 120:2081-93. [PMID: 20458144 DOI: 10.1172/jci40676] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 03/03/2010] [Indexed: 01/07/2023] Open
Abstract
Epithelial-mesenchymal interactions regulate normal gut epithelial homeostasis and have a putative role in inflammatory bowel disease and colon cancer pathogenesis. Epimorphin is a mesenchymal and myofibroblast protein with antiproliferative, promorphogenic effects in intestinal epithelium. We previously showed that deletion of epimorphin partially protects mice from acute colitis, associated with an increase in crypt cell proliferation. Here we explored the potential therapeutic utility of modulating epimorphin expression by examining the effects of epimorphin deletion on chronic inflammation-associated colon carcinogenesis using the azoxymethane/dextran sodium sulfate (AOM/DSS) model. We found that mice in which epimorphin expression was absent had a marked reduction in incidence and extent of colonic dysplasia. Furthermore, epimorphin deletion in myofibroblasts altered the morphology and growth of cocultured epithelial cells. Loss of epimorphin affected secretion of soluble mesenchymal regulators of the stem cell niche such as Chordin. Importantly, IL-6 secretion from LPS-treated epimorphin-deficient myofibroblasts was completely inhibited, and stromal IL-6 expression was reduced in vivo. Taken together, these data show that epimorphin deletion inhibits chronic inflammation-associated colon carcinogenesis in mice, likely as a result of increased epithelial repair, decreased myofibroblast IL-6 secretion, and diminished IL-6-induced inflammation. Furthermore, we believe that modulation of epimorphin expression may have therapeutic benefits in appropriate clinical settings.
Collapse
Affiliation(s)
- Anisa Shaker
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gracz AD, Ramalingam S, Magness ST. Sox9 expression marks a subset of CD24-expressing small intestine epithelial stem cells that form organoids in vitro. Am J Physiol Gastrointest Liver Physiol 2010; 298:G590-600. [PMID: 20185687 PMCID: PMC2867430 DOI: 10.1152/ajpgi.00470.2009] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The inability to identify, isolate, and culture intestinal epithelial stem cells (IESCs) has been prohibitive to the study and therapeutic utilization of these cells. Using a Sox9(EGFP) mouse model, we demonstrate that Sox9(EGFP) fluorescence signatures can be used to differentiate between and enrich for progenitors (Sox9(EGFPsubLo)) and multipotent IESCs (Sox9(EGFPlo)). Sox9(EGFPlo) cells generate "organoids" in a recently defined culture system that mimics the native IESC niche. These organoids possess all four differentiated cell types of the small intestine epithelium, demonstrating the multipotent capacity of Sox9(EGFPlo) cells. Our results are consistent with the previously reported observation that single IESCs generate cryptlike units without a detectable mesenchymal cell component. A prospective search revealed that CD24 is expressed in the Sox9(EGFPlo) population and marks IESCs that form organoids in culture. CD24 represents the first cell surface marker that facilitates fluorescence-activated cell sorting enrichment of IESCs with widely available antibodies without requiring a specialized fluorescent reporter gene mouse model.
Collapse
Affiliation(s)
- Adam D. Gracz
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sendhilnathan Ramalingam
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Scott T. Magness
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
31
|
The role of the basement membrane as a modulator of intestinal epithelial-mesenchymal interactions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 96:175-206. [PMID: 21075345 DOI: 10.1016/b978-0-12-381280-3.00008-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal development is a process of continuous dynamic bidirectional crosstalk between epithelial and underlying mesenchymal cells. This crosstalk is mediated by well-dissected signaling pathways. Another crucial actor in the epithelio-mesenchymal interactions is the stromal microenvironment, which is composed of extracellular matrix molecules. Among them, the basement membrane (BM) molecules are secreted by the epithelium and mesenchyme in a complementary manner. These molecules signal back to the cells via the integrins or other specific receptors. In this review, we mainly focus on the BM molecules, particularly laminins. The major BM molecules are organized in a complex molecular network, which is highly variable among organs. Cell culture, coculture, and grafting models have been of great interest in understanding the importance of these molecules. Mouse gene ablation of laminin chains are interesting models, which often lead to embryonic death and are frequently accompanied by compensatory processes. Overall, the BM molecules have a crucial role in the careful maintenance of intestinal homeostasis.
Collapse
|
32
|
Abstract
The rapid specification and differentiation of the embryonic zebrafish gut is essential to provide contractility for the digestion of food. The role of microRNAs in modulating gut epithelial or smooth muscle differentiation is currently not known. Here we show that the microRNA miR-145 is strongly expressed in zebrafish gut smooth muscle and regulates its development. Modulation of miR-145 levels results in gut smooth muscle and epithelium maturation defects. Loss of miR-145 results in defects of smooth muscle function as measured by decreased nitric oxide production but also leads to increased expression of the embryonic smooth muscle markers sm22alpha-b, nm-mhc-b, and smoothelin. Defects in gut epithelial maturation are also present as observed by immature morphology and a complete loss of alkaline phosphatase expression. Loss or gain of miR-145 function phenocopies defects observed with altered gata6 expression and accordingly, we show that miR-145 directly represses gata6, and that gata6 is a major miR-145 target in vitro and in vivo. miR-145 therefore plays a critical role in promoting the maturation of both layers of the gut during development through regulation of gata6.
Collapse
|
33
|
Kwek J, De Iongh R, Nicholas K, Familari M. Molecular insights into evolution of the vertebrate gut: focus on stomach and parietal cells in the marsupial,Macropus eugenii. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2009; 312:613-24. [DOI: 10.1002/jez.b.21227] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
34
|
Abstract
The epithelial cell lining of the gastrointestinal tract is the most rapidly proliferating tissue in the body. The constant state of renewal of differentiated epithelial cells is sustained by a continual supply of progeny from multipotent progenitors that originate from stem cells located within the intestinal crypts. In addition to supporting normal epithelial homeostasis, intestinal stem cells (ISC) are thought to play an important role in the rapid expansion of the gut during development, tissue regeneration following injury or surgical loss, and malignancy. Because of the lack of specific ISC markers required to isolate and characterize these cells, our current knowledge of the biology of ISC results largely from indirect measures of their behavior published during the past 40 years. The recent description of several potential ISC markers and the use of transgenic mice, both as a tool to lineage trace and to isolate specific cells expressing these markers, have provided a tremendous advancement to our current understanding of these cells. This brief review provides a general historical overview of our understanding of ISC and the tools available to study their behavior in the context of normal and pathological conditions, as well as potential future clinical applications that may result from this exciting area of research.
Collapse
|
35
|
Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, van Es JH, Abo A, Kujala P, Peters PJ, Clevers H. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009; 459:262-5. [PMID: 19329995 DOI: 10.1038/nature07935] [Citation(s) in RCA: 4862] [Impact Index Per Article: 303.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Accepted: 02/24/2009] [Indexed: 02/06/2023]
Abstract
The intestinal epithelium is the most rapidly self-renewing tissue in adult mammals. We have recently demonstrated the presence of about six cycling Lgr5(+) stem cells at the bottoms of small-intestinal crypts. Here we describe the establishment of long-term culture conditions under which single crypts undergo multiple crypt fission events, while simultanously generating villus-like epithelial domains in which all differentiated cell types are present. Single sorted Lgr5(+) stem cells can also initiate these cryptvillus organoids. Tracing experiments indicate that the Lgr5(+) stem-cell hierarchy is maintained in organoids. We conclude that intestinal cryptvillus units are self-organizing structures, which can be built from a single stem cell in the absence of a non-epithelial cellular niche.
Collapse
Affiliation(s)
- Toshiro Sato
- Hubrecht Institute and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Walton KD, Warner J, Hertzler PH, McClay DR. Hedgehog signaling patterns mesoderm in the sea urchin. Dev Biol 2009; 331:26-37. [PMID: 19393640 DOI: 10.1016/j.ydbio.2009.04.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 03/30/2009] [Accepted: 04/17/2009] [Indexed: 10/20/2022]
Abstract
The Hedgehog (Hh) signaling pathway is essential for patterning many structures in vertebrates including the nervous system, chordamesoderm, limb and endodermal organs. In the sea urchin, a basal deuterostome, Hh signaling is shown to participate in organizing the mesoderm. At gastrulation the Hh ligand is expressed by the endoderm downstream of the Brachyury and FoxA transcription factors in the endomesoderm gene regulatory network. The co-receptors Patched (Ptc) and Smoothened (Smo) are expressed by the neighboring skeletogenic and non-skeletogenic mesoderm. Perturbations of Hh, Ptc and Smo cause embryos to develop with skeletal defects and inappropriate non-skeletogenic mesoderm patterning, although initial specification of mesoderm occurs without detectable abnormalities. Perturbations of the pathway caused late defects in skeletogenesis and in the non-skeletogenic mesoderm, including altered numbers of pigment and blastocoelar cells, randomized left-right asymmetry of coelomic pouches, and disorganized circumesophageal muscle causing an inability to swallow. Together the data support the requirement of Hh signaling in patterning each of the mesoderm subtypes in the sea urchin embryo.
Collapse
Affiliation(s)
- Katherine D Walton
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
37
|
Kalabis J, Li G, Fukunaga-Kalabis M, Rustgi AK, Herlyn M. Endothelin-3 stimulates survival of goblet cells in organotypic cultures of fetal human colonic epithelium. Am J Physiol Gastrointest Liver Physiol 2008; 295:G1182-9. [PMID: 18832450 PMCID: PMC2604801 DOI: 10.1152/ajpgi.90294.2008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cells within the normal human colonic epithelium undergo a dynamic cycle of growth, differentiation, and death. The organotypic culture system of human fetal colonic epithelial cells seeded on top of collagen gels with embedded colonic fibroblasts allowed prolonged culture of the colonic epithelial cells (Kalabis J, Patterson MJ, Enders GM, Marian B, Iozzo RV, Rogler G, Gimotty PA, Herlyn M. FASEB J 17: 1115-1117, 2003). Herein, we have evaluated the role of endothelin-3 (ET3) and both cognate endothelin receptors (ETRA, ETRB) for human colonic epithelial cell growth and survival. ET3 was produced continuously by the fibroblasts as a result of adenovirus-mediated gene transfer. The presence and function of the endothelin receptors (ETRs) in epithelial cells was evaluated by [(3)H]thymidine incorporation using primary epithelial cells in monoculture and by immunohistochemistry on human fetal and adult paraffin-embedded tissues. In organotypic culture, ET3 increased the number of goblet cells but not of enteroendocrine cells. The increase in goblet cells was caused by prolonged cell survival and differentiation. The inhibition of both ETRA and ETRB significantly decreased the number of goblet cells and proliferation in epithelial cells, whereas the number of enteroendocrine cells remained unchanged. ET3 induced activation of IkappaB and MAPK in the epithelial cells, suggesting that these signaling pathways mediate its proproliferation and prosurvival activities. Our results demonstrate that ET3 is involved in regulating human colonic epithelial cell proliferation and survival, particularly for goblet cells, and may be an important component of colonic homeostasis.
Collapse
Affiliation(s)
- Jiri Kalabis
- Molecular and Cellular Oncogenesis Program, The Wistar Institute; and Division of Gastroenterology, Department of Medicine, Department of Genetics, and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gang Li
- Molecular and Cellular Oncogenesis Program, The Wistar Institute; and Division of Gastroenterology, Department of Medicine, Department of Genetics, and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mizuho Fukunaga-Kalabis
- Molecular and Cellular Oncogenesis Program, The Wistar Institute; and Division of Gastroenterology, Department of Medicine, Department of Genetics, and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anil K. Rustgi
- Molecular and Cellular Oncogenesis Program, The Wistar Institute; and Division of Gastroenterology, Department of Medicine, Department of Genetics, and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program, The Wistar Institute; and Division of Gastroenterology, Department of Medicine, Department of Genetics, and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
38
|
Barros R, Pereira B, Duluc I, Azevedo M, Mendes N, Camilo V, Jacobs RJ, Paulo P, Santos-Silva F, van Seuningen I, van den Brink GR, David L, Freund JN, Almeida R. Key elements of the BMP/SMAD pathway co-localize with CDX2 in intestinal metaplasia and regulate CDX2 expression in human gastric cell lines. J Pathol 2008; 215:411-20. [PMID: 18498120 DOI: 10.1002/path.2369] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori infection induces intestinal metaplasia of the stomach, a preneoplastic lesion associated with an increased risk for gastric cancer development. Intestinal metaplasia is induced by the intestine-specific transcription factor CDX2 but the mechanisms responsible for this ectopic expression have never been described. We hypothesized that the BMP/SMAD pathway has a role in CDX2 regulation, in this context, for the following reasons: (1) the BMP pathway is crucial for normal intestinal differentiation and (2) there is an influx of BMP2 and BMP4-producing cells to the stomach upon Helicobacter pylori infection. We evaluated the expression of key elements of the BMP pathway in human stomach specimens with IM. Growth factor treatments, with BMP2 and BMP4, were performed in cultured cells and a knock-down experiment of SMAD4 was done using RNAi. We showed overexpression in IM of BMP2/4, BMPR1A, and SMAD4 in 56% of IM foci, and pSMAD1/5/8 in 100% of IM foci as compared to adjacent mucosa. In vitro, treatment of AGS cells with BMP2 and BMP4 increased endogenous CDX2 expression as well as the intestinal differentiation markers MUC2 and LI-cadherin. On the other hand, SMAD4 knock-down led to decreased endogenous CDX2, MUC2, and LI-cadherin in AGS. Treatment of the SMAD4 knock-down cells had no influence on CDX2 expression as opposed to wild-type cells. A 9.3 kb CDX2 promoter could be transactivated by SMAD4 and SMAD1 in a cell-dependent manner. In conclusion, we identified for the first time that the BMP pathway is active in intestinal metaplasia and that BMP2 and BMP4 regulate CDX2 expression and promote intestinal differentiation through the canonical signal transducers.
Collapse
Affiliation(s)
- R Barros
- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Stringer EJ, Pritchard CA, Beck F. Cdx2 initiates histodifferentiation of the midgut endoderm. FEBS Lett 2008; 582:2555-60. [PMID: 18577384 DOI: 10.1016/j.febslet.2008.06.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 06/06/2008] [Accepted: 06/16/2008] [Indexed: 10/21/2022]
Abstract
Null mutation or haploinsufficiency of Cdx2 results in the development of heterotopic lesions with a gastric phenotype in the midgut endoderm. Conversely transgenic expression of Cdx2 in the stomach causes the endoderm to differentiate into intestinal-type mucosa. We demonstrate that the mesoderm adjacent to intestinal heterotopic areas expresses stomach specific Barx1 while the surrounding mesoderm is Barx1 negative. We conclude that the initiation of gut histodifferentiation lies in the endodermal expression of Cdx2 and that endodermal/mesodermal cross-talk involving Barx1 with appropriate feedback loops results in the development of the postnatal gut phenotype.
Collapse
Affiliation(s)
- Emma J Stringer
- Department of Biochemistry, University of Leicester, Leicester, UK
| | | | | |
Collapse
|
40
|
Juuti-Uusitalo K, Mäki M, Kainulainen H, Isola J, Kaukinen K. Gluten affects epithelial differentiation-associated genes in small intestinal mucosa of coeliac patients. Clin Exp Immunol 2007; 150:294-305. [PMID: 17888028 PMCID: PMC2219351 DOI: 10.1111/j.1365-2249.2007.03500.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2007] [Indexed: 01/25/2023] Open
Abstract
In coeliac disease gluten induces an immunological reaction in genetically susceptible patients, and influences on epithelial cell proliferation and differentiation in the small-bowel mucosa. Our aim was to find novel genes which operate similarly in epithelial proliferation and differentiation in an epithelial cell differentiation model and in coeliac disease patient small-bowel mucosal biopsy samples. The combination of cDNA microarray data originating from a three-dimensional T84 epithelial cell differentiation model and small-bowel mucosal biopsy samples from untreated and treated coeliac disease patients and healthy controls resulted in 30 genes whose mRNA expression was similarly affected. Nine of 30 were located directly or indirectly in the receptor tyrosine kinase pathway starting from the epithelial growth factor receptor. Removal of gluten from the diet resulted in a reversion in the expression of 29 of the 30 genes in the small-bowel mucosal biopsy samples. Further characterization by blotting and labelling revealed increased epidermal growth factor receptor and beta-catenin protein expression in the small-bowel mucosal epithelium in untreated coeliac disease patients compared to healthy controls and treated coeliac patients. We found 30 genes whose mRNA expression was affected similarly in the epithelial cell differentiation model and in the coeliac disease patient small-bowel mucosal biopsy samples. In particular, those genes involved in the epithelial growth factor-mediated signalling pathways may be involved in epithelial cell differentiation and coeliac disease pathogenesis. The epithelial cell differentiation model is a useful tool for studying gene expression changes in the crypt-villus axis.
Collapse
Affiliation(s)
- K Juuti-Uusitalo
- Paediatric Research Centre, Medical School and Department of Paediatrics, Tampere University Hospital and Medical School, University of Tampere, Tampere, Finland
| | | | | | | | | |
Collapse
|
41
|
Tou L, Liu Q, Shivdasani RA. Histone deacetylases 1 and 2 redundantly regulate cardiac morphogenesis, growth, and contractility. Genes Dev 2007; 24:3132-9. [PMID: 15060137 PMCID: PMC381684 DOI: 10.1128/mcb.24.8.3132-3139.2004] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Histone deacetylases (HDACs) tighten chromatin structure and repress gene expression through the removal of acetyl groups from histone tails. The class I HDACs, HDAC1 and HDAC2, are expressed ubiquitously, but their potential roles in tissue-specific gene expression and organogenesis have not been defined. To explore the functions of HDAC1 and HDAC2 in vivo, we generated mice with conditional null alleles of both genes. Whereas global deletion of HDAC1 results in death by embryonic day 9.5, mice lacking HDAC2 survive until the perinatal period, when they succumb to a spectrum of cardiac defects, including obliteration of the lumen of the right ventricle, excessive hyperplasia and apoptosis of cardiomyocytes, and bradycardia. Cardiac-specific deletion of either HDAC1 or HDAC2 does not evoke a phenotype, whereas cardiac-specific deletion of both genes results in neonatal lethality, accompanied by cardiac arrhythmias, dilated cardiomyopathy, and up-regulation of genes encoding skeletal muscle-specific contractile proteins and calcium channels. Our results reveal cell-autonomous and non-cell-autonomous functions for HDAC1 and HDAC2 in the control of myocardial growth, morphogenesis, and contractility, which reflect partially redundant roles of these enzymes in tissue-specific transcriptional repression.
Collapse
Affiliation(s)
- Liqiang Tou
- Department of Medical Oncology and Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | |
Collapse
|
42
|
Kim BM, Mao J, Taketo MM, Shivdasani RA. Phases of canonical Wnt signaling during the development of mouse intestinal epithelium. Gastroenterology 2007; 133:529-38. [PMID: 17681174 DOI: 10.1053/j.gastro.2007.04.072] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Accepted: 04/19/2007] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND AIMS Intestinal crypts constitute a niche in which epithelial progenitors respond to Wnt signals, replicate, and prepare to differentiate. Because mutations in Wnt pathway genes lead to intestinal cancer, the role of Wnt signaling in gut epithelial homeostasis is a subject of intense investigation. We studied how Wnt signaling is established during intestine development. METHODS We studied spatiotemporal features of Wnt signaling at formative stages in mouse embryos, when villous projections appear and crypt precursors occupy intervillus regions. We used TOP-GAL transgenic and Axin2(LacZ) mice, which report faithfully on canonical Wnt activity, relevant molecular markers, and embryos with aberrant beta-catenin activation. RESULTS Developing intestines first display evidence for Wnt signaling after appearance of villi. During villus morphogenesis, intervillus cells proliferate actively but lack signs of canonical Wnt signaling. Surprisingly, in late gestation and briefly thereafter, conspicuous Wnt activity is evident in differentiated, postmitotic villus epithelium. Neither Tcf4, a principal transcriptional effector of intestinal Wnt signals, nor candidate Wnt targets CD44 and cyclinD1 are expressed in late fetal villus cells that show high Wnt activity. Instead, those cells express the related factor Tcf3 and a different Wnt target, c-Myc. Premature and deregulated beta-catenin activation causes severe villus dysmorphogenesis in transgenic mice. CONCLUSIONS Relationships among Wnt signaling, epithelial proliferation, and tissue differentiation are reversed in the developing and adult gut. The canonical Wnt pathway has independent, albeit possibly overlapping, functions in early intestinal villi and adult crypts. These observations advance understanding of Wnt functions in intestinal development and disease.
Collapse
Affiliation(s)
- Byeong-Moo Kim
- Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
43
|
de Koning BAE, Lindenbergh-Kortleve DJ, Pieters R, Büller HA, Renes IB, Einerhand AWC. Alterations in epithelial and mesenchymal intestinal gene expression during doxorubicin-induced mucositis in mice. Dig Dis Sci 2007; 52:1814-25. [PMID: 17415656 PMCID: PMC1914222 DOI: 10.1007/s10620-006-9174-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 01/25/2006] [Indexed: 01/28/2023]
Abstract
In the current study we aimed to gain insight into epithelial-mesenchymal cross-talk and progenitor compartment modulation during doxorubicin (DOX)-induced mucositis in mice. Intestinal segments were collected on various days after DOX treatment. DOX-induced damage at day 1-2 was characterized by increased epithelial proliferation and apoptosis and a decrease in the expression of epithelial differentiation markers. Concurrently, T-cell factor-4 (TCF4) levels increased and the epithelial differentiation enhancing factor, bone morphogenic protein-4 (BMP4), decreased. During severe damage (day 3), BMP4 levels were significantly increased, which inversely correlated with epithelial proliferation. At the same time, the expression of the epithelial differentiation markers was increasing again. At day 7, BMP4 levels were down-regulated, while the levels of the epithelial differentiation markers and TCF4 were normalized again. These data suggest that in response to DOX-induced damage, BMP4 and TCF4 are modulated in such a way that homeostasis of the progenitor compartment is partly preserved.
Collapse
Affiliation(s)
- Barbara A E de Koning
- Department of Pediatrics, Divisions of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
44
|
Golaz JL, Vonlaufen N, Hemphill A, Burgener IA. Establishment and characterization of a primary canine duodenal epithelial cell culture. In Vitro Cell Dev Biol Anim 2007; 43:176-85. [PMID: 17577610 DOI: 10.1007/s11626-007-9034-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Accepted: 04/25/2007] [Indexed: 12/23/2022]
Abstract
Many mechanisms involved in the pathogenesis of chronic enteropathies or host-pathogen interactions in canine intestine have not been elucidated so far. Next to the clinical and in vivo research tools, an in vitro model of canine intestinal cell culture would be very helpful for studies at the cellular level. Therefore, the purpose of this study was to establish and characterize a primary canine duodenal epithelial cell culture. Neonatal duodenum was disrupted with trypsin-ethylenediaminetetraacetic acid (EDTA) and the mucosa scraped off and digested with collagenase and dispase. After centrifugation on a 2% sorbitol gradient, the cells were incubated at 37 degrees C in OptiMEM supplemented with Primocin, epidermal growth factor, insulin, hydrocortisone, and 10% fetal calf serum (FCS). After 24 h, the FCS concentration was reduced to 2.5%, and the temperature decreased to 33 degrees C. With this method, the cultures were growing to confluent monolayers within 5-6 d and remained viable for an average of 2 wk. Their epithelial nature was confirmed by electron microscopy and immunofluorescence staining using antibodies directed against specific cytokeratins, desmosomes, and tight junctions. The intestinal cells proliferated, as evidenced by immunolabeling with a Ki-67 antibody, and cryptal cell subpopulations could be identified. Furthermore, alkaline phosphatase and sucrase activity were detected.
Collapse
Affiliation(s)
- Julia L Golaz
- Department of Clinical Veterinary Medicine, Division of Small Animal Internal Medicine, Vetsuisse Faculty of the University of Bern, Länggassstrasse 122, 3001 Bern, Switzerland
| | | | | | | |
Collapse
|
45
|
Li X, Madison BB, Zacharias W, Kolterud A, States D, Gumucio DL. Deconvoluting the intestine: molecular evidence for a major role of the mesenchyme in the modulation of signaling cross talk. Physiol Genomics 2007; 29:290-301. [PMID: 17299133 DOI: 10.1152/physiolgenomics.00269.2006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Reciprocal cross talk between the endodermally derived epithelium and the underlying mesenchyme is required for regional patterning and proper differentiation of the developing mammalian intestine. Though both epithelium and mesenchyme participate in patterning, the mesenchyme is thought to play a prominent role in the determination of the epithelial phenotype during development and in adult life. However, the molecular basis for this instructional dominance is unclear. In fact, surprisingly little is known about the cellular origins of many of the critical signaling molecules and the gene transcriptional events that they impact. Here, we profile genes that are expressed in the separate mesenchymal and epithelial compartments of the perinatal mouse intestine. The data indicate that the vast majority of soluble inhibitors and modulators of signaling pathways such as Hedgehog, Bmp, Wnt, Fgf, and Igf are expressed predominantly or exclusively by the mesenchyme, accounting for its ability to dominate instructional cross talk. We also catalog the most highly enriched transcription factors in both compartments. The results bolster previous evidence suggesting a major role for Hnf4gamma and Hnf4alpha in the regulation of epithelial genes. Finally, we find that while epithelially enriched genes tend to be highly tissue restricted in their expression, mesenchymally enriched genes tend to be broadly expressed in multiple tissues. Thus, the unique tissue-specific signature that characterizes the intestinal epithelium is instructed and supported by a mesenchyme that itself expresses genes that are largely nontissue specific.
Collapse
Affiliation(s)
- Xing Li
- Bioinformatics Program, Departments of Cell and Developmental Biology and Human Genetics, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | |
Collapse
|
46
|
Furuya S, Furuya K. Subepithelial fibroblasts in intestinal villi: roles in intercellular communication. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 264:165-223. [PMID: 17964923 DOI: 10.1016/s0074-7696(07)64004-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ingestion of food and water induces chemical and mechanical signals that trigger peristaltic reflexes in the gut. Intestinal villi are motile, equipped with chemosensors and mechanosensors, and transduce signaling to sensory neurons, but the exact mechanisms have not yet been elucidated. Subepithelial fibroblasts located under the villous epithelium form contractile cellular networks via gap junctions. The networks ensheathe lamina propria and are in close contact with epithelium, neural and capillary networks, smooth muscles, and immune cells. Unique characteristics of subepithelial fibroblasts have been revealed by primary cultures isolated from rat duodenal villi. They include rapid reversal changes in cell shape by cAMP reagents and endothelins, cell shape-dependent mechanosensitivity that induces ATP release as a paracrine mediator, contractile ability, and expression of various receptors for vasoactive and neuroactive substances. Herein, we review these characteristics that play a key role in the villi. They serve as a barrier/sieve, flexible mechanical frame, mechanosensor, and signal transduction machinery in the intestinal villi, which are regulated locally and dynamically by rapid cell shape conversion.
Collapse
Affiliation(s)
- Sonoko Furuya
- Section of Brain Structure, Center for Brain Experiment, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | | |
Collapse
|
47
|
Jones RG, Li X, Gray PD, Kuang J, Clayton F, Samowitz WS, Madison BB, Gumucio DL, Kuwada SK. Conditional deletion of beta1 integrins in the intestinal epithelium causes a loss of Hedgehog expression, intestinal hyperplasia, and early postnatal lethality. ACTA ACUST UNITED AC 2006; 175:505-14. [PMID: 17088430 PMCID: PMC2064526 DOI: 10.1083/jcb.200602160] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Conditional deletion of β1 integrins in the intestinal epithelium, unlike in epidermal and mammary epithelia, of mice does not result in decreased cell adhesion and proliferation, but instead causes a profound increase in epithelial proliferation with dysplasia and polypoid structures. The increased epithelial proliferation inhibited epithelial differentiation that caused severe malnutrition and early postnatal lethality. The striking similarities between β1 integrin–deleted mice and neonatal mice with defective Hedgehog signaling led to the discovery that Hedgehog expression was markedly reduced in the former mice. β1 integrins were found to drive the expression of Hedgehogs in intestinal epithelial cells in an HNF-3β (Foxa2)–dependent fashion. The expression of Tcf-4, a transcription factor known to be required for intestinal epithelial stem cell proliferation, was increased and mislocalized in the intestinal epithelia of the β1 integrin–deleted mice and in newborn mice treated with the Hedgehog signaling inhibitor cyclopamine. This study shows that β1 integrins are key regulators of proliferation and homeostasis in the intestine and achieve this not through anchorage-dependent effects but by generating Hh expression and signaling.
Collapse
Affiliation(s)
- Robert G Jones
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Simon-Assmann P, Turck N, Sidhoum-Jenny M, Gradwohl G, Kedinger M. In vitro models of intestinal epithelial cell differentiation. Cell Biol Toxicol 2006; 23:241-56. [PMID: 17171431 DOI: 10.1007/s10565-006-0175-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Accepted: 10/23/2006] [Indexed: 11/28/2022]
Abstract
The intestinal epithelium is a particularly interesting tissue as (1) it is in a constant cell renewal from a stem cell pool located in the crypts which form, with the underlying fibroblasts, a stem cell niche and (2) the pluripotent stem cells give rise to four main cell types: enterocytes, mucus, endocrine, and Paneth cells. The mechanisms leading to the determination of phenotype commitment and cell-specific expressions are still poorly understood. Although transgenic mouse models are powerful tools for elucidating the molecular cascades implicated in these processes, cell culture approaches bring easy and elegant ways to study cellular behavior, cell interactions, and cell signaling pathways for example. In the present review, we will describe the major tissue culture technologies that allow differentiation of epithelial cells from undifferentiated embryonic or crypt cells. We will point to the necessity of the re-creation of a complex microenvironment that allows full differentiation process to occur. We will also summarize the characteristics and interesting properties of the cell lines established from human colorectal tumors.
Collapse
|
49
|
Gene expression in TGFbeta-induced epithelial cell differentiation in a three-dimensional intestinal epithelial cell differentiation model. BMC Genomics 2006; 7:279. [PMID: 17074098 PMCID: PMC1635984 DOI: 10.1186/1471-2164-7-279] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Accepted: 10/31/2006] [Indexed: 11/17/2022] Open
Abstract
Background The TGFβ1-induced signal transduction processes involved in growth and differentiation are only partly known. The three-dimensional epithelial differentiation model, in which T84 epithelial cells are induced to differentiate either with TGFβ1 or IMR-90 mesenchymal cell-secreted soluble factors, is previously shown to model epithelial cell differentiation seen in intestine. That model has not been used for large scale gene expression studies, such as microarray method. Therefore the gene expression changes were studied in undifferentiated and differentiated three-dimensional T84 cultures with cDNA microarray method in order to study the molecular changes and find new players in epithelial cell differentiation. Results The expression of 372 genes out of 5188 arrayed sequences was significantly altered, and 47 of them were altered by both mediators. The data were validated and the altered genes are presented in ontology classes. For the genes tested the expressions in protein level were in accordance with the mRNA results. We also found 194 genes with no known function to be potentially important in epithelial cell differentiation. The mRNA expression changes induced by TGFβ1 were bigger than changes induced by soluble factors secreted by IMR-90 mesenchymal cells. The gene expression data was depicted in already known signaling pathway routes. Conclusion Our results reveal potential new signaling pathways and several new genes affected by TGFβ in epithelial cell differentiation. The differentiation induced by TGFβ1 appears to be more potent than the differentiation induced by mesenchymal cells. This study indicates that our cell culture model is a suitable tool in studying regulatory mechanisms during epithelial cell differentiation in intestine. Furthermore the present results indicate that our model is a good tool for finding new players acting in the differentiation of epithelial cells.
Collapse
|
50
|
Al-Ashy R, Chakroun I, El-Sabban ME, Homaidan FR. The role of NF-κB in mediating the anti-inflammatory effects of IL-10 in intestinal epithelial cells. Cytokine 2006; 36:1-8. [PMID: 17161612 DOI: 10.1016/j.cyto.2006.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Revised: 07/19/2006] [Accepted: 10/03/2006] [Indexed: 12/31/2022]
Abstract
In inflammatory bowel disease, cells that infiltrate the mucosa regulate intestinal epithelial cell function partly through release of pro- and anti-inflammatory cytokines. The aim of this study is to evaluate the role of the anti-inflammatory cytokine, IL-10, on normal mouse intestinal epithelial cells (Mode-K) in the absence or presence of IL-1. Western blotting assays and immunocytochemistry were used to identify the presence of IL-1 and IL-10 receptors on Mode-K cells; and electrophoretic mobility shift assays were used to study the activation of NF-kappaB transcription factor. Stimulation of Mode-K cells with IL-1 or IL-10 did not modify IL-1 and IL-10 receptor expression levels. IL-1 induced the synthesis of the enzyme cyclooxygenase-2 (COX-2) through the activation and translocation of p65 subunit of NF-kappaB. Inhibition of translocated p65 binding to DNA, inhibited COX-2 production and induced apoptosis. IL-10 inhibited IL-1-induced effects on IKB-alpha and IKB-beta proteins through stabilizing these proteins; subsequently causing inhibition of NF-kappaB translocation to the nucleus and any subsequent induction of COX-2. These data support a role for IL-10 in the regulation of IEC function under inflammatory conditions and the involvement of COX-2 in inhibiting apoptosis in mouse intestinal epithelial cells.
Collapse
Affiliation(s)
- Randa Al-Ashy
- Department of Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | |
Collapse
|