1
|
Abud HE, Amarasinghe SL, Micati D, Jardé T. Stromal Niche Signals That Orchestrate Intestinal Regeneration. Cell Mol Gastroenterol Hepatol 2024; 17:679-685. [PMID: 38342301 PMCID: PMC10957453 DOI: 10.1016/j.jcmgh.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
Stromal cell populations have a central role in providing signals that support the maintenance, differentiation, and function of the intestinal epithelium. The behavior and fate of epithelial cells is directed by the spatial organization of stromal cells that either sustain stem and progenitor cell identity or drive differentiation. A combination of single-cell analyses, mouse models, and organoid coculture assays have provided insight into the diversity of signals delivered by stromal cells. Signaling gradients are established and fine-tuned by the expression of signaling agonists and antagonists along the crypt-villus axis. On epithelial injury, there are disruptions to the abundance and organization of stromal populations. There are also distinct changes in the signals originating from these cells that impact remodeling of the epithelium. How these signals coordinate to mediate epithelial repair or sustain tissue injury in inflammatory bowel diseases is beginning to emerge. Understanding of these processes may lead to opportunities to target stromal cell populations as a strategy to modify disease states.
Collapse
Affiliation(s)
- Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| | - Shanika L Amarasinghe
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Diana Micati
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
2
|
Ho GT, Theiss AL. Mitochondria and Inflammatory Bowel Diseases: Toward a Stratified Therapeutic Intervention. Annu Rev Physiol 2021; 84:435-459. [PMID: 34614372 DOI: 10.1146/annurev-physiol-060821-083306] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondria serve numerous critical cellular functions, rapidly responding to extracellular stimuli and cellular demands while dynamically communicating with other organelles. Mitochondrial function in the gastrointestinal epithelium plays a critical role in maintaining intestinal health. Emerging studies implicate the involvement of mitochondrial dysfunction in inflammatory bowel disease (IBD). This review presents mitochondrial metabolism, function, and quality control that converge in intestinal epithelial stemness, differentiation programs, barrier integrity, and innate immunity to influence intestinal inflammation. Intestinal and disease characteristics that set the stage for mitochondrial dysfunction being a key factor in IBD, and in turn, pathogenic mitochondrial mechanisms influencing and potentiating the development of IBD, are discussed. These findings establish the basis for potential mitochondrial-targeted interventions for IBD therapy. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Gwo-Tzer Ho
- Edinburgh IBD Science Unit, Centre for Inflammation Research, Queens Medical Research Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA;
| |
Collapse
|
3
|
Abud HE, Chan WH, Jardé T. Source and Impact of the EGF Family of Ligands on Intestinal Stem Cells. Front Cell Dev Biol 2021; 9:685665. [PMID: 34350179 PMCID: PMC8327171 DOI: 10.3389/fcell.2021.685665] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/27/2021] [Indexed: 12/27/2022] Open
Abstract
Epidermal Growth Factor (EGF) has long been known for its role in promoting proliferation of intestinal epithelial cells. EGF is produced by epithelial niche cells at the base of crypts in vivo and is routinely added to the culture medium to support the growth of intestinal organoids ex vivo. The recent identification of diverse stromal cell populations that reside underneath intestinal crypts has enabled the characterization of key growth factor cues supplied by these cells. The nature of these signals and how they are delivered to drive intestinal epithelial development, daily homeostasis and tissue regeneration following injury are being investigated. It is clear that aside from EGF, other ligands of the family, including Neuregulin 1 (NRG1), have distinct roles in supporting the function of intestinal stem cells through the ErbB pathway.
Collapse
Affiliation(s)
- Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Wing Hei Chan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| |
Collapse
|
4
|
Yu X, Li S, Xu Y, Zhang Y, Ma W, Liang C, Lu H, Ji Y, Liu C, Chen D, Li J. Androgen Maintains Intestinal Homeostasis by Inhibiting BMP Signaling via Intestinal Stromal Cells. Stem Cell Reports 2020; 15:912-925. [PMID: 32916121 PMCID: PMC7561494 DOI: 10.1016/j.stemcr.2020.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 01/13/2023] Open
Abstract
Research shows a higher incidence of colorectal cancer in men. However, the molecular mechanisms for this gender disparity remain unknown. We report the roles of androgen in proliferation and differentiation of intestinal stem cells via targeting of the androgen receptor (AR) on intestinal stromal cells by negatively regulating BMP signaling. Orchidectomy (ORX) or the AR antagonist promotes expansion of intestinal epithelium but suppresses intestinal stem cell (ISC) proliferation. Conversely, the AR agonist inhibits ISC differentiation but augments proliferation in ovariectomized mice. Mechanistically, activation of the AR increases expression of BMP antagonists but lowers expression of BMP4 and Wnt antagonists in primary stromal cells, which promotes intestinal organoid growth. Interestingly, the BMP pathway inhibitor LDN-193189 reverses the ORX-induced effects. Our results highlight that stromal cells constitute the intestinal stem cell niche and provide a possible explanation for higher incidence rates of colorectal cancer in men.
Collapse
Affiliation(s)
- Xin Yu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Shiguang Li
- School of Clinical Medicine, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Yiming Xu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan Wangfujing Dongcheng District, 100730 Beijing, China
| | - Yundi Zhang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Wenlong Ma
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Changchun Liang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Haodong Lu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Yuge Ji
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Chuanyong Liu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China
| | - Dawei Chen
- Laboratory of Medical Chemistry, Interdisciplinary Cluster for Applied Genoproteomics (GIGA), University of Liège, CHU, Sart-Tilman, 4000 Liège, Belgium,Corresponding author
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012 Jinan, China,Corresponding author
| |
Collapse
|
5
|
Jardé T, Chan WH, Rossello FJ, Kaur Kahlon T, Theocharous M, Kurian Arackal T, Flores T, Giraud M, Richards E, Chan E, Kerr G, Engel RM, Prasko M, Donoghue JF, Abe SI, Phesse TJ, Nefzger CM, McMurrick PJ, Powell DR, Daly RJ, Polo JM, Abud HE. Mesenchymal Niche-Derived Neuregulin-1 Drives Intestinal Stem Cell Proliferation and Regeneration of Damaged Epithelium. Cell Stem Cell 2020; 27:646-662.e7. [PMID: 32693086 DOI: 10.1016/j.stem.2020.06.021] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/13/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022]
Abstract
Epidermal growth factor (EGF) maintains intestinal stem cell (ISC) proliferation and is a key component of organoid growth media yet is dispensable for intestinal homeostasis, suggesting roles for multiple EGF family ligands in ISC function. Here, we identified neuregulin 1 (NRG1) as a key EGF family ligand that drives tissue repair following injury. NRG1, but not EGF, is upregulated upon damage and is expressed in mesenchymal stromal cells, macrophages, and Paneth cells. NRG1 deletion reduces proliferation in intestinal crypts and compromises regeneration capacity. NRG1 robustly stimulates proliferation in crypts and induces budding in organoids, in part through elevated and sustained activation of mitogen-activated protein kinase (MAPK) and AKT. Consistently, NRG1 treatment induces a proliferative gene signature and promotes organoid formation from progenitor cells and enhances regeneration following injury. These data suggest mesenchymal-derived NRG1 is a potent mediator of tissue regeneration and may inform the development of therapies for enhancing intestinal repair after injury.
Collapse
Affiliation(s)
- Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia.
| | - Wing Hei Chan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Fernando J Rossello
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; University of Melbourne Centre for Cancer Research, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Tanvir Kaur Kahlon
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Mandy Theocharous
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia; Cancer Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Teni Kurian Arackal
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Tracey Flores
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Mégane Giraud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Elizabeth Richards
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Eva Chan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Genevieve Kerr
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Rebekah M Engel
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, VIC 3144, Australia
| | - Mirsada Prasko
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Jacqueline F Donoghue
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Royal Women's Hospital, Melbourne University, Melbourne, VIC 3052, Australia
| | - Shin-Ichi Abe
- Center for Education, Kumamoto Health Science University, Kumamoto 861-5598, Japan
| | - Toby J Phesse
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK; Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Christian M Nefzger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Paul J McMurrick
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, VIC 3144, Australia
| | - David R Powell
- Monash Bioinformatics Platform, Monash University, Clayton, VIC 3800, Australia
| | - Roger J Daly
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia; Cancer Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia.
| |
Collapse
|
6
|
Kondo S, Mizuno S, Hashita T, Iwao T, Matsunaga T. Establishment of a novel culture method for maintaining intestinal stem cells derived from human induced pluripotent stem cells. Biol Open 2020; 9:bio049064. [PMID: 31919043 PMCID: PMC6955217 DOI: 10.1242/bio.049064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023] Open
Abstract
The small intestine plays an important role in the pharmacokinetics of orally administered drugs due to the presence of drug transporters and drug-metabolizing enzymes. However, few appropriate methods exist to investigate intestinal pharmacokinetics. Induced pluripotent stem (iPS) cells can form various types of cells and represent a potentially useful tool for drug discovery. We previously reported that differentiated enterocytes from human iPS cells are useful for pharmacokinetic studies; however, the process is time and resource intensive. Here, we established a new two-dimensional culture method for maintaining human iPS-cell-derived intestinal stem cells (ISCs) with differentiation potency and evaluated their ability to differentiate into enterocytes exhibiting appropriate pharmacokinetic function. The culture method used several factors to activate signalling pathways required for maintaining stemness, followed by differentiation into enterocytes. Functional evaluation was carried out to verify epithelial-marker expression and inducibility and activity of metabolic enzymes and transporters. Our results confirmed the establishment of an ISC culture method for maintaining stemness and verified that the differentiated enterocytes from the maintained ISCs demonstrated proper pharmacokinetic function. Thus, our findings describe a time- and cost-effective approach that can be used as a general evaluation tool for evaluating intestinal pharmacokinetics.
Collapse
Affiliation(s)
- Satoshi Kondo
- Department of Drug Safety Research, Nonclinical Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Shota Mizuno
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Tadahiro Hashita
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
7
|
Jeon MK, Kaemmerer E, Schneider U, Schiffer M, Klaus C, Hennings J, Clahsen T, Ackerstaff T, Niggemann M, Schippers A, Longerich T, Sellge G, Trautwein C, Wagner N, Liedtke C, Gassler N. Notch inhibition counteracts Paneth cell death in absence of caspase-8. Virchows Arch 2018; 473:71-83. [PMID: 29770852 DOI: 10.1007/s00428-018-2368-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 04/09/2018] [Accepted: 04/29/2018] [Indexed: 02/07/2023]
Abstract
Opposing activities of Notch and Wnt signaling regulate mucosal barrier homeostasis and differentiation of intestinal epithelial cells. Specifically, Wnt activity is essential for differentiation of secretory cells including Wnt3-producing Paneth cells, whereas Notch signaling strongly promotes generation of absorptive cells. Loss of caspase-8 in intestinal epithelium (casp8∆int) is associated with fulminant epithelial necroptosis, severe Paneth cell death, secondary intestinal inflammation, and an increase in Notch activity. Here, we found that pharmacological Notch inhibition with dibenzazepine (DBZ) is able to essentially rescue the loss of Paneth cells, deescalate the inflammatory phenotype, and reduce intestinal permeability in casp8∆int mice. The secretory cell metaplasia in DBZ-treated casp8∆int animals is proliferative, indicating for Notch activities partially insensitive to gamma-secretase inhibition in a casp8∆int background. Our data suggest that casp8 acts in the intestinal Notch network.
Collapse
Affiliation(s)
- M K Jeon
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - E Kaemmerer
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
- Department of Pediatrics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - U Schneider
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - M Schiffer
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - C Klaus
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - J Hennings
- Department of Medicine III, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - T Clahsen
- Department of Pediatrics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - T Ackerstaff
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - M Niggemann
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52056, Aachen, Germany
| | - A Schippers
- Department of Pediatrics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - T Longerich
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - G Sellge
- Department of Medicine III, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - C Trautwein
- Department of Medicine III, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - N Wagner
- Department of Pediatrics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - C Liedtke
- Department of Medicine III, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - N Gassler
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Klinikum Braunschweig, Institute of Pathology, Celler Strasse 28, 38114, Braunschweig, Germany.
| |
Collapse
|
8
|
Dorfman T, Pollak Y, Sohotnik R, Coran AG, Bejar J, Sukhotnik I. Enhanced intestinal epithelial cell proliferation in diabetic rats correlates with β-catenin accumulation. J Endocrinol 2015; 226:135-43. [PMID: 26297291 DOI: 10.1530/joe-14-0725] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The Wnt/β-catenin signaling cascade is implicated in the control of stem cell activity, cell proliferation, and cell survival of the gastrointestinal epithelium. Recent evidence indicates that the Wnt/β-catenin pathway is activated under diabetic conditions. The purpose of this study was to evaluate the role of Wnt/β-catenin signaling during diabetes-induced enteropathy in a rat model. Male rats were divided into three groups: control rats received injections of vehicle; diabetic rats received injections of one dose of streptozotocin (STZ); and diabetic-insulin rats received injections of STZ and were treated with insulin given subcutaneously at a dose of 1 U/kg twice daily. Rats were killed on day 7. Wnt/β-catenin-related genes and expression of proteins was determined using real-time PCR, western blotting, and immunohistochemistry. Among 13 genes identified by real-time PCR, seven genes were upregulated in diabetic rats compared with control animals including the target genes c-Myc and Tcf4. Diabetic rats also showed a significant increase in β-catenin protein compared with control animals. Treatment of diabetic rats attenuated the stimulating effect of diabetes on intestinal cell proliferation and Wnt/β-catenin signaling. In conclusion, enhanced intestinal epithelial cell proliferation in diabetic rats correlates with β-catenin accumulation.
Collapse
Affiliation(s)
- Tatiana Dorfman
- Laboratory of Intestinal Adaptation and RecoveryThe Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, IsraelDepartments of Pediatric Surgery BPathologyBnai Zion Medical Center, 47 Golomb Street, PO Box 4940, Haifa 31048, IsraelSection of Pediatric SurgeryC.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yulia Pollak
- Laboratory of Intestinal Adaptation and RecoveryThe Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, IsraelDepartments of Pediatric Surgery BPathologyBnai Zion Medical Center, 47 Golomb Street, PO Box 4940, Haifa 31048, IsraelSection of Pediatric SurgeryC.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Rima Sohotnik
- Laboratory of Intestinal Adaptation and RecoveryThe Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, IsraelDepartments of Pediatric Surgery BPathologyBnai Zion Medical Center, 47 Golomb Street, PO Box 4940, Haifa 31048, IsraelSection of Pediatric SurgeryC.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Arnold G Coran
- Laboratory of Intestinal Adaptation and RecoveryThe Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, IsraelDepartments of Pediatric Surgery BPathologyBnai Zion Medical Center, 47 Golomb Street, PO Box 4940, Haifa 31048, IsraelSection of Pediatric SurgeryC.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jacob Bejar
- Laboratory of Intestinal Adaptation and RecoveryThe Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, IsraelDepartments of Pediatric Surgery BPathologyBnai Zion Medical Center, 47 Golomb Street, PO Box 4940, Haifa 31048, IsraelSection of Pediatric SurgeryC.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Igor Sukhotnik
- Laboratory of Intestinal Adaptation and RecoveryThe Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, IsraelDepartments of Pediatric Surgery BPathologyBnai Zion Medical Center, 47 Golomb Street, PO Box 4940, Haifa 31048, IsraelSection of Pediatric SurgeryC.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA Laboratory of Intestinal Adaptation and RecoveryThe Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, IsraelDepartments of Pediatric Surgery BPathologyBnai Zion Medical Center, 47 Golomb Street, PO Box 4940, Haifa 31048, IsraelSection of Pediatric SurgeryC.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
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: 30] [Impact Index Per Article: 3.0] [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
|
10
|
Horvay K, Jardé T, Casagranda F, Perreau VM, Haigh K, Nefzger CM, Akhtar R, Gridley T, Berx G, Haigh JJ, Barker N, Polo JM, Hime GR, Abud HE. Snai1 regulates cell lineage allocation and stem cell maintenance in the mouse intestinal epithelium. EMBO J 2015; 34:1319-35. [PMID: 25759216 DOI: 10.15252/embj.201490881] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/02/2015] [Indexed: 12/17/2022] Open
Abstract
Snail family members regulate epithelial-to-mesenchymal transition (EMT) during invasion of intestinal tumours, but their role in normal intestinal homeostasis is unknown. Studies in breast and skin epithelia indicate that Snail proteins promote an undifferentiated state. Here, we demonstrate that conditional knockout of Snai1 in the intestinal epithelium results in apoptotic loss of crypt base columnar stem cells and bias towards differentiation of secretory lineages. In vitro organoid cultures derived from Snai1 conditional knockout mice also undergo apoptosis when Snai1 is deleted. Conversely, ectopic expression of Snai1 in the intestinal epithelium in vivo results in the expansion of the crypt base columnar cell pool and a decrease in secretory enteroendocrine and Paneth cells. Following conditional deletion of Snai1, the intestinal epithelium fails to produce a proliferative response following radiation-induced damage indicating a fundamental requirement for Snai1 in epithelial regeneration. These results demonstrate that Snai1 is required for regulation of lineage choice, maintenance of CBC stem cells and regeneration of the intestinal epithelium following damage.
Collapse
Affiliation(s)
- Katja Horvay
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| | - Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| | - Franca Casagranda
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Victoria M Perreau
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Katharina Haigh
- Australian Centre for Blood Diseases, Monash University & Alfred Health, Melbourne, Vic., Australia Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christian M Nefzger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia Australian Regenerative Medicine Institute, Monash University, Clayton, Vic., Australia
| | - Reyhan Akhtar
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| | - Thomas Gridley
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium Molecular and Cellular Oncology, Inflammation Research Center, VIB, Ghent, Belgium
| | - Jody J Haigh
- Australian Centre for Blood Diseases, Monash University & Alfred Health, Melbourne, Vic., Australia Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Nick Barker
- A*STAR Institute of Medical Biology, Singapore City, Singapore
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia Australian Regenerative Medicine Institute, Monash University, Clayton, Vic., Australia
| | - Gary R Hime
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| |
Collapse
|
11
|
Sukhotnik I, Geyer T, Pollak Y, Mogilner JG, Coran AG, Berkowitz D. The role of Wnt/β-catenin signaling in enterocyte turnover during methotrexate-induced intestinal mucositis in a rat. PLoS One 2014; 9:e110675. [PMID: 25375224 PMCID: PMC4222808 DOI: 10.1371/journal.pone.0110675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 09/21/2014] [Indexed: 01/16/2023] Open
Abstract
Background/Aims Intestinal mucositis is a common side-effect in patients who receive aggressive chemotherapy. The Wnt signaling pathway is critical for establishing and maintaining the proliferative compartment of the intestine. In the present study, we tested whether Wnt/β-catenin signaling is involved in methotrexate (MTX)-induced intestinal damage in a rat model. Methods Non-pretreated and pretreated with MTX Caco-2 cells were evaluated for cell proliferation and apoptosis using FACS analysis. Adult rats were divided into three experimental groups: Control rats; MTX-2 animals were treated with a single dose of MTX given IP and were sacrificed on day 2, and MTX-4 rats were treated with MTX similar to group B and were sacrificed on day 4. Intestinal mucosal damage, mucosal structural changes, enterocyte proliferation, and enterocyte apoptosis were measured at sacrifice. Real Time PCR and Western blot was used to determine the level of Wnt/β-catenin related genes and protein expression. Results In the vitro experiment, treatment with MTX resulted in marked decrease in early cell proliferation rates following by a 17-fold increase in late cell proliferation rates compared to early proliferation. Treatment with MTX resulted in a significant increase in early and late apoptosis compared to Caco-2 untreated cells. In the vivo experiment, MTX-2 and MTX-4 rats demonstrated intestinal mucosal hypoplasia. MTX-2 rats demonstrated a significant decrease in FRZ-2, Wnt 3A Wnt 5A, β-catenin, c-myc mRNA expression and a significant decrease in β-catenin and Akt protein levels compared to control animals. Four days following MTX administration, rats demonstrated a trend toward a restoration of Wnt/β-catenin signaling especially in ileum. Conclusions Wnt/β-catenin signaling is involved in enterocyte turnover during MTX-induced intestinal mucositis in a rat.
Collapse
Affiliation(s)
- Igor Sukhotnik
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Laboratory of intestinal adaptation and recovery, Dept of Pediatric Surgery, Bnai Zion Medical Center, Haifa, Israel
- * E-mail:
| | - Tatiana Geyer
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Laboratory of intestinal adaptation and recovery, Haifa, Israel
| | - Yulia Pollak
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Laboratory of intestinal adaptation and recovery, Haifa, Israel
| | - Jorge G. Mogilner
- The Bruce Rappaport Faculty of Medicine, Dept of Pediatric Surgery, Bnai Zion Medical Center, Haifa, Israel
| | - Arnold G. Coran
- Section of Pediatric Surgery C.S. Mott Children's Hospital and University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Drora Berkowitz
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Dept of Pediatric Gastroenterology, Bnai Zion Medical Center, Haifa, Israel
| |
Collapse
|
12
|
Abstract
BACKGROUND Growing evidence suggests that the Wnt/β-catenin signaling cascade is implicated in the control of stem cell activity, cell proliferation, lineage commitment, and cell survival during normal development and tissue regeneration of the gastrointestinal epithelium. The roles of this signaling cascade in stimulation of cell proliferation after massive small bowel resection are unknown. The purpose of this study was to evaluate the role of Wnt/β-catenin signaling during late stages of intestinal adaptation in a rat model of short bowel syndrome (SBS). METHODS Male rats were divided into two groups: sham rats underwent bowel transection and SBS rats underwent a 75 % bowel resection. Parameters of intestinal adaptation, enterocyte proliferation and apoptosis were determined 2 weeks after operation. Illumina's digital gene expression analysis was used to determine Wnt/β-catenin signaling gene expression profiling. Twelve Wnt/β-catenin-related genes and β-catenin protein expression were determined using real-time PCR, western blotting and immunohistochemistry. RESULTS From the total number of 20,000 probes, 20 genes related to Wnt/β-catenin signaling were investigated. From these genes, seven genes were found to be up-regulated and eight genes to be down-regulated in SBS vs. sham animals with a relative change in gene expression level of 20 % or more. From 12 genes determined by real-time PCR, nine genes were down-regulated in SBS rats compared to control animals including target gene c-Myc. SBS rats also showed a significant decrease in β-catenin protein compared to control animals. CONCLUSION Two weeks following massive bowel resection in rats, Wnt/β-catenin signaling pathway is inhibited. In addition, it appears that cell differentiation rather than proliferation is most important in the late stages of intestinal adaptation.
Collapse
|
13
|
Zhang K, Ai WB, Liu CB, Wu JF. Progress in understanding the relationship between Notch signaling pathway and hepatic stellate cell activation. Shijie Huaren Xiaohua Zazhi 2013; 21:3611-3616. [DOI: 10.11569/wcjd.v21.i33.3611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Multiple signaling pathways are involved in the pathogenesis of hepatic fibrosis, and the Notch signaling pathway plays an important role in promoting the activation of hepatic stellate cells (HSCs). This pathway participates in the activation of HSCs mainly by cooperating with transforming growth factor β (TGF-β)/BMP, nuclear factor-kappa B (NF-κB), and Wnt signaling pathways directly or indirectly. This review aims to explore the relationship between the Notch signaling pathway and the activation of HSCs as well as the cooperative actions between TGF-β/BMP, NF-κB, and Wnt and the Notch signaling pathway in the process of the activation of HSCs.
Collapse
|