501
|
Koch S. Extrinsic control of Wnt signaling in the intestine. Differentiation 2017; 97:1-8. [PMID: 28802143 DOI: 10.1016/j.diff.2017.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/28/2017] [Accepted: 08/04/2017] [Indexed: 12/15/2022]
Abstract
The canonical Wnt/β-catenin signaling pathway is a central regulator of development and tissue homeostasis. In the intestine, Wnt signaling is primarily known as the principal organizer of epithelial stem cell identity and proliferation. Within the last decade, numerous scientific breakthroughs have shed light on epithelial self-organization in the gut, and organoids are now routinely used to study stem cell biology and intestinal pathophysiology. The contribution of non-epithelial cells to Wnt signaling in the gut has received less attention. However, there is mounting evidence that stromal cells are a rich source of Wnt pathway activators and inhibitors, which can dynamically shape Wnt signaling to control epithelial proliferation and restitution. Elucidating the extent and mechanisms of paracrine Wnt signaling in the intestine has the potential to broaden our understanding of epithelial homeostasis, and may be of particular relevance for disorders such as inflammatory bowel diseases and colitis-associated cancers.
Collapse
Affiliation(s)
- Stefan Koch
- Department of Clinical and Experimental Medicine (IKE), Faculty of Health Sciences, Linköping University, S-581 85 Linköping, Sweden; Wallenberg Centre for Molecular Medicine (WCMM), Linköping University, S-581 85 Linköping, Sweden.
| |
Collapse
|
502
|
Intrinsic Autophagy Is Required for the Maintenance of Intestinal Stem Cells and for Irradiation-Induced Intestinal Regeneration. Cell Rep 2017; 20:1050-1060. [DOI: 10.1016/j.celrep.2017.07.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/24/2016] [Accepted: 07/10/2017] [Indexed: 12/19/2022] Open
|
503
|
Mills KM, Szczerkowski JLA, Habib SJ. Wnt ligand presentation and reception: from the stem cell niche to tissue engineering. Open Biol 2017; 7:rsob.170140. [PMID: 28814649 PMCID: PMC5577451 DOI: 10.1098/rsob.170140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/21/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cells reside in niches where spatially restricted signals maintain a delicate balance between stem cell self-renewal and differentiation. Wnt family proteins are particularly suited for this role as they are modified by lipids, which constrain and spatially regulate their signalling range. In recent years, Wnt/β-catenin signalling has been shown to be essential for the self-renewal of a variety of mammalian stem cells. In this review, we discuss Wnt-responsive stem cells in their niche, and mechanisms by which Wnt ligands are presented to responsive cells. We also highlight recent progress in molecular visualization that has allowed for the monitoring of Wnt signalling within the stem cell compartment and new approaches to recapitulate this niche signalling in vitro Indeed, new technologies that present Wnt in a localized manner and mimic the three-dimensional microenvironment of stem cells will advance our understanding of Wnt signalling in the stem cell niche. These advances will expand current horizons to exploit Wnt ligands in the rapidly evolving fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Kate M Mills
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - James L A Szczerkowski
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| |
Collapse
|
504
|
Yousefi M, Li L, Lengner CJ. Hierarchy and Plasticity in the Intestinal Stem Cell Compartment. Trends Cell Biol 2017; 27:753-764. [PMID: 28732600 DOI: 10.1016/j.tcb.2017.06.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 01/17/2023]
Abstract
Somatic stem cells maintain tissue homeostasis by organizing themselves in such a way that they can maintain proliferative output while simultaneously protecting themselves from DNA damage that may lead to oncogenic transformation. There is considerable debate about how such stem cell compartments are organized. Burgeoning evidence from the small intestine and colon provides support for a two-stem cell model involving an actively proliferating but injury-sensitive stem cell and a rare, injury-resistant pool of quiescent stem cells. Parallel with this evidence, recent studies have revealed considerable plasticity within the intestinal stem cell (ISC) compartment. We discuss the evidence for plasticity and hierarchy within the ISC compartment and how these properties govern tissue regeneration and contribute to oncogenic transformation leading to colorectal cancers.
Collapse
Affiliation(s)
- Maryam Yousefi
- Department of Biomedical Sciences, School of Veterinary Medicine and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, Missouri, MO 64110, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66101, USA.
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
505
|
Abstract
Purpose of review The intestine is a dynamic organ with rapid stem cell division generating epithelial cells that mature and apoptose in 3-5 days. Rapid turnover maintains the epithelial barrier and homeostasis. Current insights on intestinal stem cells (ISCs) and their regulation are discussed here. Recent findings The Lgr5+ ISCs maintain intestinal homeostasis by dividing asymmetrically, but also divide symmetrically to extinguish or replace ISCs. Following radiation or mucosal injury, reserve BMI1+ ISCs as well as other crypt cells can de-differentiate into Lgr5+ ISCs. ISC niche cells, including Paneth, immune and myofibroblast cells secrete factors that regulate ISC proliferation. Finally, several studies indicate that the microbiome metabolites regulate ISC growth. Summary ISC cells can be plastic and integrate a complexity of environmental/niche cues to trigger or suppress proliferation as needed.
Collapse
Affiliation(s)
- Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | | |
Collapse
|
506
|
Yan KS, Gevaert O, Zheng GXY, Anchang B, Probert CS, Larkin KA, Davies PS, Cheng ZF, Kaddis JS, Han A, Roelf K, Calderon RI, Cynn E, Hu X, Mandleywala K, Wilhelmy J, Grimes SM, Corney DC, Boutet SC, Terry JM, Belgrader P, Ziraldo SB, Mikkelsen TS, Wang F, von Furstenberg RJ, Smith NR, Chandrakesan P, May R, Chrissy MAS, Jain R, Cartwright CA, Niland JC, Hong YK, Carrington J, Breault DT, Epstein J, Houchen CW, Lynch JP, Martin MG, Plevritis SK, Curtis C, Ji HP, Li L, Henning SJ, Wong MH, Kuo CJ. Intestinal Enteroendocrine Lineage Cells Possess Homeostatic and Injury-Inducible Stem Cell Activity. Cell Stem Cell 2017; 21:78-90.e6. [PMID: 28686870 PMCID: PMC5642297 DOI: 10.1016/j.stem.2017.06.014] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 04/17/2017] [Accepted: 06/20/2017] [Indexed: 12/22/2022]
Abstract
Several cell populations have been reported to possess intestinal stem cell (ISC) activity during homeostasis and injury-induced regeneration. Here, we explored inter-relationships between putative mouse ISC populations by comparative RNA-sequencing (RNA-seq). The transcriptomes of multiple cycling ISC populations closely resembled Lgr5+ ISCs, the most well-defined ISC pool, but Bmi1-GFP+ cells were distinct and enriched for enteroendocrine (EE) markers, including Prox1. Prox1-GFP+ cells exhibited sustained clonogenic growth in vitro, and lineage-tracing of Prox1+ cells revealed long-lived clones during homeostasis and after radiation-induced injury in vivo. Single-cell mRNA-seq revealed two subsets of Prox1-GFP+ cells, one of which resembled mature EE cells while the other displayed low-level EE gene expression but co-expressed tuft cell markers, Lgr5 and Ascl2, reminiscent of label-retaining secretory progenitors. Our data suggest that the EE lineage, including mature EE cells, comprises a reservoir of homeostatic and injury-inducible ISCs, extending our understanding of cellular plasticity and stemness.
Collapse
Affiliation(s)
- Kelley S Yan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Columbia Center for Human Development, Columbia Stem Cell Initiative, Department of Medicine, Division of Digestive and Liver Diseases, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Olivier Gevaert
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Benedict Anchang
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christopher S Probert
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kathryn A Larkin
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paige S Davies
- Oregon Health & Science University, Department of Cell, Developmental and Cancer Biology, Portland, OR 97239, USA
| | - Zhuan-Fen Cheng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John S Kaddis
- Department of Diabetes and Cancer Discovery Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Arnold Han
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Columbia Center for Translational Immunology, Department of Medicine, Division of Digestive and Liver Diseases, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Kelly Roelf
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ruben I Calderon
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Department of Medicine, Division of Digestive and Liver Diseases, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Esther Cynn
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Department of Medicine, Division of Digestive and Liver Diseases, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Xiaoyi Hu
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Department of Medicine, Division of Digestive and Liver Diseases, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Komal Mandleywala
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Department of Medicine, Division of Digestive and Liver Diseases, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Julie Wilhelmy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sue M Grimes
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David C Corney
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | - Fengchao Wang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Nicholas R Smith
- Oregon Health & Science University, Department of Cell, Developmental and Cancer Biology, Portland, OR 97239, USA
| | - Parthasarathy Chandrakesan
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Randal May
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mary Ann S Chrissy
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Joyce C Niland
- Department of Diabetes and Cancer Discovery Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Young-Kwon Hong
- Departments of Surgery and of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jill Carrington
- National Institutes of Health, Division of Digestive Diseases and Nutrition, NIDDK, Bethesda, MD 20892, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jonathan Epstein
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Courtney W Houchen
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - John P Lynch
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Martin G Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sylvia K Plevritis
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christina Curtis
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hanlee P Ji
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Susan J Henning
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Melissa H Wong
- Oregon Health & Science University, Department of Cell, Developmental and Cancer Biology, Portland, OR 97239, USA
| | - Calvin J Kuo
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
507
|
Multifaceted Interpretation of Colon Cancer Stem Cells. Int J Mol Sci 2017; 18:ijms18071446. [PMID: 28678194 PMCID: PMC5535937 DOI: 10.3390/ijms18071446] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 12/11/2022] Open
Abstract
Colon cancer is one of the leading causes of cancer-related deaths worldwide, despite recent advances in clinical oncology. Accumulating evidence sheds light on the existence of cancer stem cells and their role in conferring therapeutic resistance. Cancer stem cells are a minor fraction of cancer cells, which enable tumor heterogeneity and initiate tumor formation. In addition, these cells are resistant to various cytotoxic factors. Therefore, elimination of cancer stem cells is difficult but essential to cure the malignant foci completely. Herein, we review the recent evidence for intestinal stem cells and colon cancer stem cells, methods to detect the tumor-initiating cells, and clinical significance of cancer stem cell markers. We also describe the emerging problems of cancer stem cell theory, including bidirectional conversion and intertumoral heterogeneity of stem cell phenotype.
Collapse
|
508
|
Flanagan DJ, Vincan E, Phesse TJ. Winding back Wnt signalling: potential therapeutic targets for treating gastric cancers. Br J Pharmacol 2017; 174:4666-4683. [PMID: 28568899 DOI: 10.1111/bph.13890] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/17/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer persists as a frequent and deadly disease that claims over 700 000 lives annually. Gastric cancer is a multifactorial disease that is genetically, cytologically and architecturally more heterogeneous than other gastrointestinal cancers, making it therapeutically challenging. As such, and largely attributed to late-stage diagnosis, gastric cancer patients show only partial response to standard chemo and targeted molecular therapies, highlighting an urgent need to develop new targeted therapies for this disease. Wnt signalling has a well-documented history in the genesis of many cancers and is, therefore, an attractive therapeutic target. As such, drug discovery has focused on developing inhibitors that target multiple nodes of the Wnt signalling cascade, some of which have progressed to clinical trials. The collective efforts of patient genomic profiling has uncovered genetic lesions to multiple components of the Wnt pathway in gastric cancer patients, which strongly suggest that Wnt-targeted therapies could offer therapeutic benefits for gastric cancer patients. These data have been supported by studies in mouse models of gastric cancer, which identify Wnt signalling as a driver of gastric tumourigenesis. Here, we review the current literature regarding Wnt signalling in gastric cancer and highlight the suitability of each class of Wnt inhibitor as a potential treatment for gastric cancer patients, in relation to the type of Wnt deregulation observed. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Dustin J Flanagan
- Molecular Oncology Laboratory, University of Melbourne, Melbourne, VIC, Australia.,Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, VIC, Australia
| | - Elizabeth Vincan
- Molecular Oncology Laboratory, University of Melbourne, Melbourne, VIC, Australia.,Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, VIC, Australia.,School of Biomedical Sciences, Curtin University, Perth, WA, Australia
| | - Toby J Phesse
- Molecular Oncology Laboratory, University of Melbourne, Melbourne, VIC, Australia.,Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, VIC, Australia.,Cell Signalling and Cancer Laboratory, European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
509
|
Smith RJ, Rao-Bhatia A, Kim TH. Signaling and epigenetic mechanisms of intestinal stem cells and progenitors: insight into crypt homeostasis, plasticity, and niches. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017. [PMID: 28644919 DOI: 10.1002/wdev.281] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The rapid turnover of intestinal epithelial cells is maintained by a small number of stem cells located in pocket-like gland structures called crypts. While our understanding of the identity and function of intestinal stem cells (ISCs) has rapidly progressed, epigenetic and transcriptional regulation in crypt stem cell and progenitor pools remains an active field of investigation. Surrounded by various types of cells in the stroma, crypt progenitors display high levels of plasticity, harboring the ability to interconvert in the face of epithelial damage. Recent studies analyzing epigenetic patterns of intestinal epithelial cells have provided evidence that plasticity is maintained by a broadly permissive epigenomic state, wherein cell-lineage specification is directed through activation of signaling pathways and transcription factor (TF) expression. New studies also have shown that the ISC niche, which is comprised of surrounding epithelial and mesenchymal tissues, plays a crucial role in supporting the maintenance and differentiation of stem cells by providing contextual information in the form of signaling cascades, such as Wnt, Notch, and Hippo. These cascades ultimately govern TF expression to promote early cell-lineage decisions in both crypt stem cells and progenitors. Highlighting recent studies investigating signaling, transcriptional, and epigenetic mechanisms of intestinal epithelial cells, we will discuss the mechanisms underlying crypt homeostasis, plasticity, and niches. WIREs Dev Biol 2017, 6:e281. doi: 10.1002/wdev.281 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Ryan J Smith
- Program of Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Abilasha Rao-Bhatia
- Program of Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Tae-Hee Kim
- Program of Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
510
|
Jadhav U, Saxena M, O'Neill NK, Saadatpour A, Yuan GC, Herbert Z, Murata K, Shivdasani RA. Dynamic Reorganization of Chromatin Accessibility Signatures during Dedifferentiation of Secretory Precursors into Lgr5+ Intestinal Stem Cells. Cell Stem Cell 2017. [PMID: 28648363 DOI: 10.1016/j.stem.2017.05.001] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Replicating Lgr5+ stem cells and quiescent Bmi1+ cells behave as intestinal stem cells (ISCs) in vivo. Disrupting Lgr5+ ISCs triggers epithelial renewal from Bmi1+ cells, from secretory or absorptive progenitors, and from Paneth cell precursors, revealing a high degree of plasticity within intestinal crypts. Here, we show that GFP+ cells from Bmi1GFP mice are preterminal enteroendocrine cells and we identify CD69+CD274+ cells as related goblet cell precursors. Upon loss of native Lgr5+ ISCs, both populations revert toward an Lgr5+ cell identity. While active histone marks are distributed similarly between Lgr5+ ISCs and progenitors of both major lineages, thousands of cis elements that control expression of lineage-restricted genes are selectively open in secretory cells. This accessibility signature dynamically converts to that of Lgr5+ ISCs during crypt regeneration. Beyond establishing the nature of Bmi1GFP+ cells, these findings reveal how chromatin status underlies intestinal cell diversity and dedifferentiation to restore ISC function and intestinal homeostasis.
Collapse
Affiliation(s)
- Unmesh Jadhav
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Madhurima Saxena
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Nicholas K O'Neill
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Assieh Saadatpour
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard TH Chan School of Public Health, Boston, MA 02215, USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard TH Chan School of Public Health, Boston, MA 02215, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Zachary Herbert
- Molecular Biology Core Facility, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kazutaka Murata
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA.
| |
Collapse
|
511
|
Kretzschmar K, Clevers H. Organoids: Modeling Development and the Stem Cell Niche in a Dish. Dev Cell 2017; 38:590-600. [PMID: 27676432 DOI: 10.1016/j.devcel.2016.08.014] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 02/08/2023]
Abstract
Organoids are three-dimensional in-vitro-grown cell clusters with near-native microanatomy that arise from self-organizing mammalian pluripotent or adult stem cells. Although monolayer stem cell cultures were established more than 40 years ago, organoid technology has recently emerged as an essential tool for both fundamental and biomedical research. For developmental biologists, organoids provide powerful means for ex vivo modeling of tissue morphogenesis and organogenesis. Here we discuss how organoid cultures of the intestine and other tissues have been established and how they are utilized as an in vitro model system for stem cell research and developmental biology.
Collapse
Affiliation(s)
- Kai Kretzschmar
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands; Princess Máxima Centre, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, UMC Utrecht, 3584 CG Utrecht, the Netherlands.
| |
Collapse
|
512
|
|
513
|
Leushacke M, Tan SH, Wong A, Swathi Y, Hajamohideen A, Tan LT, Goh J, Wong E, Denil SLIJ, Murakami K, Barker N. Lgr5-expressing chief cells drive epithelial regeneration and cancer in the oxyntic stomach. Nat Cell Biol 2017; 19:774-786. [PMID: 28581476 DOI: 10.1038/ncb3541] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/28/2017] [Indexed: 02/08/2023]
Abstract
The daily renewal of the corpus epithelium is fuelled by adult stem cells residing within tubular glands, but the identity of these stem cells remains controversial. Lgr5 marks homeostatic stem cells and 'reserve' stem cells in multiple tissues. Here, we report Lgr5 expression in a subpopulation of chief cells in mouse and human corpus glands. Using a non-variegated Lgr5-2A-CreERT2 mouse model, we show by lineage tracing that Lgr5-expressing chief cells do not behave as corpus stem cells during homeostasis, but are recruited to function as stem cells to effect epithelial renewal following injury by activating Wnt signalling. Ablation of Lgr5+ cells severely impairs epithelial homeostasis in the corpus, indicating an essential role for these Lgr5+ cells in maintaining the homeostatic stem cell pool. We additionally define Lgr5+ chief cells as a major cell-of-origin of gastric cancer. These findings reveal clinically relevant insights into homeostasis, repair and cancer in the corpus.
Collapse
Affiliation(s)
| | - Si Hui Tan
- A*STAR Institute of Medical Biology, 138648, Singapore
| | - Angeline Wong
- A*STAR Institute of Medical Biology, 138648, Singapore
| | - Yada Swathi
- A*STAR Institute of Medical Biology, 138648, Singapore
| | | | | | - Jasmine Goh
- A*STAR Institute of Medical Biology, 138648, Singapore
| | - Esther Wong
- A*STAR Institute of Medical Biology, 138648, Singapore
| | | | - Kazuhiro Murakami
- Cancer Research Institute, Kanazawa University, Kakuma-machi Kanazawa 920-1192, Japan
| | - Nick Barker
- A*STAR Institute of Medical Biology, 138648, Singapore.,Cancer Research Institute, Kanazawa University, Kakuma-machi Kanazawa 920-1192, Japan.,Centre for Regenerative Medicine, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
514
|
Charepalli V, Reddivari L, Radhakrishnan S, Eriksson E, Xiao X, Kim SW, Shen F, Vijay-Kumar M, Li Q, Bhat VB, Knight R, Vanamala JKP. Pigs, Unlike Mice, Have Two Distinct Colonic Stem Cell Populations Similar to Humans That Respond to High-Calorie Diet prior to Insulin Resistance. Cancer Prev Res (Phila) 2017; 10:442-450. [PMID: 28576788 DOI: 10.1158/1940-6207.capr-17-0010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/10/2017] [Accepted: 05/30/2017] [Indexed: 12/12/2022]
Abstract
Basal colonic crypt stem cells are long lived and play a role in colon homeostasis. Previous evidence has shown that high-calorie diet (HCD) enhances colonic stem cell numbers and expansion of the proliferative zone, an important biomarker for colon cancer. However, it is not clear how HCD drives dysregulation of colon stem cell/colonocyte proliferative kinetics. We used a human-relevant pig model and developed an immunofluorescence technique to detect and quantify colonic stem cells. Pigs (n = 8/group) were provided either standard diet (SD; 5% fat) or HCD (23% fat) for 13 weeks. HCD- and SD-consuming pigs had similar total calorie intake, serum iron, insulin, and glucose levels. However, HCD elevated both colonic proliferative zone (KI-67) and stem cell zone (ASCL-2 and BMI-1). Proliferative zone correlated with elevated innate colonic inflammatory markers TLR-4, NF-κB, IL6, and lipocalin-2 (r ≥ 0.62, P = 0.02). Elevated gut bacterial phyla proteobacteria and firmicutes in HCD-consuming pigs correlated with proliferative and stem cell zone. Colonic proteome data revealed the upregulation of proteins involved in cell migration and proliferation and correlated with proliferative and stem cell zone expansion. Our study suggests that pig colon, unlike mice, has two distinct stem cells (ASCL-2 and BMI-1) similar to humans, and HCD increases expansion of colonic proliferative and stem cell zone. Thus, pig model can aid in the development of preventive strategies against gut bacterial dysbiosis and inflammation-promoted diseases, such as colon cancer. Cancer Prev Res; 10(8); 442-50. ©2017 AACR.
Collapse
Affiliation(s)
- Venkata Charepalli
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania
| | - Lavanya Reddivari
- Department of Plant Science, The Pennsylvania State University, University Park, Pennsylvania
| | - Sridhar Radhakrishnan
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania
| | - Elisabeth Eriksson
- Biotechnology, Applied Nutrition and Food Chemistry, Lund University, Lund, Sweden
| | - Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina
| | - Frank Shen
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania.,Department of Medicine, The Pennsylvania State University Medical Center, Hershey, Pennsylvania
| | - Qunhua Li
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania
| | | | - Rob Knight
- Department of Pediatrics, University of California, San Diego, California.,Department of Computer Science and Engineering, University of California, La Jolla, California
| | - Jairam K P Vanamala
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania. .,The Penn State Hershey Cancer Institute, Hershey, Pennsylvania.,Center for Molecular Immunology and Infectious Diseases, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
515
|
McKinley ET, Sui Y, Al-Kofahi Y, Millis BA, Tyska MJ, Roland JT, Santamaria-Pang A, Ohland CL, Jobin C, Franklin JL, Lau KS, Gerdes MJ, Coffey RJ. Optimized multiplex immunofluorescence single-cell analysis reveals tuft cell heterogeneity. JCI Insight 2017; 2:93487. [PMID: 28570279 DOI: 10.1172/jci.insight.93487] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Intestinal tuft cells are a rare, poorly understood cell type recently shown to be a critical mediator of type 2 immune response to helminth infection. Here, we present advances in segmentation algorithms and analytical tools for multiplex immunofluorescence (MxIF), a platform that enables iterative staining of over 60 antibodies on a single tissue section. These refinements have enabled a comprehensive analysis of tuft cell number, distribution, and protein expression profiles as a function of anatomical location and physiological perturbations. Based solely on DCLK1 immunoreactivity, tuft cell numbers were similar throughout the mouse small intestine and colon. However, multiple subsets of tuft cells were uncovered when protein coexpression signatures were examined, including two new intestinal tuft cell markers, Hopx and EGFR phosphotyrosine 1068. Furthermore, we identified dynamic changes in tuft cell number, composition, and protein expression associated with fasting and refeeding and after introduction of microbiota to germ-free mice. These studies provide a foundational framework for future studies of intestinal tuft cell regulation and demonstrate the utility of our improved MxIF computational methods and workflow for understanding cellular heterogeneity in complex tissues in normal and disease states.
Collapse
Affiliation(s)
- Eliot T McKinley
- Epithelial Biology Center and.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yunxia Sui
- General Electric Global Research Center, Niskayuna, New York, USA
| | - Yousef Al-Kofahi
- General Electric Global Research Center, Niskayuna, New York, USA
| | - Bryan A Millis
- Department of Cell and Developmental Biology.,Cell Imaging Shared Resource, and
| | - Matthew J Tyska
- Epithelial Biology Center and.,Department of Cell and Developmental Biology
| | - Joseph T Roland
- Epithelial Biology Center and.,Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | - Christian Jobin
- Department of Medicine.,Department of Infectious Diseases and Pathology, and.,Department of Anatomy and Cell Physiology, University of Florida, Gainesville, Florida, USA
| | - Jeffrey L Franklin
- Epithelial Biology Center and.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cell and Developmental Biology.,Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Ken S Lau
- Epithelial Biology Center and.,Department of Cell and Developmental Biology
| | - Michael J Gerdes
- General Electric Global Research Center, Niskayuna, New York, USA
| | - Robert J Coffey
- Epithelial Biology Center and.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cell and Developmental Biology.,Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
516
|
Abstract
Elimination of self-renewing cancer stem cells (CSCs) is necessary to permanently eradicate malignant tissues. In a recent article in Nature, de Sousa e Melo et al. (2017) reveal that intestinal tumors can contain dynamic pools of functionally distinct CSC populations, which seem to interconvert depending on the host tissue microenvironment.
Collapse
Affiliation(s)
- Piero Dalerba
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Department of Medicine (Division of Digestive and Liver Diseases), Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, NY 10032, USA.
| |
Collapse
|
517
|
Stem cell plasticity enables hair regeneration following Lgr5+ cell loss. Nat Cell Biol 2017; 19:666-676. [DOI: 10.1038/ncb3535] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 04/19/2017] [Indexed: 12/22/2022]
|
518
|
Goto N, Ueo T, Fukuda A, Kawada K, Sakai Y, Miyoshi H, Taketo MM, Chiba T, Seno H. Distinct Roles of HES1 in Normal Stem Cells and Tumor Stem-like Cells of the Intestine. Cancer Res 2017; 77:3442-3454. [PMID: 28536281 DOI: 10.1158/0008-5472.can-16-3192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/17/2017] [Accepted: 05/08/2017] [Indexed: 11/16/2022]
|
519
|
Hou Q, Ye L, Huang L, Yu Q. The Research Progress on Intestinal Stem Cells and Its Relationship with Intestinal Microbiota. Front Immunol 2017; 8:599. [PMID: 28588586 PMCID: PMC5440531 DOI: 10.3389/fimmu.2017.00599] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022] Open
Abstract
The intestine is home to trillions of microorganisms, and the vast diversity within this gut microbiota exists in a balanced state to protect the intestinal mucosal barrier. Research into the association of the intestinal microbiota with health and disease (including diet, nutrition, obesity, inflammatory bowel disease, and cancer) continues to expand, with the field advancing at a rapid rate. Intestinal stem cells (ISCs) are the fundamental component of the mucosal barrier; they undergo continuous proliferation to replace the epithelium, which is also intimately involved in intestinal diseases. The intestinal microbiota, such as Lactobacillus, communicates with ISCs both directly and indirectly to regulate the proliferation and differentiation of ISCs. Moreover, Salmonella infection significantly decreased the expression of intestinal stem cell markers Lgr5 and Bmi1. However, the detailed interaction of intestinal microbiota and ISCs are still unclear. This review considers the progress of research on the model and niches of ISCs, as well as the complex interplay between the gut microbiota and ISCs, which will be crucial for explaining the mechanisms of intestinal diseases related to imbalances in the intestinal microbiota and ISCs.
Collapse
Affiliation(s)
- Qihang Hou
- College of veterinary medicine, Nanjing Agricultural University, Nanjing, China
| | - Lulu Ye
- College of veterinary medicine, Nanjing Agricultural University, Nanjing, China
| | - Lulu Huang
- College of veterinary medicine, Nanjing Agricultural University, Nanjing, China
| | - Qinghua Yu
- College of veterinary medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
520
|
Colak S, Medema JP. Human colonic fibroblasts regulate stemness and chemotherapy resistance of colon cancer stem cells. Cell Cycle 2017; 15:1531-7. [PMID: 25483065 DOI: 10.4161/15384101.2014.973321] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
There is increasing evidence that cancers are heterogeneous and contain a hierarchical organization consisting of cancer stem cells and their differentiated cell progeny. These cancer stem cells are at the core of the tumor as they represent the clonogenic cells within a tumor. Moreover, these cells are considered to contain selective therapy resistance, which suggests a pivotal role in therapy resistance and tumor relapse. Here we show that differentiated cells can re-acquire stemness through factors secreted from fibroblasts. This induced CSC state also coincides with re-acquisition of resistance to chemotherapy. Resistance induced in newly formed CSCs is mediated by the anti-apoptotic molecule BCLXL and inhibition of BCLXL with the BH3 mimetic ABT-737 sensitizes these cancer cells toward chemotherapy. These data point to an important interplay between tumor cells and their microenvironment in the regulation of stemness and therapy resistance.
Collapse
Affiliation(s)
- S Colak
- a LEXOR (Laboratory of Experimental Oncology and Radiobiology), Center for Experimental Molecular Medicine Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands
| | - J P Medema
- a LEXOR (Laboratory of Experimental Oncology and Radiobiology), Center for Experimental Molecular Medicine Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands.,b Cancer Genomics Center , The Netherlands
| |
Collapse
|
521
|
Kretzschmar K, Clevers H. Wnt/β-catenin signaling in adult mammalian epithelial stem cells. Dev Biol 2017; 428:273-282. [PMID: 28526587 DOI: 10.1016/j.ydbio.2017.05.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/06/2023]
Abstract
Adult stem cells self-renew and replenish differentiated cells in various organs and tissues throughout a mammal's life. Over the last 25 years an ever-growing body of knowledge has unraveled the essential regulation of adult mammalian epithelia by the canonical Wnt signaling with its key intracellular effector β-catenin. In this review, we discuss the principles of the signaling pathway and its role in adult epithelial stem cells of the intestine and skin during homeostasis and tumorigenesis. We further highlight the research that led to the identification of new stem cell markers and methods to study adult stem cells ex vivo.
Collapse
Affiliation(s)
- Kai Kretzschmar
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, UMC Utrecht, 3584 CG Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, UMC Utrecht, 3584 CG Utrecht, The Netherlands; Princess Máxima Centre for Pediatric Oncology, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
522
|
Viswanathan V, Damle S, Zhang T, Opdenaker L, Modarai S, Accerbi M, Schmidt S, Green P, Galileo D, Palazzo J, Fields J, Haghighat S, Rigoutsos I, Gonye G, Boman BM. An miRNA Expression Signature for the Human Colonic Stem Cell Niche Distinguishes Malignant from Normal Epithelia. Cancer Res 2017; 77:3778-3790. [PMID: 28487386 DOI: 10.1158/0008-5472.can-16-2388] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/02/2017] [Accepted: 05/05/2017] [Indexed: 02/02/2023]
Abstract
Malignant transformation of tissue stem cells (SC) may be the root of most cancer. Accordingly, we identified miRNA expression patterns in the normal human colonic SC niche to understand how cancer stem cells (CSC) may arise. In profiling miRNA expression in SC-enriched crypt subsections isolated from fresh, normal surgical specimens, we identified 16 miRNAs that were differentially expressed in the crypt bottom, creating an SC signature for normal colonic epithelia (NCE). A parallel analysis of colorectal cancer tissues showed differential expression of 83 miRNAs relative to NCE. Within the 16 miRNA signature for the normal SC niche, we found that miR-206, miR-007-3, and miR-23b individually could distinguish colorectal cancer from NCE. Notably, miR-23b, which was increased in colorectal cancer, was predicted to target the SC-expressed G protein-coupled receptor LGR5. Cell biology investigations showed that miR-23b regulated CSC phenotypes globally at the level of proliferation, cell cycle, self-renewal, epithelial-mesenchymal transition, invasion, and resistance to the colorectal cancer chemotherapeutic agent 5-fluorouracil. In mechanistic experiments, we found that miR-23b decreased LGR5 expression and increased ALDH+ CSCs. CSC analyses confirmed that levels of LGR5 and miR-23b are inversely correlated in ALDH+ CSCs and that distinct subpopulations of LGR5+ and ALDH+ CSCs exist. Overall, our results define a critical function for miR-23b, which, by targeting LGR5, contributes to overpopulation of ALDH+ CSCs and colorectal cancer. Cancer Res; 77(14); 3778-90. ©2017 AACR.
Collapse
Affiliation(s)
- Vignesh Viswanathan
- Center for Translational Cancer Research, Helen F Graham Cancer Center and Research Institute, Newark, Delaware.,Department of Biological Sciences, University of Delaware, Newark, Delaware.,Department of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Shirish Damle
- Thomas Jefferson University and Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Tao Zhang
- Center for Translational Cancer Research, Helen F Graham Cancer Center and Research Institute, Newark, Delaware.,Department of Biological Sciences, University of Delaware, Newark, Delaware.,Thomas Jefferson University and Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Lynn Opdenaker
- Center for Translational Cancer Research, Helen F Graham Cancer Center and Research Institute, Newark, Delaware.,Department of Biological Sciences, University of Delaware, Newark, Delaware
| | - Shirin Modarai
- Center for Translational Cancer Research, Helen F Graham Cancer Center and Research Institute, Newark, Delaware.,Department of Biological Sciences, University of Delaware, Newark, Delaware
| | - Monica Accerbi
- Department of Plant and Soil Sciences, Delaware Biotechnology Institute, Newark, Delaware
| | - Skye Schmidt
- Department of Plant and Soil Sciences, Delaware Biotechnology Institute, Newark, Delaware
| | - Pamela Green
- Department of Plant and Soil Sciences, Delaware Biotechnology Institute, Newark, Delaware
| | - Deni Galileo
- Department of Biological Sciences, University of Delaware, Newark, Delaware
| | - Juan Palazzo
- Thomas Jefferson University and Kimmel Cancer Center, Philadelphia, Pennsylvania
| | | | - Sepehr Haghighat
- Center for Translational Cancer Research, Helen F Graham Cancer Center and Research Institute, Newark, Delaware.,Department of Biological Sciences, University of Delaware, Newark, Delaware.,Thomas Jefferson University and Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Isidore Rigoutsos
- Thomas Jefferson University and Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Greg Gonye
- Thomas Jefferson University and Kimmel Cancer Center, Philadelphia, Pennsylvania.,Nanostring Technologies, Seattle, Washington
| | - Bruce M Boman
- Center for Translational Cancer Research, Helen F Graham Cancer Center and Research Institute, Newark, Delaware. .,Department of Biological Sciences, University of Delaware, Newark, Delaware.,Thomas Jefferson University and Kimmel Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
523
|
Giroux V, Lento AA, Islam M, Pitarresi JR, Kharbanda A, Hamilton KE, Whelan KA, Long A, Rhoades B, Tang Q, Nakagawa H, Lengner CJ, Bass AJ, Wileyto EP, Klein-Szanto AJ, Wang TC, Rustgi AK. Long-lived keratin 15+ esophageal progenitor cells contribute to homeostasis and regeneration. J Clin Invest 2017; 127:2378-2391. [PMID: 28481227 DOI: 10.1172/jci88941] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 03/09/2017] [Indexed: 12/30/2022] Open
Abstract
The esophageal lumen is lined by a stratified squamous epithelium comprised of proliferative basal cells that differentiate while migrating toward the luminal surface and eventually desquamate. Rapid epithelial renewal occurs, but the specific cell of origin that supports this high proliferative demand remains unknown. Herein, we have described a long-lived progenitor cell population in the mouse esophageal epithelium that is characterized by expression of keratin 15 (Krt15). Genetic in vivo lineage tracing revealed that the Krt15 promoter marks a long-lived basal cell population able to self-renew, proliferate, and generate differentiated cells, consistent with a progenitor/stem cell population. Transcriptional profiling demonstrated that Krt15+ basal cells are molecularly distinct from Krt15- basal cells. Depletion of Krt15-derived cells resulted in decreased proliferation, thereby leading to atrophy of the esophageal epithelium. Further, Krt15+ cells were radioresistant and contributed to esophageal epithelial regeneration following radiation-induced injury. These results establish the presence of a long-lived and indispensable Krt15+ progenitor cell population that provides additional perspective on esophageal epithelial biology and the widely prevalent diseases that afflict this epithelium.
Collapse
Affiliation(s)
- Véronique Giroux
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ashley A Lento
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mirazul Islam
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jason R Pitarresi
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akriti Kharbanda
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathryn E Hamilton
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kelly A Whelan
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Apple Long
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ben Rhoades
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qiaosi Tang
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hiroshi Nakagawa
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam J Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - E Paul Wileyto
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andres J Klein-Szanto
- Department of Pathology and Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Timothy C Wang
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University, New York, New York, USA
| | - Anil K Rustgi
- Division of Gastroenterology, Department of Medicine, and.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
524
|
Jin Y, Patel PH, Kohlmaier A, Pavlovic B, Zhang C, Edgar BA. Intestinal Stem Cell Pool Regulation in Drosophila. Stem Cell Reports 2017; 8:1479-1487. [PMID: 28479306 PMCID: PMC5469868 DOI: 10.1016/j.stemcr.2017.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 01/07/2023] Open
Abstract
Intestinal epithelial renewal is mediated by intestinal stem cells (ISCs) that exist in a state of neutral drift, wherein individual ISC lineages are regularly lost and born but ISC numbers remain constant. To test whether an active mechanism maintains stem cell pools in the Drosophila midgut, we performed partial ISC depletion. In contrast to the mouse intestine, Drosophila ISCs failed to repopulate the gut after partial depletion. Even when the midgut was challenged to regenerate by infection, ISCs retained normal proportions of asymmetric division and ISC pools did not increase. We discovered, however, that the loss of differentiated midgut enterocytes (ECs) slows when ISC division is suppressed and accelerates when ISC division increases. This plasticity in rates of EC turnover appears to facilitate epithelial homeostasis even after stem cell pools are compromised. Our study identifies unique behaviors of Drosophila midgut cells that maintain epithelial homeostasis.
Collapse
Affiliation(s)
- Yinhua Jin
- Center for Molecular Biology, University of Heidelberg (ZMBH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Parthive H Patel
- Center for Molecular Biology, University of Heidelberg (ZMBH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexander Kohlmaier
- Center for Molecular Biology, University of Heidelberg (ZMBH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Bojana Pavlovic
- Center for Molecular Biology, University of Heidelberg (ZMBH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Chenge Zhang
- Center for Molecular Biology, University of Heidelberg (ZMBH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Bruce A Edgar
- Center for Molecular Biology, University of Heidelberg (ZMBH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
525
|
Non-equivalence of Wnt and R-spondin ligands during Lgr5 + intestinal stem-cell self-renewal. Nature 2017; 545:238-242. [PMID: 28467820 PMCID: PMC5641471 DOI: 10.1038/nature22313] [Citation(s) in RCA: 323] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 03/30/2017] [Indexed: 12/24/2022]
Abstract
The canonical Wnt/β-catenin signaling pathway governs diverse developmental, homeostatic and pathologic processes. Palmitoylated Wnt ligands engage cell surface Frizzled (Fzd) receptors and Lrp5/6 co-receptors enabling β-catenin nuclear translocation and Tcf/Lef-dependent gene transactivation1–3. Mutations in Wnt downstream signaling components have revealed diverse functions presumptively attributed to Wnt ligands themselves, although direct attribution remains elusive, as complicated by redundancy between 19 mammalian Wnts and 10 Fzds1 and Wnt hydrophobicity2,3. For example, individual Wnt ligand mutations have not revealed homeostatic phenotypes in the intestinal epithelium4, an archetypal canonical Wnt pathway-dependent rapidly self-renewing tissue whose regeneration is fueled by proliferative crypt Lgr5+ intestinal stem cells (ISCs)5–9. R-spondin ligands (Rspo1–4) engage distinct Lgr4-6 and Rnf43/Znrf3 receptor classes10–13, markedly potentiate canonical Wnt/β-catenin signaling and induce intestinal organoid growth in vitro and Lgr5+ ISCs in vivo8,14–17. However, the interchangeability, functional cooperation and relative contributions of Wnt versus Rspo ligands to in vivo canonical Wnt signaling and ISC biology remain unknown. Here, we deconstructed functional roles of Wnt versus Rspo ligands in the intestinal crypt stem cell niche. We demonstrate that the default fate of Lgr5+ ISCs is lineage commitment, escape from which requires both Rspo and Wnt ligands. However, gain-of-function studies using Rspo versus a novel non-lipidated Wnt analog reveal qualitatively distinct, non-interchangeable roles for these ligands in ISCs. Wnts are insufficient to induce Lgr5+ ISC self-renewal, but rather confer a basal competency by maintaining Rspo receptor expression that enables Rspo to actively drive and specify the extent of stem cell expansion. This functionally non-equivalent yet cooperative interplay between Wnt and Rspo ligands establishes a molecular precedent for regulation of mammalian stem cells by distinct priming and self-renewal factors, with broad implications for precision control of tissue regeneration.
Collapse
|
526
|
Wieck MM, Schlieve CR, Thornton ME, Fowler KL, Isani M, Grant CN, Hilton AE, Hou X, Grubbs BH, Frey MR, Grikscheit TC. Prolonged Absence of Mechanoluminal Stimulation in Human Intestine Alters the Transcriptome and Intestinal Stem Cell Niche. Cell Mol Gastroenterol Hepatol 2017; 3:367-388.e1. [PMID: 28462379 PMCID: PMC5403975 DOI: 10.1016/j.jcmgh.2016.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS For patients with short-bowel syndrome, intestinal adaptation is required to achieve enteral independence. Although adaptation has been studied extensively in animal models, little is known about this process in human intestine. We hypothesized that analysis of matched specimens with and without luminal flow could identify new potential therapeutic pathways. METHODS Fifteen paired human ileum samples were collected from children aged 2-20 months during ileostomy-reversal surgery after short-segment intestinal resection and diversion. The segment exposed to enteral feeding was denoted as fed, and the diverted segment was labeled as unfed. Morphometrics and cell differentiation were compared histologically. RNA Sequencing and Gene Ontology Enrichment Analysis identified over-represented and under-represented pathways. Immunofluorescence staining and Western blot evaluated proteins of interest. Paired data were compared with 1-tailed Wilcoxon rank-sum tests with a P value less than .05 considered significant. RESULTS Unfed ileum contained shorter villi, shallower crypts, and fewer Paneth cells. Genes up-regulated by the absence of mechanoluminal stimulation were involved in digestion, metabolism, and transport. Messenger RNA expression of LGR5 was significantly higher in unfed intestine, accompanied by increased levels of phosphorylated signal transducer and activator of transcription 3 protein, and CCND1 and C-MYC messenger RNA. However, decreased proliferation and fewer LGR5+, OLFM4+, and SOX9+ intestinal stem cells (ISCs) were observed in unfed ileum. CONCLUSIONS Even with sufficient systemic caloric intake, human ileum responds to the chronic absence of mechanoluminal stimulation by up-regulating brush-border enzymes, transporters, structural genes, and ISC genes LGR5 and ASCL2. These data suggest that unfed intestine is primed to replenish the ISC population upon re-introduction of enteral feeding. Therefore, the elucidation of pathways involved in these processes may provide therapeutic targets for patients with intestinal failure. RNA sequencing data are available at Gene Expression Omnibus series GSE82147.
Collapse
Affiliation(s)
- Minna M. Wieck
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
- Department of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
| | - Christopher R. Schlieve
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
- Department of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
| | - Matthew E. Thornton
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California
| | - Kathryn L. Fowler
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
| | - Mubina Isani
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
- Department of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
| | - Christa N. Grant
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
- Department of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
| | - Ashley E. Hilton
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Xiaogang Hou
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
| | - Brendan H. Grubbs
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California
| | - Mark R. Frey
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
- Department of Pediatrics and Biochemistry, Department of Molecular Biology, University of Southern California, Los Angeles, California
| | - Tracy C. Grikscheit
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Los Angeles, California
- Department of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
- Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
527
|
Yang H, Adam RC, Ge Y, Hua ZL, Fuchs E. Epithelial-Mesenchymal Micro-niches Govern Stem Cell Lineage Choices. Cell 2017; 169:483-496.e13. [PMID: 28413068 DOI: 10.1016/j.cell.2017.03.038] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/09/2017] [Accepted: 03/23/2017] [Indexed: 01/19/2023]
Abstract
Adult tissue stem cells (SCs) reside in niches, which, through intercellular contacts and signaling, influence SC behavior. Once activated, SCs typically give rise to short-lived transit-amplifying cells (TACs), which then progress to differentiate into their lineages. Here, using single-cell RNA-seq, we unearth unexpected heterogeneity among SCs and TACs of hair follicles. We trace the roots of this heterogeneity to micro-niches along epithelial-mesenchymal interfaces, where progenitors display molecular signatures reflective of spatially distinct local signals and intercellular interactions. Using lineage tracing, temporal single-cell analyses, and chromatin landscaping, we show that SC plasticity becomes restricted in a sequentially and spatially choreographed program, culminating in seven spatially arranged unilineage progenitors within TACs of mature follicles. By compartmentalizing SCs into micro-niches, tissues gain precise control over morphogenesis and regeneration: some progenitors specify lineages immediately, whereas others retain potency, preserving self-renewing features established early while progressively restricting lineages as they experience dynamic changes in microenvironment.
Collapse
Affiliation(s)
- Hanseul Yang
- Robin Neustein Laboratory of Mammalian Development and Cell Biology, The Rockefeller University, New York, NY 10065, USA
| | - Rene C Adam
- Robin Neustein Laboratory of Mammalian Development and Cell Biology, The Rockefeller University, New York, NY 10065, USA
| | - Yejing Ge
- Robin Neustein Laboratory of Mammalian Development and Cell Biology, The Rockefeller University, New York, NY 10065, USA
| | - Zhong L Hua
- Robin Neustein Laboratory of Mammalian Development and Cell Biology, The Rockefeller University, New York, NY 10065, USA
| | - Elaine Fuchs
- Robin Neustein Laboratory of Mammalian Development and Cell Biology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
528
|
Kharas MG, Lengner CJ. Stem Cells, Cancer, and MUSASHI in Blood and Guts. Trends Cancer 2017; 3:347-356. [PMID: 28718412 DOI: 10.1016/j.trecan.2017.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 01/08/2023]
Abstract
The mammalian MSI family of RNA-binding proteins (RBPs) have important roles as oncoproteins in an array of tumor types, including leukemias, glioblastomas, and pancreatic, breast, lung, and colorectal cancers. The mammalian Msi genes, Msi1 and Msi2, have been most thoroughly investigated in two highly proliferative tissues prone to oncogenic transformation: the hematopoietic lineage and the intestinal epithelium. Despite their vast phenotypic differences, MSI proteins appear to have an analogous role in governing the stem cell compartment in both of these tissues, potentially providing a paradigm for a broader understanding of MSI function and oncogenic activities. In this review, we focus on the function of MSI in the blood and the intestine, and discuss therapeutic strategies for targeting this pathway.
Collapse
Affiliation(s)
- Michael G Kharas
- Molecular Pharmacology Program and Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA.
| |
Collapse
|
529
|
Stokes K, Cooke A, Chang H, Weaver DR, Breault DT, Karpowicz P. The Circadian Clock Gene BMAL1 Coordinates Intestinal Regeneration. Cell Mol Gastroenterol Hepatol 2017; 4:95-114. [PMID: 28593182 PMCID: PMC5453906 DOI: 10.1016/j.jcmgh.2017.03.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/24/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The gastrointestinal syndrome is an illness of the intestine caused by high levels of radiation. It is characterized by extensive loss of epithelial tissue integrity, which initiates a regenerative response by intestinal stem and precursor cells. The intestine has 24-hour rhythms in many physiological functions that are believed to be outputs of the circadian clock: a molecular system that produces 24-hour rhythms in transcription/translation. Certain gastrointestinal illnesses are worsened when the circadian rhythms are disrupted, but the role of the circadian clock in gastrointestinal regeneration has not been studied. METHODS We tested the timing of regeneration in the mouse intestine during the gastrointestinal syndrome. The role of the circadian clock was tested genetically using the BMAL1 loss of function mouse mutant in vivo, and in vitro using intestinal organoid culture. RESULTS The proliferation of the intestinal epithelium follows a 24-hour rhythm during the gastrointestinal syndrome. The circadian clock runs in the intestinal epithelium during this pathologic state, and the loss of the core clock gene, BMAL1, disrupts both the circadian clock and rhythmic proliferation. Circadian activity in the intestine involves a rhythmic production of inflammatory cytokines and subsequent rhythmic activation of the JNK stress response pathway. CONCLUSIONS Our results show that a circadian rhythm in inflammation and regeneration occurs during the gastrointestinal syndrome. The study and treatment of radiation-induced illnesses, and other gastrointestinal illnesses, should consider 24-hour timing in physiology and pathology.
Collapse
Affiliation(s)
- Kyle Stokes
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, Canada
| | - Abrial Cooke
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, Canada
| | - Hanna Chang
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, Canada
| | - David R. Weaver
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - David T. Breault
- Harvard Stem Cell Institute, Cambridge, Massachusetts,Division of Endocrinology, Boston Children’s Hospital, Boston, Massachusetts,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Phillip Karpowicz
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, Canada,Correspondence Address correspondence to: Phillip Karpowicz, PhD, Department of Biological Sciences, University of Windsor, Windsor, Ontario, Canada N9B 3P4.Department of Biological SciencesUniversity of WindsorWindsorOntarioCanada N9B 3P4
| |
Collapse
|
530
|
Abstract
Optimizing the management of colorectal cancer (CRC) risk in IBD requires a fundamental understanding of the evolutionary process underpinning tumorigenesis. In IBD, clonal evolution begins long before the development of overt neoplasia, and is probably accelerated by the repeated cycles of epithelial wounding and repair that are characteristic of the condition. Here, we review the biological drivers of mutant clone selection in IBD with particular reference to the unique histological architecture of the intestinal epithelium coupled with the inflammatory microenvironment in IBD, and the unique mutation patterns seen in IBD-driven neoplasia when compared with sporadic adenomas and CRC. How these data can be leveraged as evolutionary-based biomarkers to predict cancer risk is discussed, as well as how the efficacy of CRC surveillance programmes and the management of dysplasia can be improved. From a research perspective, the longitudinal surveillance of patients with IBD provides an under-exploited opportunity to investigate the biology of the human gastrointestinal tract over space and time.
Collapse
Affiliation(s)
- Chang-Ho R Choi
- Evolution and Cancer Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Inflammatory Bowel Disease Unit, Level 4 St Mark's Hospital, Watford Road, London HA1 3UJ, UK
| | - Ibrahim Al Bakir
- Evolution and Cancer Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Inflammatory Bowel Disease Unit, Level 4 St Mark's Hospital, Watford Road, London HA1 3UJ, UK
| | - Ailsa L Hart
- Inflammatory Bowel Disease Unit, Level 4 St Mark's Hospital, Watford Road, London HA1 3UJ, UK
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
531
|
Ponsioen B, Snippert HJ. Cancer systems biology: Live imaging of intestinal tissue in health and disease. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.coisb.2017.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
532
|
Costa BP, Gonçalves AC, Abrantes AM, Alves R, Matafome P, Seiça R, Sarmento-Ribeiro AB, Botelho MF, Castro-Sousa F. Intestinal Epithelial Stem Cells: Distinct Behavior After Surgical Injury and Teduglutide Administration. J INVEST SURG 2017; 31:243-252. [PMID: 28362133 DOI: 10.1080/08941939.2017.1294217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous studies suggest that intestinal epithelial stem cells (IESC), critical drivers of homeostasis and regeneration, include two subpopulations: crypt-based columnar and "position +4" stem cells, identified by Lgr5 and Bmi1 biomarkers, respectively. Teduglutide is an enterotrophic counterpart of glucagon-like peptide 2. This study aimed to investigate the response of putative IESC to surgical injury and teduglutide administration on an animal model of intestinal resection and anastomosis. METHODS Wistar rats (n = 59) were distributed into four groups: "Ileal Resection" versus "Laparotomy", subsequently subdivided into "Postoperative Teduglutide Administration" versus "No Treatment"; and sacrificed at third or seventh days, with ileal sample harvesting. Flow cytometry was used to analyze epithelial stem cells with monoclonal antibodies against Lgr5, Bmi1 and also CD44, CD24, CD166, and Grp78 surface markers. RESULTS Surgical trauma induced an increase of epithelial stem cells population at third day (9.0 ± 0.3 versus 5.7 ± 0.3%, p = 0.0001), which was more intense and involved all subpopulations after ileal resection. At seventh day, teduglutide was significantly associated with higher proportion of Lgr5+/Bmi1- cells (5.8 ± 0.1 versus 2.9 ± 0.3%, p = 0.005) and, on the contrary, lower percentage of Lgr5-/Bmi1+ cells (0.03 ± 0.01 versus 1.9 ± 0.1%, p = 0.049) after ileal resection; and higher proportion of Lgr5+/Bmi1+ cells (1.7 ± 0.1 versus 1.1 ± 0.2%, p = 0.028) after isolated laparotomy. After surgery, Lgr5+/Bmi1- and Lgr5-/Bmi1+ subpopulations demonstrated an inverse correlation and both correlated negatively with Grp78 labeling index. Lgr5-/Bmi1+ and CD44+/CD24low/CD166+/Grp78+ cells proportions exhibited a high grade positive correlation. CONCLUSION Those observations support the existence of two epithelial stem cells subpopulations with distinct behavior after surgical injury and teduglutide treatment.
Collapse
Affiliation(s)
- Beatriz P Costa
- a Department of Surgery A , Hospital da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, EPE , Coimbra , Portugal.,b University Clinic of Surgery , Faculty of Medicine, University of Coimbra , Coimbra , Portugal
| | - Ana C Gonçalves
- c Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology Unit, Faculty of Medicine , University of Coimbra , Coimbra , Portugal.,d Center for Neuroscience and Cell Biology (CNC) and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine , University of Medicine , Coimbra , Portugal.,e Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Ana M Abrantes
- d Center for Neuroscience and Cell Biology (CNC) and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine , University of Medicine , Coimbra , Portugal.,e Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine , University of Coimbra , Coimbra , Portugal.,f Institute of Biophysics, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Raquel Alves
- c Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology Unit, Faculty of Medicine , University of Coimbra , Coimbra , Portugal.,d Center for Neuroscience and Cell Biology (CNC) and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine , University of Medicine , Coimbra , Portugal.,e Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Paulo Matafome
- d Center for Neuroscience and Cell Biology (CNC) and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine , University of Medicine , Coimbra , Portugal.,g Institute of Physiology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Raquel Seiça
- d Center for Neuroscience and Cell Biology (CNC) and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine , University of Medicine , Coimbra , Portugal.,g Institute of Physiology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Ana B Sarmento-Ribeiro
- c Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology Unit, Faculty of Medicine , University of Coimbra , Coimbra , Portugal.,d Center for Neuroscience and Cell Biology (CNC) and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine , University of Medicine , Coimbra , Portugal.,e Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine , University of Coimbra , Coimbra , Portugal.,h Department of Clinical Hematology , Coimbra University Hospital, Centro Hospitalar e Universitário de Coimbra, EPE , Coimbra , Portugal
| | - M Filomena Botelho
- d Center for Neuroscience and Cell Biology (CNC) and Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine , University of Medicine , Coimbra , Portugal.,e Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine , University of Coimbra , Coimbra , Portugal.,f Institute of Biophysics, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Francisco Castro-Sousa
- a Department of Surgery A , Hospital da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, EPE , Coimbra , Portugal.,b University Clinic of Surgery , Faculty of Medicine, University of Coimbra , Coimbra , Portugal
| |
Collapse
|
533
|
Visualization and targeting of LGR5 + human colon cancer stem cells. Nature 2017; 545:187-192. [PMID: 28355176 DOI: 10.1038/nature22081] [Citation(s) in RCA: 539] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 03/23/2017] [Indexed: 12/25/2022]
Abstract
The cancer stem cell (CSC) theory highlights a self-renewing subpopulation of cancer cells that fuels tumour growth. The existence of human CSCs is mainly supported by xenotransplantation of prospectively isolated cells, but their clonal dynamics and plasticity remain unclear. Here, we show that human LGR5+ colorectal cancer cells serve as CSCs in growing cancer tissues. Lineage-tracing experiments with a tamoxifen-inducible Cre knock-in allele of LGR5 reveal the self-renewal and differentiation capacity of LGR5+ tumour cells. Selective ablation of LGR5+ CSCs in LGR5-iCaspase9 knock-in organoids leads to tumour regression, followed by tumour regrowth driven by re-emerging LGR5+ CSCs. KRT20 knock-in reporter marks differentiated cancer cells that constantly diminish in tumour tissues, while reverting to LGR5+ CSCs and contributing to tumour regrowth after LGR5+ CSC ablation. We also show that combined chemotherapy potentiates targeting of LGR5+ CSCs. These data provide insights into the plasticity of CSCs and their potential as a therapeutic target in human colorectal cancer.
Collapse
|
534
|
Qin W, Zheng Y, Qian BZ, Zhao M. Prostate Cancer Stem Cells and Nanotechnology: A Focus on Wnt Signaling. Front Pharmacol 2017; 8:153. [PMID: 28400729 PMCID: PMC5368180 DOI: 10.3389/fphar.2017.00153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/09/2017] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer is the most common cancer among men worldwide. However, current treatments for prostate cancer patients in advanced stage often fail because of relapse. Prostate cancer stem cells (PCSCs) are resistant to most standard therapies, and are considered to be a major mechanism of cancer metastasis and recurrence. In this review, we summarized current understanding of PCSCs and their self-renewal signaling pathways with a specific focus on Wnt signaling. Although multiple Wnt inhibitors have been developed to target PCSCs, their application is still limited by inefficient delivery and toxicity in vivo. Recently, nanotechnology has opened a new avenue for cancer drug delivery, which significantly increases specificity and reduces toxicity. These nanotechnology-based drug delivery methods showed great potential in targeting PCSCs. Here, we summarized current advancement of nanotechnology-based therapeutic strategies for targeting PCSCs and highlighted the challenges and perspectives in designing future therapies to eliminate PCSCs.
Collapse
Affiliation(s)
- Wei Qin
- The Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China; Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China
| | - Yongjiang Zheng
- The Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, China
| | - Bin-Zhi Qian
- Edinburgh Cancer Research UK Centre and MRC University of Edinburgh Centre for Reproductive Health, University of Edinburgh Edinburgh, UK
| | - Meng Zhao
- The Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China; Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
535
|
Mutual reinforcement between telomere capping and canonical Wnt signalling in the intestinal stem cell niche. Nat Commun 2017; 8:14766. [PMID: 28303901 PMCID: PMC5357864 DOI: 10.1038/ncomms14766] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/27/2017] [Indexed: 12/30/2022] Open
Abstract
Critical telomere shortening (for example, secondary to partial telomerase deficiency in the rare disease dyskeratosis congenita) causes tissue pathology, but underlying mechanisms are not fully understood. Mice lacking telomerase (for example, mTR−/− telomerase RNA template mutants) provide a model for investigating pathogenesis. In such mice, after several generations of telomerase deficiency telomeres shorten to the point of uncapping, causing defects most pronounced in high-turnover tissues including intestinal epithelium. Here we show that late-generation mTR−/− mutants experience marked downregulation of Wnt pathway genes in intestinal crypt epithelia, including crypt base columnar stem cells and Paneth cells, and in underlying stroma. The importance of these changes was revealed by rescue of crypt apoptosis and Wnt pathway gene expression upon treatment with Wnt pathway agonists. Rescue was associated with reduced telomere-dysfunction-induced foci and anaphase bridges, indicating improved telomere capping. Thus a mutually reinforcing feedback loop exists between telomere capping and Wnt signalling, and telomere capping can be impacted by extracellular cues in a fashion independent of telomerase. Mice lacking telomerase provide a model to study pathogenesis caused by critical telomere shortening. Here, the authors provide evidence that telomere shortening causes downregulation of Wnt signalling in intestinal crypts and that defects can be partially rescued by treatment with Wnt agonists.
Collapse
|
536
|
Yamaguchi J, Yokoyama Y, Kokuryo T, Ebata T, Nagino M. Cells of origin of pancreatic neoplasms. Surg Today 2017; 48:9-17. [PMID: 28260136 DOI: 10.1007/s00595-017-1501-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/07/2017] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignant disease associated with poor prognosis, despite recent medical advances. It is of great importance to understand the initial events and cells of origin of pancreatic cancer to prevent the development and progression of PDAC. There are three distinct precursor lesions that develop into PDAC: pancreatic intraepithelial neoplasms (PanINs), intraductal papillary mucinous neoplasms (IPMNs), and mucinous cystic neoplasms (MCNs). Studies on genetically engineered mouse models have revealed that the initiation and development of these lesions largely depend on genetic alterations. These lesions originate from different populations in the pancreas. PanIN development seems to be the result of the transdifferentiation of acinar cells, whereas IPMNs most likely arise from the progenitor niche of the pancreatic ductal epithelium. Pancreatic carcinogenesis is dependent on various events, including gene alterations, environmental insults, and cell types. However, further studies are needed to fully understand the initial processes of pancreatic cancer.
Collapse
Affiliation(s)
- Junpei Yamaguchi
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan.
| | - Yukihiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| | - Masato Nagino
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8560, Japan
| |
Collapse
|
537
|
Fan P, Liu P, Song P, Chen X, Ma X. Moderate dietary protein restriction alters the composition of gut microbiota and improves ileal barrier function in adult pig model. Sci Rep 2017; 7:43412. [PMID: 28252026 PMCID: PMC5333114 DOI: 10.1038/srep43412] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
This study was conducted to investigate impacts of dietary protein levels on gut bacterial community and gut barrier. The intestinal microbiota of finishing pigs, fed with 16%, 13% and 10% crude protein (CP) in diets, respectively, were investigated using Illumina MiSeq sequencing. The ileal bacterial richness tended to decrease when the dietary protein concentration reduced from 16% to 10%. The proportion of Clostridium_sensu_stricto_1 in ileum significantly decreased, whereas Escherichia-Shigella increased with reduction of protein concentration. In colon, the proportion of Clostridium_sensu_stricto_1 and Turicibacter increased, while the proportion of RC9_gut_group significantly decreased with the dietary protein reduction. Notably, the proportion of Peptostreptococcaceae was higher in both ileum and colon of 13% CP group. As for metabolites, the intestinal concentrations of SCFAs and biogenic amines decreased with the dietary protein reduction. The 10% CP dietary treatment damaged ileal mucosal morphology, and decreased the expression of biomarks of intestinal cells (Lgr5 and Bmi1), whereas the expression of tight junction proteins (occludin and claudin) in 13% CP group were higher than the other two groups. In conclusion, moderate dietary protein restriction (13% CP) could alter the bacterial community and metabolites, promote colonization of beneficial bacteria in both ileum and colon, and improve gut barrier function.
Collapse
Affiliation(s)
- Peixin Fan
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Ping Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Peixia Song
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiyue Chen
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
- Department of Internal Medicine, Department of Biochemistry, Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9113, USA
| |
Collapse
|
538
|
López-Posadas R, Neurath MF, Atreya I. Molecular pathways driving disease-specific alterations of intestinal epithelial cells. Cell Mol Life Sci 2017; 74:803-826. [PMID: 27624395 PMCID: PMC11107577 DOI: 10.1007/s00018-016-2363-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/06/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022]
Abstract
Due to the fact that chronic inflammation as well as tumorigenesis in the gut is crucially impacted by the fate of intestinal epithelial cells, our article provides a comprehensive overview of the composition, function, regulation and homeostasis of the gut epithelium. In particular, we focus on those aspects which were found to be altered in the context of inflammatory bowel diseases or colorectal cancer and also discuss potential molecular targets for a disease-specific therapeutic intervention.
Collapse
Affiliation(s)
- Rocío López-Posadas
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Ulmenweg 18, 91054, Erlangen, Germany.
| |
Collapse
|
539
|
Hayakawa Y, Fox JG, Wang TC. The Origins of Gastric Cancer From Gastric Stem Cells: Lessons From Mouse Models. Cell Mol Gastroenterol Hepatol 2017; 3:331-338. [PMID: 28462375 PMCID: PMC5404024 DOI: 10.1016/j.jcmgh.2017.01.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/28/2017] [Indexed: 12/24/2022]
Abstract
The cellular origin of digestive cancers has been a long-standing question in the cancer field. Mouse models have identified long-lived stem cells in most organ systems, including the luminal gastrointestinal tract, and numerous studies have pointed to tissue resident stem cells as the main cellular origin of cancer. During gastric carcinogenesis, chronic inflammation induces genetic and epigenetic alterations in long-lived stem cells, along with expansion of stem cell niches, eventually leading to invasive cancer. The gastric corpus and antrum have distinct stem cells and stem cell niches, suggesting differential regulation of cancer initiation at the 2 sites. In this short review, we discuss recent experimental models and human studies, which provide important insights into the pathogenesis of gastric cancer.
Collapse
Affiliation(s)
- Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York,Correspondence Address correspondence to: Timothy C. Wang, MD, Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, 1130 St Nicholas Avenue, Room 925, New York, New York 10032-3802. fax: (212) 851-4590.Division of Digestive and Liver DiseasesDepartment of Medicine and Irving Cancer Research CenterColumbia University Medical Center1130 St Nicholas AvenueRoom 925New YorkNew York 10032-3802
| |
Collapse
|
540
|
Abstract
The corneal epithelium acts as a protective barrier on the anterior ocular surface and is essential for maintaining transparency of the cornea and thus visual acuity. During both homeostasis and repair, the corneal epithelium is maintained by self-renewing stem cells, which persist throughout the lifetime of the organism. Importantly, as in other self-renewing tissues, the functional activity of corneal epithelial stem cells (CSCEs) is tightly regulated by the surrounding microenvironment, or niche, which provides a range of cues that maintain the stem cell population. This Cell Science at a Glance article and the accompanying poster will therefore aim to summarise our current understanding of the corneal epithelial stem cell niche and its role in regulating stem cell activity during homeostasis, repair and disease.
Collapse
Affiliation(s)
- Craig S Nowell
- CMU, Department for Pathology and Immunology, Rue Michel Servet, 1211 Geneva, Switzerland
| | - Freddy Radtke
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research, Lausanne, Vaud, 1015, Switzerland
| |
Collapse
|
541
|
Induced Quiescence of Lgr5+ Stem Cells in Intestinal Organoids Enables Differentiation of Hormone-Producing Enteroendocrine Cells. Cell Stem Cell 2017; 20:177-190.e4. [DOI: 10.1016/j.stem.2016.11.001] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 08/30/2016] [Accepted: 10/28/2016] [Indexed: 02/06/2023]
|
542
|
Abstract
The development of sustainable intestinal organoid cell culture has emerged as a new modality for the study of intestinal function and cellular processes. Organoid culture is providing a new testbed for therapeutic research and development. Intestinal organoids, self-renewing 3-dimensional structures comprised intestinal stem cells and their differentiated epithelial progeny allow for more facile and robust exploration of cellular activity, cell organization and structure, genetic manipulation, and vastly more physiologic modeling of intestinal response to stimuli as compared to traditional 2-dimensional cell line cultures. Intestinal organoids are affecting a wide variety of research into gastrointestinal pathology. The purpose of this review is to discuss the current state-of-the-art and future effect of research using enteroids and colonoids (organoids grown from the small and large intestines, respectively).
Collapse
|
543
|
Smith NR, Davies PS, Levin TG, Gallagher AC, Keene DR, Sengupta SK, Wieghard N, El Rassi E, Wong MH. Cell Adhesion Molecule CD166/ALCAM Functions Within the Crypt to Orchestrate Murine Intestinal Stem Cell Homeostasis. Cell Mol Gastroenterol Hepatol 2017; 3:389-409. [PMID: 28462380 PMCID: PMC5404029 DOI: 10.1016/j.jcmgh.2016.12.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 12/04/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Intestinal epithelial homeostasis is maintained by active-cycling and slow-cycling stem cells confined within an instructive crypt-based niche. Exquisite regulating of these stem cell populations along the proliferation-to-differentiation axis maintains a homeostatic balance to prevent hyperproliferation and cancer. Although recent studies focus on how secreted ligands from mesenchymal and epithelial populations regulate intestinal stem cells (ISCs), it remains unclear what role cell adhesion plays in shaping the regulatory niche. Previously we have shown that the cell adhesion molecule and cancer stem cell marker, CD166/ALCAM (activated leukocyte cell adhesion molecule), is highly expressed by both active-cycling Lgr5+ ISCs and adjacent Paneth cells within the crypt base, supporting the hypothesis that CD166 functions to mediate ISC maintenance and signal coordination. METHODS Here we tested this hypothesis by analyzing a CD166-/- mouse combined with immunohistochemical, flow cytometry, gene expression, and enteroid culture. RESULTS We found that animals lacking CD166 expression harbored fewer active-cycling Lgr5+ ISCs. Homeostasis was maintained by expansion of the transit-amplifying compartment and not by slow-cycling Bmi1+ ISC stimulation. Loss of active-cycling ISCs was coupled with deregulated Paneth cell homeostasis, manifested as increased numbers of immature Paneth progenitors due to decreased terminal differentiation, linked to defective Wnt signaling. CD166-/- Paneth cells expressed reduced Wnt3 ligand expression and depleted nuclear β-catenin. CONCLUSIONS These data support a function for CD166 as an important cell adhesion molecule that shapes the signaling microenvironment by mediating ISC-niche cell interactions. Furthermore, loss of CD166 expression results in decreased ISC and Paneth cell homeostasis and an altered Wnt microenvironment.
Collapse
Key Words
- BrdU, bromodeoxyuridine
- CD166
- CLEM, correlative light and electron microscopy
- FACS, fluorescence-activated cell sorting
- FITC, fluorescein isothiocyanate
- GFP, green fluorescent protein
- HBSS, Hank’s balanced salt solution
- Homeostasis
- IHC, immunohistochemistry
- ISC, intestinal stem cell
- Intestinal Stem Cell
- Lyz, lysozyme
- Muc2, mucin 2
- Paneth Cell
- SEM, standard error of the mean
- Stem Cell Niche
- TA, transit-amplifying
- TEM, transmission electron microscopy
- WT, wild-type
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
Collapse
Affiliation(s)
- Nicholas R. Smith
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | - Paige S. Davies
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | - Trevor G. Levin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Alexandra C. Gallagher
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Sidharth K. Sengupta
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA
| | - Nikki Wieghard
- Department of Surgery, Oregon Health & Science University, Portland, Oregon
| | - Edward El Rassi
- Department of Otolaryngology, Oregon Health & Science University, Portland, Oregon
| | - Melissa H. Wong
- Department of Cell, Developmental and Cancer Biology and Oregon Health & Science University, Portland, OR 97239, USA,OHSU Stem Cell Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon,Correspondence Address correspondence to: Melissa H. Wong, PhD, Oregon Health & Science University, Department of Cell, Developmental and Cancer Biology, 3181 SW Sam Jackson Park Road, Mail Code L215, Portland, Oregon 97239. fax: (503) 494-4253.Oregon Health & Science UniversityDepartment of CellDevelopmental and Cancer Biology3181 SW Sam Jackson Park RoadMail Code L215PortlandOregon 97239
| |
Collapse
|
544
|
Zhu X, Ching T, Pan X, Weissman SM, Garmire L. Detecting heterogeneity in single-cell RNA-Seq data by non-negative matrix factorization. PeerJ 2017; 5:e2888. [PMID: 28133571 PMCID: PMC5251935 DOI: 10.7717/peerj.2888] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 12/08/2016] [Indexed: 01/08/2023] Open
Abstract
Single-cell RNA-Sequencing (scRNA-Seq) is a fast-evolving technology that enables the understanding of biological processes at an unprecedentedly high resolution. However, well-suited bioinformatics tools to analyze the data generated from this new technology are still lacking. Here we investigate the performance of non-negative matrix factorization (NMF) method to analyze a wide variety of scRNA-Seq datasets, ranging from mouse hematopoietic stem cells to human glioblastoma data. In comparison to other unsupervised clustering methods including K-means and hierarchical clustering, NMF has higher accuracy in separating similar groups in various datasets. We ranked genes by their importance scores (D-scores) in separating these groups, and discovered that NMF uniquely identifies genes expressed at intermediate levels as top-ranked genes. Finally, we show that in conjugation with the modularity detection method FEM, NMF reveals meaningful protein-protein interaction modules. In summary, we propose that NMF is a desirable method to analyze heterogeneous single-cell RNA-Seq data. The NMF based subpopulation detection package is available at: https://github.com/lanagarmire/NMFEM.
Collapse
Affiliation(s)
- Xun Zhu
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, United States; Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, United States
| | - Travers Ching
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, United States; Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, United States
| | - Xinghua Pan
- Department of Genetics, Yale University , New Haven , CT , United States
| | - Sherman M Weissman
- Department of Genetics, Yale University , New Haven , CT , United States
| | - Lana Garmire
- Epidemiology Program, University of Hawaii Cancer Center , Honolulu , HI , United States
| |
Collapse
|
545
|
Augenlicht LH. Environmental Impact on Intestinal Stem Cell Functions in Mucosal Homeostasis and Tumorigenesis. J Cell Biochem 2017; 118:943-952. [PMID: 27584938 DOI: 10.1002/jcb.25719] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 08/30/2016] [Indexed: 12/22/2022]
Abstract
Multiple cell compartments at or near the base of the intestinal crypt have been identified as contributing intestinal stem cells for homeostasis of the rapidly turning over intestinal mucosa and cells that can initiate tumor development upon appropriate genetic changes. There is a strong literature establishing the importance of the frequently dividing Lgr5+ crypt base columnar cells as the fundamental cell in providing these stem cell-associated functions, but there are also clear data that more quiescent cells from other compartments can be mobilized to provide these stem cell functions upon compromise of Lgr5+ cells. We review the data that vitamin D, a pleiotropic hormone, is essential for Lgr5 stem cell functions by signaling through the vitamin D receptor. Moreover, we discuss the implications of this role of vitamin D and its impact on relatively long-lived stem cells in regards to the fact that virtually all the data on normal functioning of mouse Lgr5 stem cells is derived from mice exposed to vitamin D levels well above those that characterize the human population. Thus, there are still many questions regarding how dietary and environmental factors influence the complement of cells providing stem cell functions and the mechanisms by which this is determined, and the importance of this in human colorectal tumor development. J. Cell. Biochem. 118: 943-952, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Leonard H Augenlicht
- Departments of Medicine and Cell Biology, Albert Einstein College of Medicine, New York 10461, New York
| |
Collapse
|
546
|
Bartfeld S, Koo BK. Adult gastric stem cells and their niches. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017; 6. [PMID: 28044412 DOI: 10.1002/wdev.261] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/18/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022]
Abstract
Adult gastric stem cells replenish the gastric epithelium throughout life. Recent studies have identified diverse populations of stem cells, progenitor cells, and even differentiated cells that can regain stem cell capacity, so highlighting an unexpected plasticity within the gastric epithelium, both in the corpus and antrum. Two niches seem to co-exist in the gastric unit: one in the isthmus region and the other at the base of the gland, although the precise features of the cell populations and the two niches are currently under debate. A variety of gastric organoid models have been established, providing new insights into niche factors required by the gastric stem cell populations. Here we review our current knowledge of gastric stem cell populations, their markers and interactions, important niche factors, and different gastric organoid systems. WIREs Dev Biol 2017, 6:e261. doi: 10.1002/wdev.261 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Sina Bartfeld
- Research Centre for Infectious Diseases, University of Wuerzburg, Wuerzburg, Germany
| | - Bon-Kyoung Koo
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
547
|
Li L, Newton PT, Bouderlique T, Sejnohova M, Zikmund T, Kozhemyakina E, Xie M, Krivanek J, Kaiser J, Qian H, Dyachuk V, Lassar AB, Warman ML, Barenius B, Adameyko I, Chagin AS. Superficial cells are self-renewing chondrocyte progenitors, which form the articular cartilage in juvenile mice. FASEB J 2016; 31:1067-1084. [PMID: 27965322 DOI: 10.1096/fj.201600918r] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/22/2016] [Indexed: 01/20/2023]
Abstract
Articular cartilage has little regenerative capacity. Recently, genetic lineage tracing experiments have revealed chondrocyte progenitors at the articular surface. We further characterized these progenitors by using in vivo genetic approaches. Histone H2B-green fluorescent protein retention revealed that superficial cells divide more slowly than underlying articular chondrocytes. Clonal genetic tracing combined with immunohistochemistry revealed that superficial cells renew their number by symmetric division, express mesenchymal stem cell markers, and generate chondrocytes via both asymmetric and symmetric differentiation. Quantitative analysis of cellular kinetics, in combination with phosphotungstic acid-enhanced micro-computed tomography, showed that superficial cells generate chondrocytes and contribute to the growth and reshaping of articular cartilage. Furthermore, we found that cartilage renewal occurs as the progeny of superficial cells fully replace fetal chondrocytes during early postnatal life. Thus, superficial cells are self-renewing progenitors that are capable of maintaining their own population and fulfilling criteria of unipotent adult stem cells. Furthermore, the progeny of these cells reconstitute adult articular cartilage de novo, entirely substituting fetal chondrocytes.-Li, L., Newton, P. T., Bouderlique, T., Sejnohova, M., Zikmund, T., Kozhemyakina, E., Xie, M., Krivanek, J., Kaiser, J., Qian, H., Dyachuk, V., Lassar, A. B., Warman, M. L., Barenius, B., Adameyko, I., Chagin, A. S. Superficial cells are self-renewing chondrocyte progenitors, which form the articular cartilage in juvenile mice.
Collapse
Affiliation(s)
- Lei Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Phillip T Newton
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Thibault Bouderlique
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Sejnohova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Elena Kozhemyakina
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Meng Xie
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jan Krivanek
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Hong Qian
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Andrew B Lassar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew L Warman
- Orthopedic Research Labs, Boston Children's Hospital, Boston, Massachusetts, USA; and
| | - Björn Barenius
- Orthopaedic Section, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Andrei S Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden;
| |
Collapse
|
548
|
Hirsch D, Ried T. Targeting colorectal cancer (stem-like) cells using LGR5 directed antibody drug conjugates. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:508. [PMID: 28149870 DOI: 10.21037/atm.2016.11.78] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Daniela Hirsch
- Institute of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Ried
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
549
|
Yousefi M, Li N, Nakauka-Ddamba A, Wang S, Davidow K, Schoenberger J, Yu Z, Jensen ST, Kharas MG, Lengner CJ. Msi RNA-binding proteins control reserve intestinal stem cell quiescence. J Cell Biol 2016; 215:401-413. [PMID: 27799368 PMCID: PMC5100293 DOI: 10.1083/jcb.201604119] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/12/2016] [Accepted: 09/29/2016] [Indexed: 12/14/2022] Open
Abstract
Regeneration of the intestinal epithelium is driven by multiple intestinal stem cell (ISC) types, including an active, radiosensitive Wnthigh ISC that fuels turnover during homeostasis and a reserve, radioresistant Wntlow/off ISC capable of generating active Wnthigh ISCs. We examined the role of the Msi family of oncoproteins in the ISC compartment. We demonstrated that Msi proteins are dispensable for normal homeostasis and self-renewal of the active ISC, despite their being highly expressed in these cells. In contrast, Msi proteins are required specifically for activation of reserve ISCs, where Msi activity is both necessary and sufficient to drive exit from quiescence and entry into the cell cycle. Ablation of Msi activity in reserve ISCs rendered the epithelium unable to regenerate in response to injury that ablates the active stem cell compartment. These findings delineate a molecular mechanism governing reserve ISC quiescence and demonstrate a necessity for the activity of this rare stem cell population in intestinal regeneration.
Collapse
Affiliation(s)
- Maryam Yousefi
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Cell and Molecular Biology Graduate Program, University of Pennsylvania, Philadelphia, PA 19104
| | - Ning Li
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Angela Nakauka-Ddamba
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Shan Wang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100083, China
| | - Kimberly Davidow
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jenna Schoenberger
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100083, China
| | - Shane T Jensen
- Department of Biostatistics, the Wharton School, University of Pennsylvania, Philadelphia, PA 19104
| | - Michael G Kharas
- Molecular Pharmacology and Chemistry Program, Experimental Therapeutics Center and Center for Stem Cell Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Cell and Developmental Biology, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Cell and Molecular Biology Graduate Program, University of Pennsylvania, Philadelphia, PA 19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
550
|
Karatas OF, Suer I, Yuceturk B, Yilmaz M, Oz B, Guven G, Cansiz H, Creighton CJ, Ittmann M, Ozen M. Identification of microRNA profile specific to cancer stem-like cells directly isolated from human larynx cancer specimens. BMC Cancer 2016; 16:853. [PMID: 27816053 PMCID: PMC5097853 DOI: 10.1186/s12885-016-2863-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 10/04/2016] [Indexed: 12/14/2022] Open
Abstract
Background Emerging evidences proposed that microRNAs are associated with regulation of distinct physio-pathological processes including development of normal stem cells and carcinogenesis. In this study we aimed to investigate microRNA profile of cancer stem-like cells (CSLCs) isolated form freshly resected larynx cancer (LCa) tissue samples. Methods CD133 positive (CD133+) stem-like cells were isolated from freshly resected LCa tumor specimens. MicroRNA profile of 12 pair of CD133+ and CD133− cells was determined using microRNA microarray and differential expressions of selvected microRNAs were validated by quantitative real time PCR (qRT-PCR). Results MicroRNA profiling of CD133+ and CD133− LCa samples with microarray revealed that miR-26b, miR-203, miR-200c, and miR-363-3p were significantly downregulated and miR-1825 was upregulated in CD133+ larynx CSLCs. qRT-PCR analysis in a total of 25 CD133+/CD133− sample pairs confirmed the altered expressions of these five microRNAs. Expressions of miR-26b, miR-200c, and miR-203 were significantly correlated with miR-363-3p, miR-203, and miR-363-3p expressions, respectively. Furthermore, in silico analysis revealed that these microRNAs target both cancer and stem-cell associated signaling pathways. Conclusions Our results showed that certain microRNAs in CD133+ cells could be used as cancer stem cell markers. Based on these results, we propose that this panel of microRNAs might carry crucial roles in LCa pathogenesis through regulating stem cell properties of tumor cells. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2863-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Omer Faruk Karatas
- Molecular Biology and Genetics Department, Erzurum Technical University, Erzurum, Turkey
| | - Ilknur Suer
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
| | - Betul Yuceturk
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey.,Advanced Genomics and Bioinformatics Research Center, The Scientific and Technological Research Council of Turkey (TUBITAK), Gebze, Kocaeli, Turkey
| | - Mehmet Yilmaz
- Department of Otorhinolaryngology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - Buge Oz
- Department of Pathology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - Gulgun Guven
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
| | - Harun Cansiz
- Department of Otorhinolaryngology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - Chad J Creighton
- Department of Medicine and Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, USA
| | - Michael Ittmann
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA.,Michael E. DeBakey VAMC, Houston, TX, 77030, USA
| | - Mustafa Ozen
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey. .,Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|