51
|
Epigenetic modification of TLE1 induce abnormal differentiation in diabetic mice intestinal epithelium. Mol Cell Biochem 2017; 438:85-96. [PMID: 28744818 DOI: 10.1007/s11010-017-3116-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/15/2017] [Indexed: 12/20/2022]
Abstract
The intestinal epithelium cells (IECs) in diabetes mellitus (DM) patients have been proven to be abnormally differentiated. During the differentiation of IECs, epigenetic modification acts as an important regulator. In this study, we aimed to examine the epigenetic alteration of Transducin-like Enhancer of Split 1 (TLE1), a multitask transcriptional co-repressor, contributing to the differentiation homeostasis in IECs of DM mice. The IECs of type 2 diabetic mice model were isolated and collected. Methylation states of whole genomic DNA promoter regions were investigated by microarray. Methylated-specific PCR was used to detect the methylation state of TLE1 promoter in DM mice IECs. The expression of TLE1, Hes1, and differentiated cell markers were measured through real-time PCR, Western blots, and immunohistochemistry; by transfection assay, TLE1 or Hes1 was independently down-regulated in intestinal epithelium cell line, IEC-6. Subsequent modulation on TLE1, Hes1, and differentiated intestinal cell markers were detected. Global gene promoter regions in DM intestinal epithelium were less methylated comparing to normal control. The expression of TLE1 was significantly increased via hypomethylated activation in DM mice IECs. Hes1 was significantly suppressed and the terminal cell markers abnormally expressed in DM mice IECs (P < 0.05). Inhibition or induction on the abundance of TLE1 in IEC-6 cell line resulted in the corresponding dysregulation of Hes1 and intestinal epithelium differentiation (P < 0.05). Demethylation of TLE1 promoter region activates the self-expression in diabetic mice IECs. Subsequently, TLE1, through the transcriptional suppression on expression of Hes1, contributes to the aberrant differentiation of IECs in DM mice.
Collapse
|
52
|
Robinson SC, Klobucar K, Pierre CC, Ansari A, Zhenilo S, Prokhortchouk E, Daniel JM. Kaiso differentially regulates components of the Notch signaling pathway in intestinal cells. Cell Commun Signal 2017. [PMID: 28637464 PMCID: PMC5480165 DOI: 10.1186/s12964-017-0178-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background In mammalian intestines, Notch signaling plays a critical role in mediating cell fate decisions; it promotes the absorptive (or enterocyte) cell fate, while concomitantly inhibiting the secretory cell fate (i.e. goblet, Paneth and enteroendocrine cells). We recently reported that intestinal-specific Kaiso overexpressing mice (KaisoTg) exhibited chronic intestinal inflammation and had increased numbers of all three secretory cell types, hinting that Kaiso might regulate Notch signaling in the gut. However, Kaiso’s precise role in Notch signaling and whether the KaisoTg secretory cell fate phenotype was linked to Kaiso-induced inflammation had yet to be elucidated. Methods Intestines from 3-month old Non-transgenic and KaisoTg mice were “Swiss” rolled and analysed for the expression of Notch1, Dll-1, Jagged-1, and secretory cell markers by immunohistochemistry and immunofluorescence. To evaluate inflammation, morphological analyses and myeloperoxidase assays were performed on intestines from 3-month old KaisoTg and control mice. Notch1, Dll-1 and Jagged-1 expression were also assessed in stable Kaiso-depleted colon cancer cells and isolated intestinal epithelial cells using real time PCR and western blotting. To assess Kaiso binding to the DLL1, JAG1 and NOTCH1 promoter regions, chromatin immunoprecipitation was performed on three colon cancer cell lines. Results Here we demonstrate that Kaiso promotes secretory cell hyperplasia independently of Kaiso-induced inflammation. Moreover, Kaiso regulates several components of the Notch signaling pathway in intestinal cells, namely, Dll-1, Jagged-1 and Notch1. Notably, we found that in KaisoTg mice intestines, Notch1 and Dll-1 expression are significantly reduced while Jagged-1 expression is increased. Chromatin immunoprecipitation experiments revealed that Kaiso associates with the DLL1 and JAG1 promoter regions in a methylation-dependent manner in colon carcinoma cell lines, suggesting that these Notch ligands are putative Kaiso target genes. Conclusion Here, we provide evidence that Kaiso’s effects on intestinal secretory cell fates precede the development of intestinal inflammation in KaisoTg mice. We also demonstrate that Kaiso inhibits the expression of Dll-1, which likely contributes to the secretory cell phenotype observed in our transgenic mice. In contrast, Kaiso promotes Jagged-1 expression, which may have implications in Notch-mediated colon cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/s12964-017-0178-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shaiya C Robinson
- Department of Biology, McMaster University, Hamilton, L8S 4K1, ON, Canada
| | - Kristina Klobucar
- Department of Biology, McMaster University, Hamilton, L8S 4K1, ON, Canada.,Current address: Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, L8N 3Z5, ON, Canada
| | - Christina C Pierre
- Department of Biology, McMaster University, Hamilton, L8S 4K1, ON, Canada.,Current address: Department of Life Science, University of the West Indies at St. Augustine, St. Augustine, Trinidad and Tobago
| | - Amna Ansari
- Department of Biology, McMaster University, Hamilton, L8S 4K1, ON, Canada
| | - Svetlana Zhenilo
- Federal Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, Russian Federation, 117312
| | - Egor Prokhortchouk
- Federal Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, Russian Federation, 117312
| | - Juliet M Daniel
- Department of Biology, McMaster University, Hamilton, L8S 4K1, ON, Canada.
| |
Collapse
|
53
|
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]
|
54
|
Kay SK, Harrington HA, Shepherd S, Brennan K, Dale T, Osborne JM, Gavaghan DJ, Byrne HM. The role of the Hes1 crosstalk hub in Notch-Wnt interactions of the intestinal crypt. PLoS Comput Biol 2017; 13:e1005400. [PMID: 28245235 PMCID: PMC5363986 DOI: 10.1371/journal.pcbi.1005400] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 03/23/2017] [Accepted: 02/07/2017] [Indexed: 12/21/2022] Open
Abstract
The Notch pathway plays a vital role in determining whether cells in the intestinal epithelium adopt a secretory or an absorptive phenotype. Cell fate specification is coordinated via Notch's interaction with the canonical Wnt pathway. Here, we propose a new mathematical model of the Notch and Wnt pathways, in which the Hes1 promoter acts as a hub for pathway crosstalk. Computational simulations of the model can assist in understanding how healthy intestinal tissue is maintained, and predict the likely consequences of biochemical knockouts upon cell fate selection processes. Chemical reaction network theory (CRNT) is a powerful, generalised framework which assesses the capacity of our model for monostability or multistability, by analysing properties of the underlying network structure without recourse to specific parameter values or functional forms for reaction rates. CRNT highlights the role of β-catenin in stabilising the Notch pathway and damping oscillations, demonstrating that Wnt-mediated actions on the Hes1 promoter can induce dynamic transitions in the Notch system, from multistability to monostability. Time-dependent model simulations of cell pairs reveal the stabilising influence of Wnt upon the Notch pathway, in which β-catenin- and Dsh-mediated action on the Hes1 promoter are key in shaping the subcellular dynamics. Where Notch-mediated transcription of Hes1 dominates, there is Notch oscillation and maintenance of fate flexibility; Wnt-mediated transcription of Hes1 favours bistability akin to cell fate selection. Cells could therefore regulate the proportion of Wnt- and Notch-mediated control of the Hes1 promoter to coordinate the timing of cell fate selection as they migrate through the intestinal epithelium and are subject to reduced Wnt stimuli. Furthermore, mutant cells characterised by hyperstimulation of the Wnt pathway may, through coupling with Notch, invert cell fate in neighbouring healthy cells, enabling an aberrant cell to maintain its neighbours in mitotically active states.
Collapse
Affiliation(s)
- Sophie K. Kay
- Department of Computer Science, University of Oxford, Oxford, U.K.
| | | | - Sarah Shepherd
- School of Mathematical Sciences, University of Nottingham, Nottingham, U.K.
| | - Keith Brennan
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, U.K.
| | - Trevor Dale
- School of Biosciences, Cardiff University, Cardiff, U.K.
| | - James M. Osborne
- School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| | | | - Helen M. Byrne
- Department of Computer Science, University of Oxford, Oxford, U.K.
- Mathematical Institute, University of Oxford, Oxford, U.K.
| |
Collapse
|
55
|
Wang Y, Huang D, Chen KY, Cui M, Wang W, Huang X, Awadellah A, Li Q, Friedman A, Xin WW, Di Martino L, Cominelli F, Miron A, Chan R, Fox J, Xu Y, Shen X, Kalady MF, Markowitz S, Maillard I, Lowe JB, Xin W, Zhou L. Fucosylation Deficiency in Mice Leads to Colitis and Adenocarcinoma. Gastroenterology 2017; 152:193-205.e10. [PMID: 27639802 PMCID: PMC5164974 DOI: 10.1053/j.gastro.2016.09.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/29/2016] [Accepted: 09/07/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND & AIMS De novo synthesis of guanosine diphosphate (GDP)-fucose, a substrate for fucosylglycans, requires sequential reactions mediated by GDP-mannose 4,6-dehydratase (GMDS) and GDP-4-keto-6-deoxymannose 3,5-epimerase-4-reductase (FX or tissue specific transplantation antigen P35B [TSTA3]). GMDS deletions and mutations are found in 6%-13% of colorectal cancers; these mostly affect the ascending and transverse colon. We investigated whether a lack of fucosylation consequent to loss of GDP-fucose synthesis contributes to colon carcinogenesis. METHODS FX deficiency and GMDS deletion produce the same biochemical phenotype of GDP-fucose deficiency. We studied a mouse model of fucosylation deficiency (Fx-/- mice) and mice with the full-length Fx gene (controls). Mice were placed on standard chow or fucose-containing diet (equivalent to a control fucosylglycan phenotype). Colon tissues were collected and analyzed histologically or by enzyme-linked immunosorbent assays to measure cytokine levels; T cells also were collected and analyzed. Fecal samples were analyzed by 16s ribosomal RNA sequencing. Mucosal barrier function was measured by uptake of fluorescent dextran. We transplanted bone marrow cells from Fx-/- or control mice (Ly5.2) into irradiated 8-week-old Fx-/- or control mice (Ly5.1). We performed immunohistochemical analyses for expression of Notch and the hes family bHLH transcription factor (HES1) in colon tissues from mice and a panel of 60 human colorectal cancer specimens (27 left-sided, 33 right-sided). RESULTS Fx-/- mice developed colitis and serrated-like lesions. The intestinal pathology of Fx-/- mice was reversed by addition of fucose to the diet, which restored fucosylation via a salvage pathway. In the absence of fucosylation, dysplasia appeared and progressed to adenocarcinoma in up to 40% of mice, affecting mainly the right colon and cecum. Notch was not activated in Fx-/- mice fed standard chow, leading to decreased expression of its target Hes1. Fucosylation deficiency altered the composition of the fecal microbiota, reduced mucosal barrier function, and altered epithelial proliferation marked by Ki67. Fx-/- mice receiving control bone marrow cells had intestinal inflammation and dysplasia, and reduced expression of cytokines produced by cytotoxic T cells. Human sessile serrated adenomas and right-sided colorectal tumors with epigenetic loss of MutL homolog 1 (MLH1) had lost or had lower levels of HES1 than other colorectal tumor types or nontumor tissues. CONCLUSIONS In mice, fucosylation deficiency leads to colitis and adenocarcinoma, loss of Notch activation, and down-regulation of Hes1. HES1 loss correlates with the development of human right-sided colorectal tumors with epigenetic loss of MLH1. These findings indicate that carcinogenesis in a subset of colon cancer is consequent to a molecular mechanism driven by fucosylation deficiency and/or HES1-loss.
Collapse
Affiliation(s)
- Yiwei Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Dan Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Kai-Yuan Chen
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Min Cui
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Weihuan Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Xiaoran Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Amad Awadellah
- Department of Pathology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Qing Li
- Department of Pathology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Ann Friedman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - William W. Xin
- School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, 19104-6304, USA
| | - Luca Di Martino
- Department of Internal Medicine, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Fabio Cominelli
- Department of Internal Medicine, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Alex Miron
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ricky Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, OH 44106, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Mathew F. Kalady
- Department of Colorectal Surgery, Digestive Diseases Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Sanford Markowitz
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ivan Maillard
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - John B. Lowe
- Department of Pathology, Genentech Inc., San Francisco, CA, 94080 USA
| | - Wei Xin
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA,Department of Pathology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio; Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio.
| |
Collapse
|
56
|
Beumer J, Clevers H. Regulation and plasticity of intestinal stem cells during homeostasis and regeneration. Development 2016; 143:3639-3649. [PMID: 27802133 DOI: 10.1242/dev.133132] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The intestinal epithelium is the fastest renewing tissue in mammals and has a large flexibility to adapt to different types of damage. Lgr5+ crypt base columnar (CBC) cells act as stem cells during homeostasis and are essential during regeneration. Upon perturbation, the activity of CBCs is dynamically regulated to maintain homeostasis and multiple dedicated progenitor cell populations can reverse to the stem cell state upon damage, adding another layer of compensatory mechanisms to facilitate regeneration. Here, we review our current understanding of how intestinal stem and progenitor cells contribute to homeostasis and regeneration, and the different signaling pathways that regulate their behavior. Nutritional state and inflammation have been recently identified as upstream regulators of stem cell activity in the mammalian intestine, and we explore how these systemic signals can influence homeostasis and regeneration.
Collapse
Affiliation(s)
- Joep Beumer
- Hubrecht Institute for Developmental Biology and Stem Cell Research, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute for Developmental Biology and Stem Cell Research, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|
57
|
Oittinen M, Popp A, Kurppa K, Lindfors K, Mäki M, Kaikkonen MU, Viiri K. Polycomb Repressive Complex 2 Enacts Wnt Signaling in Intestinal Homeostasis and Contributes to the Instigation of Stemness in Diseases Entailing Epithelial Hyperplasia or Neoplasia. Stem Cells 2016; 35:445-457. [PMID: 27570105 DOI: 10.1002/stem.2479] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/11/2016] [Accepted: 07/29/2016] [Indexed: 12/15/2022]
Abstract
Canonical Wnt/β-catenin signaling regulates the homeostasis of intestinal epithelium by controlling the balance between intestinal stem cell self-renewal and differentiation but epigenetic mechanisms enacting the process are not known. We hypothesized that epigenetic regulator, Polycomb Repressive Complex-2 (PRC2), is involved in Wnt-mediated epithelial homeostasis on the crypt-villus axis and aberrancies therein are implicated both in celiac disease and in intestinal malignancies. We found that PRC2 establishes repressive crypt and villus specific trimethylation of histone H3 lysine 27 (H3K27me3) signature on genes responsible for, for example, nutrient transport and cell killing in crypts and, for example, proliferation and differentiation in mature villi, suggesting that PRC2 facilitates the Wnt-governed intestinal homeostasis. When celiac patients are on gluten-containing diet PRC2 is out-of-bounds active and consequently its target genes were found affected in intestinal epithelium. Significant set of effective intestinal PRC2 targets are also differentially expressed in colorectal adenoma and carcinomas. Our results suggest that PRC2 gives rise and maintains polar crypt and villus specific H3K27me3 signatures. As H3K27me3 is a mark enriched in developmentally important genes, identified intestinal PRC2 targets are possibly imperative drivers for enterocyte differentiation and intestinal stem cell maintenance downstream to Wnt-signaling. Our work also elucidates the mechanism sustaining the crypt hyperplasia in celiac disease and suggest that PRC2-dependent fostering of epithelial stemness is a common attribute in intestinal diseases in which epithelial hyperplasia or neoplasia prevails. Finally, this work demonstrates that in intestine PRC2 represses genes having both pro-stemness and pro-differentiation functions, fact need to be considered when designing epigenetic therapies including PRC2 as a drug target. Stem Cells 2017;35:445-457.
Collapse
Affiliation(s)
- Mikko Oittinen
- Tampere Centre for Child Health Research, University of Tampere, Department of Pediatrics and Tampere University Hospital, Tampere, Finland
| | - Alina Popp
- Tampere Centre for Child Health Research, University of Tampere, Department of Pediatrics and Tampere University Hospital, Tampere, Finland.,University of Medicine and Pharmacy "Carol Davila", Department of Pediatrics and Institute for Mother and Child Care, Bucharest, Romania
| | - Kalle Kurppa
- Tampere Centre for Child Health Research, University of Tampere, Department of Pediatrics and Tampere University Hospital, Tampere, Finland
| | - Katri Lindfors
- Tampere Centre for Child Health Research, University of Tampere, Department of Pediatrics and Tampere University Hospital, Tampere, Finland
| | - Markku Mäki
- Tampere Centre for Child Health Research, University of Tampere, Department of Pediatrics and Tampere University Hospital, Tampere, Finland
| | - Minna U Kaikkonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Keijo Viiri
- Tampere Centre for Child Health Research, University of Tampere, Department of Pediatrics and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
58
|
Chiacchiera F, Rossi A, Jammula S, Zanotti M, Pasini D. PRC2 preserves intestinal progenitors and restricts secretory lineage commitment. EMBO J 2016; 35:2301-2314. [PMID: 27585866 DOI: 10.15252/embj.201694550] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/10/2016] [Indexed: 11/09/2022] Open
Abstract
Chromatin modifications shape cell heterogeneity by activating and repressing defined sets of genes involved in cell proliferation, differentiation and development. Polycomb-repressive complexes (PRCs) act synergistically during development and differentiation by maintaining transcriptional repression of common genes. PRC2 exerts this activity by catalysing H3K27 trimethylation. Here, we show that in the intestinal epithelium PRC2 is required to sustain progenitor cell proliferation and the correct balance between secretory and absorptive lineage differentiation programs. Using genetic models, we show that PRC2 activity is largely dispensable for intestinal stem cell maintenance but is strictly required for radiation-induced regeneration by preventing Cdkn2a transcription. Combining these models with genomewide molecular analysis, we further demonstrate that preferential accumulation of secretory cells does not result from impaired proliferation of progenitor cells induced by Cdkn2a activation but rather from direct regulation of transcription factors responsible for secretory lineage commitment. Overall, our data uncover a dual role of PRC2 in intestinal homeostasis highlighting the importance of this repressive layer in controlling cell plasticity and lineage choices in adult tissues.
Collapse
Affiliation(s)
- Fulvio Chiacchiera
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Alessandra Rossi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - SriGanesh Jammula
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Marika Zanotti
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Diego Pasini
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
59
|
Reg4+ deep crypt secretory cells function as epithelial niche for Lgr5+ stem cells in colon. Proc Natl Acad Sci U S A 2016; 113:E5399-407. [PMID: 27573849 DOI: 10.1073/pnas.1607327113] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Leucine-rich repeat-containing G-protein coupled receptor 5-positive (Lgr5(+)) stem cells reside at crypt bottoms of the small and large intestine. Small intestinal Paneth cells supply Wnt3, EGF, and Notch signals to neighboring Lgr5(+) stem cells. Whereas the colon lacks Paneth cells, deep crypt secretory (DCS) cells are intermingled with Lgr5(+) stem cells at crypt bottoms. Here, we report regenerating islet-derived family member 4 (Reg4) as a marker of DCS cells. To investigate a niche function, we eliminated DCS cells by using the diphtheria-toxin receptor gene knocked into the murine Reg4 locus. Ablation of DCS cells results in loss of stem cells from colonic crypts and disrupts gut homeostasis and colon organoid growth. In agreement, sorted Reg4(+) DCS cells promote organoid formation of single Lgr5(+) colon stem cells. DCS cells can be massively produced from Lgr5(+) colon stem cells in vitro by combined Notch inhibition and Wnt activation. We conclude that Reg4(+) DCS cells serve as Paneth cell equivalents in the colon crypt niche.
Collapse
|
60
|
Takada Y, Fukuda A, Chiba T, Seno H. Brg1 plays an essential role in development and homeostasis of the duodenum through regulation of Notch signaling. Development 2016; 143:3532-3539. [PMID: 27510977 DOI: 10.1242/dev.141549] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 07/28/2016] [Indexed: 12/16/2022]
Abstract
Brg1, a core subunit of the SWI/SNF chromatin remodeling complex, is essential for development and homeostasis of various organs. However, the functional role of Brg1 in intestinal development and homeostasis, and the underlying molecular mechanism, remain unknown. We found that deletion of Brg1 in the mouse intestine resulted in growth impairment and early death associated with abnormal crypt-villous formation, skewed differentiation into secretory lineage cells, markedly increased apoptosis, and stem cell loss in the duodenum. Furthermore, we found that the Notch signaling pathway was dramatically downregulated in Brg1-deficient duodenum. Remarkably, overexpression of the Notch1 intercellular domain (ICD) partially reversed the prognosis of intestinal Brg1 mutant mice. Notch1 ICD overexpression rescued morphogenesis, prevented over-differentiation into secretory lineage cells, and restored apoptosis to normal levels in Brg1-deficient duodenum, although stem cell loss was not rescued. Our data demonstrate that Brg1 plays an essential role in development and homeostasis, including morphogenesis, stem cell differentiation and cell survival in the duodenum. Mechanistically, the rescue of the intestinal Brg1 mutant phenotype by overexpression of the Notch1 ICD indicates that Notch signaling is a key downstream target that mediates the effects of Brg1.
Collapse
Affiliation(s)
- Yutaka Takada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| |
Collapse
|
61
|
Ahadi M, Andrici J, Sioson L, Sheen A, Clarkson A, Gill AJ. Loss of Hes1 expression is associated with poor prognosis in colorectal adenocarcinoma. Hum Pathol 2016; 57:91-97. [PMID: 27476040 DOI: 10.1016/j.humpath.2016.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/01/2016] [Accepted: 07/06/2016] [Indexed: 01/08/2023]
Abstract
Alterations in the Notch signaling pathway play a role in colorectal cancer (CRC). Hes1, a Notch-induced transcription factor, has recently been reported to show decreased expression by immunohistochemistry in sessile serrated adenomas. Variable staining patterns have been reported in tubular adenomas, and existing data on Hes1 expression in CRC are limited and inconsistent. We therefore sought to investigate the expression of Hes1 by immunohistochemistry in a large and well-characterized cohort of CRC patients to determine clinicopathological associations and prognostic significance. Immunohistochemistry for Hes1 was performed on 2775 consecutive CRCs in tissue microarray format. Hes1 expression was classified into 3 categories: absent, 1302 cases (46.9%); cytoplasmic staining only with loss of nuclear staining, 1002 cases (36.1%); and nuclear with or without cytoplasmic staining, 471 cases (17%). In univariate analysis, loss of nuclear expression of HES1 was significantly associated with older age, female sex, right-sided location, mucinous or medullary histology, higher histological grade, microsatellite instability, BRAFV600E mutation, and larger tumor size. Strong and statistically significant associations with female sex, right-sided location, BRAFV600E mutation, microsatellite instability, and larger size remained in multivariate analysis. Patients with loss of nuclear expression of Hes1 had a significantly worse all-cause 5-year survival in both univariate (P = .002) and multivariate (P = .009) analysis. We conclude that loss of nuclear expression of Hes1 occurs in 83% of CRCs when studied in tissue microarray format and is associated with female sex, right-sided location, BRAFV600E mutation, microsatellite instability, larger tumor size, and significantly worse survival.
Collapse
Affiliation(s)
- Mahsa Ahadi
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065; Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
| | - Juliana Andrici
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065; Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065; Sydney Medical School, University of Sydney, NSW, Australia, 2006
| | - Loretta Sioson
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
| | - Amy Sheen
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065
| | - Adele Clarkson
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065; Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065; Sydney Medical School, University of Sydney, NSW, Australia, 2006
| | - Anthony J Gill
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia, 2065; Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, Australia, 2065; Sydney Medical School, University of Sydney, NSW, Australia, 2006.
| |
Collapse
|
62
|
Ratanasirintrawoot S, Israsena N. Stem Cells in the Intestine: Possible Roles in Pathogenesis of Irritable Bowel Syndrome. J Neurogastroenterol Motil 2016; 22:367-82. [PMID: 27184041 PMCID: PMC4930294 DOI: 10.5056/jnm16023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/08/2016] [Indexed: 12/13/2022] Open
Abstract
Irritable bowel syndrome is one of the most common functional gastrointestinal (GI) disorders that significantly impair quality of life in patients. Current available treatments are still not effective and the pathophysiology of this condition remains unclearly defined. Recently, research on intestinal stem cells has greatly advanced our understanding of various GI disorders. Alterations in conserved stem cell regulatory pathways such as Notch, Wnt, and bone morphogenic protein/TGF-β have been well documented in diseases such as inflammatory bowel diseases and cancer. Interaction between intestinal stem cells and various signals from their environment is important for the control of stem cell self-renewal, regulation of number and function of specific intestinal cell types, and maintenance of the mucosal barrier. Besides their roles in stem cell regulation, these signals are also known to have potent effects on immune cells, enteric nervous system and secretory cells in the gut, and may be responsible for various aspects of pathogenesis of functional GI disorders, including visceral hypersensitivity, altered gut motility and low grade gut inflammation. In this article, we briefly summarize the components of these signaling pathways, how they can be modified by extrinsic factors and novel treatments, and provide evidenced support of their roles in the inflammation processes. Furthermore, we propose how changes in these signals may contribute to the symptom development and pathogenesis of irritable bowel syndrome.
Collapse
Affiliation(s)
- Sutheera Ratanasirintrawoot
- Stem Cell and Cell Therapy Research Unit, Chulalongkorn University, Bangkok, Thailand.,Department of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nipan Israsena
- Stem Cell and Cell Therapy Research Unit, Chulalongkorn University, Bangkok, Thailand.,Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
63
|
Demitrack ES, Samuelson LC. Notch regulation of gastrointestinal stem cells. J Physiol 2016; 594:4791-803. [PMID: 26848053 DOI: 10.1113/jp271667] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/19/2016] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract epithelium is continuously replenished by actively cycling stem and progenitor cells. These cell compartments are regulated to balance proliferation and stem cell renewal with differentiation into the various mature cell types to maintain tissue homeostasis. In this topical review we focus on the role of the Notch signalling pathway to regulate GI stem cell function in adult small intestine and stomach. We first present the current view of stem and progenitor cell populations in these tissues and then summarize the studies that have established the Notch pathway as a key regulator of gastric and intestinal stem cell function. Notch signalling has been shown to be a niche factor required for maintenance of GI stem cells in both tissues. In addition, Notch has been described to regulate epithelial cell differentiation. Recent studies have revealed key similarities and differences in how Notch regulates stem cell function in the stomach compared to intestine. We summarize the literature regarding Notch regulation of GI stem cell proliferation and differentiation, highlighting tissue-specific functions to compare and contrast Notch in the stomach and intestine.
Collapse
Affiliation(s)
- Elise S Demitrack
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
64
|
Chronic rapamycin treatment on the nutrient utilization and metabolism of juvenile turbot (Psetta maxima). Sci Rep 2016; 6:28068. [PMID: 27305975 PMCID: PMC4910097 DOI: 10.1038/srep28068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/31/2016] [Indexed: 12/04/2022] Open
Abstract
High dietary protein inclusion is necessary in fish feeds and also represents a major cost in the aquaculture industry, which demands improved dietary conversion into body proteins in fish. In mammals, the target of rapamycin (TOR) is a key nutritionally responsive molecule governing postprandial anabolism. However, its physiological significance in teleosts has not been fully examined. In the present study, we examined the nutritional physiology of turbot after chronic rapamycin inhibition. Our results showed that a 6-week inhibition of TOR using dietary rapamycin inclusion (30 mg/kg diet) reduced growth performance and feed utilization. The rapamycin treatment inhibited TOR signaling and reduced expression of key enzymes in glycolysis, lipogenesis, cholesterol biosynthesis, while increasing the expression of enzymes involved in gluconeogenesis. Furthermore, rapamycin treatment increased intestinal goblet cell number in turbot, while the expressions of Notch and Hes1 were down regulated. It was possible that stimulated goblet cell differentiation by rapamycin was mediated through Notch-Hes1 pathway. Therefore, our results demonstrate the important role of TOR signaling in fish nutritional physiology.
Collapse
|
65
|
Abstract
Sessile serrated adenoma/polyp (SSA/p) is a precancerous lesion, and its differential diagnosis from hyperplastic polyp (HP) could be challenging in certain circumstances based on morphology alone. Hes1 is a downstream target of Notch-signaling pathway and plays an important role in intestinal development by regulating differentiation of enterocytes. In this study, we evaluated the expression patterns of Hes1 in SSA/p and HP, and determine whether Hes1 immunostaining can help differentiate between these 2 entities. Serrated polyps with cytologic dysplasia (SSA with cytologic dysplasia, tubular adenoma, and traditional serrated adenoma) were also studied. Hes1 is ubiquitously expressed in the nuclei of normal colon epithelial cells. The complete loss or a very weak expression of Hes1 is observed in the majority of the SSA/p in the study (58/63, 92%) compared with the normal expression of Hes1 in HP (35/35,100%). In SSA/p with cytologic dysplasia, dysplastic area demonstrated cytoplasmic and/or nuclear staining for Hes1. Tubular adenoma and traditional serrated adenoma showed variability of Hes1 staining within the polyp with a mixed positive and negative staining pattern. Our study suggests that loss of Hes1 could be used as a sensitive and specific marker to differentiate SSA/p from HP, which helps the diagnosis in morphologically challenging cases.
Collapse
|
66
|
Weng MT, Tsao PN, Lin HL, Tung CC, Change MC, Chang YT, Wong JM, Wei SC. Hes1 Increases the Invasion Ability of Colorectal Cancer Cells via the STAT3-MMP14 Pathway. PLoS One 2015; 10:e0144322. [PMID: 26650241 PMCID: PMC4674118 DOI: 10.1371/journal.pone.0144322] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/15/2015] [Indexed: 12/21/2022] Open
Abstract
The Notch pathway contributes to self-renewal of tumor-initiating cell and inhibition of normal colonic epithelial cell differentiation. Deregulated expression of Notch1 and Jagged1 is observed in colorectal cancer. Hairy/enhancer of split (HES) family, the most characterized targets of Notch, involved in the development of many cancers. In this study, we explored the role of Hes1 in the tumorigenesis of colorectal cancer. Knocking down Hes1 induced CRC cell senescence and decreased the invasion ability, whereas over-expression of Hes1 increased STAT3 phosphorylation activity and up-regulated MMP14 protein level. We further explored the expression of Hes1 in human colorectal cancer and found high Hes1 mRNA expression is associated with poor prognosis in CRC patients. These findings suggest that Hes1 regulates the invasion ability through the STAT3-MMP14 pathway in CRC cells and high Hes1 expression is a predictor of poor prognosis of CRC.
Collapse
Affiliation(s)
- MT Weng
- Department of Internal Medicine, Far-Eastern Memorial Hospital, New Taipei, Taiwan
- Department of Chemical Engineering & Materials Science, Yuan-Ze University, Taoyuan, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - PN Tsao
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - HL Lin
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - CC Tung
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - MC Change
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - YT Chang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - JM Wong
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - SC Wei
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
67
|
Affiliation(s)
- Shoichi Date
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 108-8345, Japan;
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 108-8345, Japan;
| |
Collapse
|
68
|
Wang SC, Lin XL, Wang HY, Qin YJ, Chen L, Li J, Jia JS, Shen HF, Yang S, Xie RY, Wei F, Gao F, Rong XX, Yang J, Zhao WT, Zhang TT, Shi JW, Yao KT, Luo WR, Sun Y, Xiao D. Hes1 triggers epithelial-mesenchymal transition (EMT)-like cellular marker alterations and promotes invasion and metastasis of nasopharyngeal carcinoma by activating the PTEN/AKT pathway. Oncotarget 2015; 6:36713-36730. [PMID: 26452025 PMCID: PMC4742206 DOI: 10.18632/oncotarget.5457] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 09/21/2015] [Indexed: 02/07/2023] Open
Abstract
Overexpression of the transcriptional factor Hes1 (hairy and enhancer of split-1) has been observed in numerous cancers, but the precise roles of Hes1 in epithelial-mesenchymal transition (EMT), cancer invasion and metastasis remain unknown. Our current study firstly revealed that Hes1 upregulation in a cohort of human nasopharyngeal carcinoma (NPC) biopsies is significantly associated with the EMT, invasive and metastatic phenotypes of cancer. In the present study, we found that Hes1 overexpression triggered EMT-like cellular marker alterations of NPC cells, whereas knockdown of Hes1 through shRNA reversed the EMT-like phenotypes, as strongly supported by Hes1-mediated EMT in NPC clinical specimens described above. Gain-of-function and loss-of-function experiments demonstrated that Hes1 promoted the migration and invasion of NPC cells in vitro. In addition, exogenous expression of Hes1 significantly enhanced the metastatic ability of NPC cells in vivo. Chromatin immunoprecipitation (ChIP) assays showed that Hes1 inhibited PTEN expression in NPC cells through binding to PTEN promoter region. Increased Hes1 expression and decreased PTEN expression were also observed in a cohort of NPC biopsies. Additional studies demonstrated that Hes1-induced EMT-like molecular changes and increased motility and invasion of NPC cells were mediated by PTEN. Taken together, our results suggest, for what we believe is the first time, that Hes1 plays an important role in the invasion and metastasis of NPC through inhibiting PTEN expression to trigger EMT-like phenotypes.
Collapse
Affiliation(s)
- Sheng-Chun Wang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
- Department of Pathology, Guangdong Medical University, Dongguan 523808, China
| | - Xiao-Lin Lin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Hui-Yan Wang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yu-Juan Qin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Lin Chen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jing Li
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jun-Shuang Jia
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Hong-Fen Shen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Sheng Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Rao-Ying Xie
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Fang Wei
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Fei Gao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xiao-Xiang Rong
- Department of Oncology, Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Jie Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wen-Tao Zhao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Ting-Ting Zhang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jun-Wen Shi
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Kai-Tai Yao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wei-Ren Luo
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yan Sun
- Joint Program in Transfusion Medicine, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Dong Xiao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
69
|
Wang SC, Lin XL, Wang HY, Qin YJ, Chen L, Li J, Jia JS, Shen HF, Yang S, Xie RY, Wei F, Gao F, Rong XX, Yang J, Zhao WT, Zhang TT, Shi JW, Yao KT, Luo WR, Sun Y, Xiao D. Hes1 triggers epithelial-mesenchymal transition (EMT)-like cellular marker alterations and promotes invasion and metastasis of nasopharyngeal carcinoma by activating the PTEN/AKT pathway. Oncotarget 2015. [PMID: 26452025 DOI: hes1 triggers epithelial-mesenchymal transition (emt)-like cellular marker alterations and promotes invasion and metastasis of nasopharyngeal carcinoma by activating the pten/akt pathway] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Overexpression of the transcriptional factor Hes1 (hairy and enhancer of split-1) has been observed in numerous cancers, but the precise roles of Hes1 in epithelial-mesenchymal transition (EMT), cancer invasion and metastasis remain unknown. Our current study firstly revealed that Hes1 upregulation in a cohort of human nasopharyngeal carcinoma (NPC) biopsies is significantly associated with the EMT, invasive and metastatic phenotypes of cancer. In the present study, we found that Hes1 overexpression triggered EMT-like cellular marker alterations of NPC cells, whereas knockdown of Hes1 through shRNA reversed the EMT-like phenotypes, as strongly supported by Hes1-mediated EMT in NPC clinical specimens described above. Gain-of-function and loss-of-function experiments demonstrated that Hes1 promoted the migration and invasion of NPC cells in vitro. In addition, exogenous expression of Hes1 significantly enhanced the metastatic ability of NPC cells in vivo. Chromatin immunoprecipitation (ChIP) assays showed that Hes1 inhibited PTEN expression in NPC cells through binding to PTEN promoter region. Increased Hes1 expression and decreased PTEN expression were also observed in a cohort of NPC biopsies. Additional studies demonstrated that Hes1-induced EMT-like molecular changes and increased motility and invasion of NPC cells were mediated by PTEN. Taken together, our results suggest, for what we believe is the first time, that Hes1 plays an important role in the invasion and metastasis of NPC through inhibiting PTEN expression to trigger EMT-like phenotypes.
Collapse
Affiliation(s)
- Sheng-Chun Wang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, Guangdong Medical University, Dongguan 523808, China
| | - Xiao-Lin Lin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Hui-Yan Wang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yu-Juan Qin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Lin Chen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jing Li
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jun-Shuang Jia
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Hong-Fen Shen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Sheng Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Rao-Ying Xie
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Fang Wei
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Fei Gao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xiao-Xiang Rong
- Department of Oncology, Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Jie Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wen-Tao Zhao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Ting-Ting Zhang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jun-Wen Shi
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Kai-Tai Yao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wei-Ren Luo
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yan Sun
- Joint Program in Transfusion Medicine, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Dong Xiao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
70
|
Wang SC, Lin XL, Wang HY, Qin YJ, Chen L, Li J, Jia JS, Shen HF, Yang S, Xie RY, Wei F, Gao F, Rong XX, Yang J, Zhao WT, Zhang TT, Shi JW, Yao KT, Luo WR, Sun Y, Xiao D. Hes1 triggers epithelial-mesenchymal transition (EMT)-like cellular marker alterations and promotes invasion and metastasis of nasopharyngeal carcinoma by activating the PTEN/AKT pathway. Oncotarget 2015. [PMID: 26452025 DOI: hes1 triggers epithelial-mesenchymal transition (emt)-like cellular marker alterations and promotes invasion and metastasis of nasopharyngeal carcinoma by activating the pten/akt pathway] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Overexpression of the transcriptional factor Hes1 (hairy and enhancer of split-1) has been observed in numerous cancers, but the precise roles of Hes1 in epithelial-mesenchymal transition (EMT), cancer invasion and metastasis remain unknown. Our current study firstly revealed that Hes1 upregulation in a cohort of human nasopharyngeal carcinoma (NPC) biopsies is significantly associated with the EMT, invasive and metastatic phenotypes of cancer. In the present study, we found that Hes1 overexpression triggered EMT-like cellular marker alterations of NPC cells, whereas knockdown of Hes1 through shRNA reversed the EMT-like phenotypes, as strongly supported by Hes1-mediated EMT in NPC clinical specimens described above. Gain-of-function and loss-of-function experiments demonstrated that Hes1 promoted the migration and invasion of NPC cells in vitro. In addition, exogenous expression of Hes1 significantly enhanced the metastatic ability of NPC cells in vivo. Chromatin immunoprecipitation (ChIP) assays showed that Hes1 inhibited PTEN expression in NPC cells through binding to PTEN promoter region. Increased Hes1 expression and decreased PTEN expression were also observed in a cohort of NPC biopsies. Additional studies demonstrated that Hes1-induced EMT-like molecular changes and increased motility and invasion of NPC cells were mediated by PTEN. Taken together, our results suggest, for what we believe is the first time, that Hes1 plays an important role in the invasion and metastasis of NPC through inhibiting PTEN expression to trigger EMT-like phenotypes.
Collapse
Affiliation(s)
- Sheng-Chun Wang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, Guangdong Medical University, Dongguan 523808, China
| | - Xiao-Lin Lin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Hui-Yan Wang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yu-Juan Qin
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Lin Chen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jing Li
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jun-Shuang Jia
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Hong-Fen Shen
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Sheng Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Rao-Ying Xie
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Fang Wei
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Fei Gao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xiao-Xiang Rong
- Department of Oncology, Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, Guangdong 510315, China
| | - Jie Yang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wen-Tao Zhao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Ting-Ting Zhang
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Jun-Wen Shi
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Kai-Tai Yao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Wei-Ren Luo
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China
| | - Yan Sun
- Joint Program in Transfusion Medicine, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Dong Xiao
- Cancer Research Institute, Southern Medical University, Guangzhou 510515, China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
71
|
Huang CZ, Yu T, Chen QK. DNA Methylation Dynamics During Differentiation, Proliferation, and Tumorigenesis in the Intestinal Tract. Stem Cells Dev 2015; 24:2733-9. [PMID: 26413818 DOI: 10.1089/scd.2015.0235] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
DNA methylation, an epigenetic control mechanism in mammals, is widely present in the intestinal tract during the differentiation and proliferation of epithelial cells. Cells in stem cell pools or villi have different patterns of DNA methylation. The process of DNA methylation is dynamic and occurs at many relevant regulatory elements during the rapid transition of stem cells into fully mature, differentiated epithelial cells. Changes in DNA methylation patterns most often take place in enhancer and promoter regions and are associated with transcription factor binding. During differentiation, enhancer regions associated with genes important to enterocyte differentiation are demethylated, activating gene expression. Abnormal patterns of DNA methylation during differentiation and proliferation in the intestinal tract can lead to the formation of aberrant crypt foci and destroy the barrier and absorptive functions of the intestinal epithelium. Accumulation of these epigenetic changes may even result in tumorigenesis. In the current review, we discuss recent findings on the association between DNA methylation and cell differentiation and proliferation in the small intestine and highlight the possible links between dysregulation of this process and tumorigenesis.
Collapse
Affiliation(s)
- Can-Ze Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, Guangdong, People's Republic of China
| | - Tao Yu
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, Guangdong, People's Republic of China
| | - Qi-Kui Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
72
|
Mirone G, Perna S, Shukla A, Marfe G. Involvement of Notch-1 in Resistance to Regorafenib in Colon Cancer Cells. J Cell Physiol 2015; 231:1097-105. [PMID: 26419617 DOI: 10.1002/jcp.25206] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/28/2015] [Indexed: 12/17/2022]
Abstract
Regorafenib, an oral small-molecule multi kinase inhibitor, is able to block Vascular Endothelial Growth Factor Receptors (VEGFR-1, 2, and 3), Platelet-Derived Growth Factor Receptors (PDGF), Fibroblast Growth Factor (FGF) receptor 1, Raf, TIE-2, and the kinases KIT, RET, and BRAF. Different studies have displayed its antitumor activity in several cancer models (both in vitro and in vivo), particularly in colorectal and gastrointestinal stromal cancers. The mechanism of resistance to regorafenib is largely unknown. In our investigation, we have generated regorafenib-resistant SW480 cells (Reg-R-SW480 cells) by culturing such cells with increasing concentration of regorafenib. Examination of intracellular signaling found that Akt signaling was activated in Reg-R-SW480 cells but not in wild-type SW480 cells, after regorafenib treatment as measured by Western Blot. The Notch pathway is a fundamental signaling system in the development and homeostasis of tissues since it regulates different cellular process such as proliferation, differentiation, and apoptosis and it can be a potential driver of resistance to a wide array of targeted therapies. In this study, we found that Notch-1 was significantly up-regulated in resistant tumor cells as well as HES1 and HEY. Additionally, inhibition of Notch-1 in resistant cells partially restored sensitivity to regorafenib treatment in vitro. Collectively, these data suggest a key role of Notch-1 in mediating the resistant effects of regorafenib in colorectal cancer cells, and also provide a rationale to improve the therapeutic efficacy of regorafenib.
Collapse
Affiliation(s)
- Giovanna Mirone
- Department of Medical Oncology B, Regina Elena National Cancer Institute, via Elio Chianesi 53, Rome, Italy
| | - Stefania Perna
- Department of Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Second University of Naples, via Vivaldi 43, Caserta, Italy
| | - Arvind Shukla
- School of Biotechnology and Bioinformatics, D.Y. Patil University, Plot No. 50, Sector-15, C.B.D. Belapur, Navi Mumbai, Maharastra, India
| | - Gabriella Marfe
- Department of Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Second University of Naples, via Vivaldi 43, Caserta, Italy
| |
Collapse
|
73
|
Shi Y, Shu B, Yang R, Xu Y, Xing B, Liu J, Chen L, Qi S, Liu X, Wang P, Tang J, Xie J. Wnt and Notch signaling pathway involved in wound healing by targeting c-Myc and Hes1 separately. Stem Cell Res Ther 2015; 6:120. [PMID: 26076648 PMCID: PMC4501079 DOI: 10.1186/s13287-015-0103-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 01/20/2015] [Accepted: 05/20/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction Wnt and Notch signaling pathways are critically involved in relative cell fate decisions within the development of cutaneous tissues. Moreover, several studies identified the above two pathways as having a significant role during wound healing. However, their biological effects during cutaneous tissues repair are unclear. Methods We employed a self-controlled model (Sprague–Dawley rats with full-thickness skin wounds) to observe the action and effect of Wnt/β-catenin and Notch signalings in vivo. The quality of wound repair relevant to the gain/loss-of-function Wnt/β-catenin and Notch activation was estimated by hematoxylin-and-eosin and Masson staining. Immunofluorescence analysis and Western blot analysis were used to elucidate the underlying mechanism of the regulation of Wnt and Notch signaling pathways in wound healing. Meanwhile, epidermal stem cells (ESCs) were cultured in keratinocyte serum-free medium with Jaggedl or in DAPT (N-[(3,5-difluorophenyl)acetyl]-L-alanyl-2-phenyl]glycine-1,1-dimethylethyl) to investigate whether the interruption of Notch signaling contributes to the expression of Wnt/β-catenin signaling. Results The results showed that in vivo the gain-of-function Wnt/β-catenin and Notch activation extended the ability to promote wound closure. We further determined that activation or inhibition of Wnt signaling and Notch signaling can affect the proliferation of ESCs, the differentiation and migration of keratinocytes, and follicle regeneration by targeting c-Myc and Hes1, which ultimately lead to enhanced or delayed wound healing. Furthermore, Western blot analysis suggested that the two pathways might interact in vivo and in vitro. Conclusion These results suggest that Wnt and Notch signalings play important roles in cutaneous repair by targeting c-Myc and Hes1 separately. What’s more, interaction between the above two pathways might act as a vital role in regulation of wound healing.
Collapse
Affiliation(s)
- Yan Shi
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Bin Shu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Ronghua Yang
- Department of Burns, Fo Shan Hospital of Sun Yat-Sen University, Lingnan avenue, Foshan, 528000, China.
| | - Yingbin Xu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Bangrong Xing
- Department of Burns, Third Affiliated Hospital of Sun Yat-sen University, Tianhe road, Guangzhou, 510620, China.
| | - Jian Liu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Lei Chen
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Shaohai Qi
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Xusheng Liu
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Peng Wang
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Jinming Tang
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| | - Julin Xie
- Department of Burns Surgery, First Affiliated Hospital of Sun Yat-Sen University, Zhongshan road, Guangzhou, 510080, China.
| |
Collapse
|
74
|
Qi Z, Chen YG. Regulation of intestinal stem cell fate specification. SCIENCE CHINA-LIFE SCIENCES 2015; 58:570-8. [PMID: 25951932 DOI: 10.1007/s11427-015-4859-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 09/17/2014] [Indexed: 02/07/2023]
Abstract
The remarkable ability of rapid self-renewal makes the intestinal epithelium an ideal model for the study of adult stem cells. The intestinal epithelium is organized into villus and crypt, and a group of intestinal stem cells located at the base of crypt are responsible for this constant self-renewal throughout the life. Identification of the intestinal stem cell marker Lgr5, isolation and in vitro culture of Lgr5+ intestinal stem cells and the use of transgenic mouse models have significantly facilitated the studies of intestinal stem cell homeostasis and differentiation, therefore greatly expanding our knowledge of the regulatory mechanisms underlying the intestinal stem cell fate determination. In this review, we summarize the current understanding of how signals of Wnt, BMP, Notch and EGF in the stem cell niche modulate the intestinal stem cell fate.
Collapse
Affiliation(s)
- Zhen Qi
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | | |
Collapse
|
75
|
Sancho R, Cremona CA, Behrens A. Stem cell and progenitor fate in the mammalian intestine: Notch and lateral inhibition in homeostasis and disease. EMBO Rep 2015; 16:571-81. [PMID: 25855643 PMCID: PMC4428041 DOI: 10.15252/embr.201540188] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/10/2015] [Accepted: 03/11/2015] [Indexed: 01/17/2023] Open
Abstract
The control of cell fate decisions is vital to build functional organs and maintain normal tissue homeostasis, and many pathways and processes cooperate to direct cells to an appropriate final identity. Because of its continuously renewing state and its carefully organised hierarchy, the mammalian intestine has become a powerful model to dissect these pathways in health and disease. One of the signalling pathways that is key to maintaining the balance between proliferation and differentiation in the intestinal epithelium is the Notch pathway, most famous for specifying distinct cell fates in adjacent cells via the evolutionarily conserved process of lateral inhibition. Here, we will review recent discoveries that advance our understanding of how cell fate in the mammalian intestine is decided by Notch and lateral inhibition, focusing on the molecular determinants that regulate protein turnover, transcriptional control and epigenetic regulation.
Collapse
Affiliation(s)
- Rocio Sancho
- Mammalian Genetics Laboratory, Cancer Research UK London Research Institute, Lincoln's Inn Fields Laboratories, London, UK
| | - Catherine A Cremona
- Mammalian Genetics Laboratory, Cancer Research UK London Research Institute, Lincoln's Inn Fields Laboratories, London, UK
| | - Axel Behrens
- Mammalian Genetics Laboratory, Cancer Research UK London Research Institute, Lincoln's Inn Fields Laboratories, London, UK School of Medicine, King's College London, London, UK
| |
Collapse
|
76
|
Kobayashi T, Iwamoto Y, Takashima K, Isomura A, Kosodo Y, Kawakami K, Nishioka T, Kaibuchi K, Kageyama R. Deubiquitinating enzymes regulate Hes1 stability and neuronal differentiation. FEBS J 2015; 282:2411-23. [PMID: 25846153 DOI: 10.1111/febs.13290] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/03/2015] [Accepted: 03/30/2015] [Indexed: 11/30/2022]
Abstract
Hairy and enhancer of split 1 (Hes1), a basic helix-loop-helix transcriptional repressor protein, regulates the maintenance of neural stem/progenitor cells by repressing proneural gene expression via Notch signaling. Previous studies showed that Hes1 expression oscillates in both mouse embryonic stem cells and neural stem cells, and that the oscillation contributes to their potency and differentiation fates. This oscillatory expression depends on the stability of Hes1, which is rapidly degraded by the ubiquitin/proteasome pathway. However, the detailed molecular mechanisms governing Hes1 stability remain unknown. We analyzed Hes1-interacting deubiquitinases purified from mouse embryonic stem cells using an Hes1-specific antibody, and identified the ubiquitin-specific protease 27x (Usp27x) as a new regulator of Hes1. We found that Hes1 was deubiquitinated and stabilized by Usp27x and its homologs ubiquitin-specific protease 22 (Usp22) and ubiquitin-specific protease 51 (Usp51). Knockdown of Usp22 shortened the half-life of Hes1, delayed its oscillation, and enhanced neuronal differentiation in mouse developing brain, whereas mis-expression of Usp27x reduced neuronal differentiation. These results suggest that these deubiquitinases modulate Hes1 protein dynamics by removing ubiquitin molecules, and thereby regulate neuronal differentiation of stem cells.
Collapse
Affiliation(s)
- Taeko Kobayashi
- Institute for Virus Research, Kyoto University, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan.,Graduate School of Medicine, Kyoto University, Japan.,Graduate School of Biostudies, Kyoto University, Japan
| | | | | | - Akihiro Isomura
- Institute for Virus Research, Kyoto University, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan
| | - Yoichi Kosodo
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Shizuoka, Japan
| | - Tomoki Nishioka
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, Japan
| | - Ryoichiro Kageyama
- Institute for Virus Research, Kyoto University, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan.,Graduate School of Medicine, Kyoto University, Japan.,Graduate School of Biostudies, Kyoto University, Japan.,World Premier International Research Initiative/Institute for Integrated Cell and Material Sciences, Kyoto University, Japan
| |
Collapse
|
77
|
Horvay K, Jardé T, Casagranda F, Perreau VM, Haigh K, Nefzger CM, Akhtar R, Gridley T, Berx G, Haigh JJ, Barker N, Polo JM, Hime GR, Abud HE. Snai1 regulates cell lineage allocation and stem cell maintenance in the mouse intestinal epithelium. EMBO J 2015; 34:1319-35. [PMID: 25759216 DOI: 10.15252/embj.201490881] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/02/2015] [Indexed: 12/17/2022] Open
Abstract
Snail family members regulate epithelial-to-mesenchymal transition (EMT) during invasion of intestinal tumours, but their role in normal intestinal homeostasis is unknown. Studies in breast and skin epithelia indicate that Snail proteins promote an undifferentiated state. Here, we demonstrate that conditional knockout of Snai1 in the intestinal epithelium results in apoptotic loss of crypt base columnar stem cells and bias towards differentiation of secretory lineages. In vitro organoid cultures derived from Snai1 conditional knockout mice also undergo apoptosis when Snai1 is deleted. Conversely, ectopic expression of Snai1 in the intestinal epithelium in vivo results in the expansion of the crypt base columnar cell pool and a decrease in secretory enteroendocrine and Paneth cells. Following conditional deletion of Snai1, the intestinal epithelium fails to produce a proliferative response following radiation-induced damage indicating a fundamental requirement for Snai1 in epithelial regeneration. These results demonstrate that Snai1 is required for regulation of lineage choice, maintenance of CBC stem cells and regeneration of the intestinal epithelium following damage.
Collapse
Affiliation(s)
- Katja Horvay
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| | - Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| | - Franca Casagranda
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Victoria M Perreau
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Katharina Haigh
- Australian Centre for Blood Diseases, Monash University & Alfred Health, Melbourne, Vic., Australia Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christian M Nefzger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia Australian Regenerative Medicine Institute, Monash University, Clayton, Vic., Australia
| | - Reyhan Akhtar
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| | - Thomas Gridley
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium Molecular and Cellular Oncology, Inflammation Research Center, VIB, Ghent, Belgium
| | - Jody J Haigh
- Australian Centre for Blood Diseases, Monash University & Alfred Health, Melbourne, Vic., Australia Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Nick Barker
- A*STAR Institute of Medical Biology, Singapore City, Singapore
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia Australian Regenerative Medicine Institute, Monash University, Clayton, Vic., Australia
| | - Gary R Hime
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia
| |
Collapse
|
78
|
Zhou Y, Rychahou P, Wang Q, Weiss HL, Evers BM. TSC2/mTORC1 signaling controls Paneth and goblet cell differentiation in the intestinal epithelium. Cell Death Dis 2015; 6:e1631. [PMID: 25654764 PMCID: PMC4669793 DOI: 10.1038/cddis.2014.588] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 12/02/2014] [Accepted: 12/05/2014] [Indexed: 12/18/2022]
Abstract
The intestinal mucosa undergoes a continual process of proliferation, differentiation and apoptosis, which is regulated by multiple signaling pathways. Notch signaling is critical for the control of intestinal stem cell maintenance and differentiation. However, the precise mechanisms involved in the regulation of differentiation are not fully understood. Previously, we have shown that tuberous sclerosis 2 (TSC2) positively regulates the expression of the goblet cell differentiation marker, MUC2, in intestinal cells. Using transgenic mice constitutively expressing a dominant negative TSC2 allele, we observed that TSC2 inactivation increased mTORC1 and Notch activities, and altered differentiation throughout the intestinal epithelium, with a marked decrease in the goblet and Paneth cell lineages. Conversely, treatment of mice with either Notch inhibitor dibenzazepine (DBZ) or mTORC1 inhibitor rapamycin significantly attenuated the reduction of goblet and Paneth cells. Accordingly, knockdown of TSC2 activated, whereas knockdown of mTOR or treatment with rapamycin decreased, the activity of Notch signaling in the intestinal cell line LS174T. Importantly, our findings demonstrate that TSC2/mTORC1 signaling contributes to the maintenance of intestinal epithelium homeostasis by regulating Notch activity.
Collapse
Affiliation(s)
- Y Zhou
- Markey Cancer Center, The University of Kentucky, Lexington, KY, USA
| | - P Rychahou
- 1] Markey Cancer Center, The University of Kentucky, Lexington, KY, USA [2] Department of Surgery, The University of Kentucky, Lexington, KY, USA
| | - Q Wang
- 1] Markey Cancer Center, The University of Kentucky, Lexington, KY, USA [2] Department of Surgery, The University of Kentucky, Lexington, KY, USA
| | - H L Weiss
- Markey Cancer Center, The University of Kentucky, Lexington, KY, USA
| | - B M Evers
- 1] Markey Cancer Center, The University of Kentucky, Lexington, KY, USA [2] Department of Surgery, The University of Kentucky, Lexington, KY, USA
| |
Collapse
|
79
|
Masjkur J, Arps-Forker C, Poser SW, Nikolakopoulou P, Toutouna L, Chenna R, Chavakis T, Chatzigeorgiou A, Chen LS, Dubrovska A, Choudhary P, Uphues I, Mark M, Bornstein SR, Androutsellis-Theotokis A. Hes3 is expressed in the adult pancreatic islet and regulates gene expression, cell growth, and insulin release. J Biol Chem 2014; 289:35503-35516. [PMID: 25371201 PMCID: PMC4271235 DOI: 10.1074/jbc.m114.590687] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 10/31/2014] [Indexed: 12/16/2022] Open
Abstract
The transcription factor Hes3 is a component of a signaling pathway that supports the growth of neural stem cells with profound consequences in neurodegenerative disease models. Here we explored whether Hes3 also regulates pancreatic islet cells. We showed that Hes3 is expressed in human and rodent pancreatic islets. In mouse islets it co-localizes with alpha and beta cell markers. We employed the mouse insulinoma cell line MIN6 to perform in vitro characterization and functional studies in conditions known to modulate Hes3 based upon our previous work using neural stem cell cultures. In these conditions, cells showed elevated Hes3 expression and nuclear localization, grew efficiently, and showed higher evoked insulin release responses, compared with serum-containing conditions. They also exhibited higher expression of the transcription factor Pdx1 and insulin. Furthermore, they were responsive to pharmacological treatments with the GLP-1 analog Exendin-4, which increased nuclear Hes3 localization. We employed a transfection approach to address specific functions of Hes3. Hes3 RNA interference opposed cell growth and affected gene expression as revealed by DNA microarrays. Western blotting and PCR approaches specifically showed that Hes3 RNA interference opposes the expression of Pdx1 and insulin. Hes3 overexpression (using a Hes3-GFP fusion construct) confirmed a role of Hes3 in regulating Pdx1 expression. Hes3 RNA interference reduced evoked insulin release. Mice lacking Hes3 exhibited increased islet damage by streptozotocin. These data suggest roles of Hes3 in pancreatic islet function.
Collapse
Affiliation(s)
| | | | | | | | | | - Ramu Chenna
- the Applied Bioinformatics Group, BioInnovations Zentrum, University of Dresden, 01307 Dresden, Germany
| | - Triantafyllos Chavakis
- the Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and
| | - Antonios Chatzigeorgiou
- the Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and
| | - Lan-Sun Chen
- the Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine and
| | - Anna Dubrovska
- Department of Medicine, OncoRay National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Pratik Choudhary
- the Diabetes Research Group, King's College London, London SE5 9RS, United Kingdom
| | - Ingo Uphues
- the Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach, Germany, and
| | - Michael Mark
- the Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach, Germany, and
| | | | | |
Collapse
|
80
|
López-Arribillaga E, Rodilla V, Pellegrinet L, Guiu J, Iglesias M, Roman AC, Gutarra S, González S, Muñoz-Cánoves P, Fernández-Salguero P, Radtke F, Bigas A, Espinosa L. Bmi1 regulates murine intestinal stem cell proliferation and self-renewal downstream of Notch. Development 2014; 142:41-50. [PMID: 25480918 DOI: 10.1242/dev.107714] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Genetic data indicate that abrogation of Notch-Rbpj or Wnt-β-catenin pathways results in the loss of the intestinal stem cells (ISCs). However, whether the effect of Notch is direct or due to the aberrant differentiation of the transit-amplifying cells into post-mitotic goblet cells is unknown. To address this issue, we have generated composite tamoxifen-inducible intestine-specific genetic mouse models and analyzed the expression of intestinal differentiation markers. Importantly, we found that activation of β-catenin partially rescues the differentiation phenotype of Rbpj deletion mutants, but not the loss of the ISC compartment. Moreover, we identified Bmi1, which is expressed in the ISC and progenitor compartments, as a gene that is co-regulated by Notch and β-catenin. Loss of Bmi1 resulted in reduced proliferation in the ISC compartment accompanied by p16(INK4a) and p19(ARF) (splice variants of Cdkn2a) accumulation, and increased differentiation to the post-mitotic goblet cell lineage that partially mimics Notch loss-of-function defects. Finally, we provide evidence that Bmi1 contributes to ISC self-renewal.
Collapse
Affiliation(s)
| | - Verónica Rodilla
- Program in Cancer Research, IMIM-Hospital del Mar, Barcelona 08003, Spain
| | - Luca Pellegrinet
- Ecole Polytechnique Federale de Lausanne, Lausanne 1015, Switzerland
| | - Jordi Guiu
- Program in Cancer Research, IMIM-Hospital del Mar, Barcelona 08003, Spain
| | - Mar Iglesias
- Department of Pathology, Hospital del Mar, Barcelona 08003, Spain
| | - Angel Carlos Roman
- Department of Biochemistry and Molecular Biology, University of Extremadura, Badajoz 06071, Spain
| | - Susana Gutarra
- Departament de Ciències Experimentals, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Susana González
- Stem Cell Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Pura Muñoz-Cánoves
- Departament de Ciències Experimentals, Universitat Pompeu Fabra, Barcelona 08003, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08003, Spain
| | - Pedro Fernández-Salguero
- Department of Biochemistry and Molecular Biology, University of Extremadura, Badajoz 06071, Spain
| | - Freddy Radtke
- Ecole Polytechnique Federale de Lausanne, Lausanne 1015, Switzerland
| | - Anna Bigas
- Program in Cancer Research, IMIM-Hospital del Mar, Barcelona 08003, Spain
| | - Lluís Espinosa
- Program in Cancer Research, IMIM-Hospital del Mar, Barcelona 08003, Spain
| |
Collapse
|
81
|
Philpott A, Winton DJ. Lineage selection and plasticity in the intestinal crypt. Curr Opin Cell Biol 2014; 31:39-45. [PMID: 25083805 PMCID: PMC4238899 DOI: 10.1016/j.ceb.2014.07.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 07/06/2014] [Accepted: 07/11/2014] [Indexed: 12/21/2022]
Abstract
We know more about the repertoire of cellular behaviours that define the stem and progenitor cells maintaining the intestinal epithelium than any other renewing tissue. Highly dynamic and stochastic processes define cell renewal. Historically the commitment step in differentiation is viewed as a ratchet, irreversibly promoting a given fate and corresponding to a programme imposed at the point of cell division. However, the emerging view of intestinal self-renewal is one of plasticity in which a stem cell state is easily reacquired. The pathway mediators of lineage selection are largely known but how they interface within highly dynamic populations to promote different lineages and yet permit plasticity is not. Advances in understanding gene regulation in the nervous system suggest possible mechanisms.
Collapse
Affiliation(s)
- Anna Philpott
- Department of Oncology, University of Cambridge, Hutchison/Medical Research Council (MRC) Research Centre, Cambridge CB2 0XZ, UK
| | - Douglas J Winton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
| |
Collapse
|
82
|
Mo P, Zhou Q, Guan L, Wang Y, Wang W, Miao M, Tong Z, Li M, Majaz S, Liu Y, Su G, Xu J, Yu C. Amplified in breast cancer 1 promotes colorectal cancer progression through enhancing notch signaling. Oncogene 2014; 34:3935-3945. [PMID: 25263446 PMCID: PMC4377317 DOI: 10.1038/onc.2014.324] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 08/01/2014] [Accepted: 08/25/2014] [Indexed: 12/21/2022]
Abstract
Aberrant activation of Notch signaling has an essential role in colorectal cancer (CRC) progression. Amplified in breast cancer 1 (AIB1), also known as steroid receptor coactivator 3 or NCOA3, is a transcriptional coactivator that promotes cancer cell proliferation and invasiveness. However, AIB1 implication in CRC progression through enhancing Notch signaling is unknown. In this study, we found that several CRC cell lines expressed high levels of AIB1, and knockdown of AIB1 decreased cell proliferation, colony formation and tumorigenesis of these CRC cells. Specifically, knockdown of AIB1 inhibited cell cycle progression at G1 phase by decreasing the mRNA levels of cyclin A2, cyclin B1, cyclin E2 and hairy and enhancer of split (Hes) 1. Furthermore, AIB1 interacted with Notch intracellular domain and Mastermind-like 1 and was recruited to the Hes1 promoter to enhance Notch signaling. Downregulation of AIB1 also decreased CRC cell invasiveness in vitro and lung metastasis in vivo. Besides that, knockout of AIB1 in mice inhibited colon carcinogenesis induced by azoxymethane/dextran sodium sulfate treatment. The mRNA levels of cyclin B1 and Hes5 were downregulated, but p27, ATOH1 and MUC2 were upregulated in the colon tumors from AIB1-deficient mice compared with those from wild-type mice. Thus, our results signify the importance of AIB1 in CRC and demonstrate that AIB1 promotes CRC progression at least in part through enhancing Notch signaling, suggesting that AIB1 is a potential molecular target for CRC treatment.
Collapse
Affiliation(s)
- Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qiling Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Lei Guan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yi Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Wei Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Mengmeng Miao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhangwei Tong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Ming Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Sidra Majaz
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yonghong Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Guoqiang Su
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
83
|
Gracz AD, Magness ST. Defining hierarchies of stemness in the intestine: evidence from biomarkers and regulatory pathways. Am J Physiol Gastrointest Liver Physiol 2014; 307:G260-73. [PMID: 24924746 PMCID: PMC4121637 DOI: 10.1152/ajpgi.00066.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
For decades, the rapid proliferation and well-defined cellular lineages of the small intestinal epithelium have driven an interest in the biology of the intestinal stem cells (ISCs) and progenitors that produce the functional cells of the epithelium. Recent and significant advances in ISC biomarker discovery have established the small intestinal epithelium as a powerful model system for studying general paradigms in somatic stem cell biology and facilitated elegant genetic and functional studies of stemness in the intestine. However, this newfound wealth of ISC biomarkers raises important questions of marker specificity. Furthermore, the ISC field must now begin to reconcile biomarker status with functional stemness, a challenge that is made more complex by emerging evidence that cellular hierarchies in the intestinal epithelium are more plastic than previously imagined, with some progenitor populations capable of dedifferentiating and functioning as ISCs following damage. In this review, we discuss the state of the ISC field in terms of biomarkers, tissue dynamics, and cellular hierarchies, and how these processes might be informed by earlier studies into signaling networks in the small intestine.
Collapse
Affiliation(s)
- A. D. Gracz
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; ,2Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - S. T. Magness
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; ,2Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and ,3Joint Department of Biomedical Engineering, University of North Carolina/North Carolina State University, Chapel Hill, North Carolina
| |
Collapse
|
84
|
Giammanco A, Blanc V, Montenegro G, Klos C, Xie Y, Kennedy S, Luo J, Chang SH, Hla T, Nalbantoglu I, Dharmarajan S, Davidson NO. Intestinal epithelial HuR modulates distinct pathways of proliferation and apoptosis and attenuates small intestinal and colonic tumor development. Cancer Res 2014; 74:5322-35. [PMID: 25085247 DOI: 10.1158/0008-5472.can-14-0726] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
HuR is a ubiquitous nucleocytoplasmic RNA-binding protein that exerts pleiotropic effects on cell growth and tumorigenesis. In this study, we explored the impact of conditional, tissue-specific genetic deletion of HuR on intestinal growth and tumorigenesis in mice. Mice lacking intestinal expression of HuR (Hur (IKO) mice) displayed reduced levels of cell proliferation in the small intestine and increased sensitivity to doxorubicin-induced acute intestinal injury, as evidenced by decreased villus height and a compensatory shift in proliferating cells. In the context of Apc(min/+) mice, a transgenic model of intestinal tumorigenesis, intestinal deletion of the HuR gene caused a three-fold decrease in tumor burden characterized by reduced proliferation, increased apoptosis, and decreased expression of transcripts encoding antiapoptotic HuR target RNAs. Similarly, Hur(IKO) mice subjected to an inflammatory colon carcinogenesis protocol [azoxymethane and dextran sodium sulfate (AOM-DSS) administration] exhibited a two-fold decrease in tumor burden. Hur(IKO) mice showed no change in ileal Asbt expression, fecal bile acid excretion, or enterohepatic pool size that might explain the phenotype. Moreover, none of the HuR targets identified in Apc(min/+)Hur(IKO) were altered in AOM-DSS-treated Hur(IKO) mice, the latter of which exhibited increased apoptosis of colonic epithelial cells, where elevation of a unique set of HuR-targeted proapoptotic factors was documented. Taken together, our results promote the concept of epithelial HuR as a contextual modifier of proapoptotic gene expression in intestinal cancers, acting independently of bile acid metabolism to promote cancer. In the small intestine, epithelial HuR promotes expression of prosurvival transcripts that support Wnt-dependent tumorigenesis, whereas in the large intestine epithelial HuR indirectly downregulates certain proapoptotic RNAs to attenuate colitis-associated cancer. Cancer Res; 74(18); 5322-35. ©2014 AACR.
Collapse
Affiliation(s)
- Antonina Giammanco
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Valerie Blanc
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Grace Montenegro
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Coen Klos
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Yan Xie
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Susan Kennedy
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Jianyang Luo
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Sung-Hee Chang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, New York
| | - Timothy Hla
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, New York
| | - Ilke Nalbantoglu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri
| | - Sekhar Dharmarajan
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Nicholas O Davidson
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri.
| |
Collapse
|
85
|
Wang L, Jin F, Qin A, Hao Y, Dong Y, Ge S, Dai K. Targeting Notch1 signaling pathway positively affects the sensitivity of osteosarcoma to cisplatin by regulating the expression and/or activity of Caspase family. Mol Cancer 2014; 13:139. [PMID: 24894297 PMCID: PMC4110525 DOI: 10.1186/1476-4598-13-139] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/26/2014] [Indexed: 12/31/2022] Open
Abstract
Background The introduction of cisplatin has improved the long-term survival rate in osteosarcoma patients. However, some patients are intrinsically resistant to cisplatin. This study reported that the activation of Notch1 is positively correlated with cisplatin sensitivity, evidenced by both clinical and in vitro data. Results In this study, a total 8 osteosarcoma specimens were enrolled and divided into two groups according to their cancer chemotherapeutic drugs sensitivity examination results. The relationship between Notch1 expression and cisplatin sensitivity of osteosarcoma patients was detected by immunohistochemistry and semi-quantitative analysis. Subsequently, two typical osteosarcoma cell lines, Saos-2 and MG63, were selected to study the changes of cisplatin sensitivity by up-regulating (NICD1 plasmid transfeciton) or decreasing (gamma-secretase complex inhibitor DAPT) the activation state of Notch1 signaling pathway. Our results showed a significant correlation between the expression of Notch1 and cisplatin sensitivity in patient specimens. In vitro, Saos-2 with higher expression of Notch1 had significantly better cisplatin sensitivity than MG63 whose Notch1 level was relatively lower. By targeting regulation in vitro, the cisplatin sensitivity of Saos-2 and MG63 had significantly increased after the activation of Notch1 signaling pathway, and vice versa. Further mechanism investigation revealed that activation/inhibition of Notch1 sensitized/desensitized cisplatin-induced apoptosis, which probably depended on the changes in the activity of Caspase family, including Caspase 3, Caspase 8 and Caspase 9 in these cells. Conclusions Our data clearly demonstrated that Notch1 is critical for cisplatin sensitivity in osteosarcoma. It can be used as a molecular marker and regulator for cisplatin sensitivity in osteosarcoma patients.
Collapse
Affiliation(s)
| | | | | | - Yongqiang Hao
- Department of Orthopaedics, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, People's Republic of China.
| | | | | | | |
Collapse
|
86
|
Chen G, Zhang Z, Cheng Y, Xiao W, Qiu Y, Yu M, Sun L, Wang W, Du G, Gu Y, Peng K, Xu C, Yang H. The canonical Notch signaling was involved in the regulation of intestinal epithelial cells apoptosis after intestinal ischemia/reperfusion injury. Int J Mol Sci 2014; 15:7883-96. [PMID: 24806344 PMCID: PMC4057709 DOI: 10.3390/ijms15057883] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 04/21/2014] [Accepted: 04/24/2014] [Indexed: 11/23/2022] Open
Abstract
Notch signaling plays a critical role in the maintenance of intestinal homeostasis. The aim of the present study was to investigate the role of Notch signaling in the apoptosis of intestinal epithelial cells after intestinal ischemia reperfusion (I/R) injury. Male C57BL/6 mice were subjected to sham operation or I/R injury. Intestinal tissue samples were collected at 12 h after reperfusion. TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling) staining showed that intestinal I/R injury induced significantly increased apoptosis of intestinal epithelial cells. Meanwhile, the mRNA expression of Jagged1, DLL1, Notch2, and Hes5, and protein expression of NICD2 and Hes5 were increased significantly after I/R injury in intestinal epithelial cells. In an in vitro IEC-6 culture model, flow cytometry analyses showed that inhibition of Notch signaling by γ-secretase inhibitor DAPT and the suppression of Hes5 expression using siRNA both significantly increased the apoptosis of IEC-6 cells under the condition of hypoxia/reoxygenation (H/R). In conclusion, the Notch2/Hes5 signaling pathway was activated and involved in the regulation of intestinal epithelial cells apoptosis in intestinal I/R injury.
Collapse
Affiliation(s)
- Guoqing Chen
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Zhicao Zhang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Yingdong Cheng
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Wensheng Wang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Guangsheng Du
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Yingchao Gu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Ke Peng
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Chao Xu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
87
|
Sheaffer KL, Kim R, Aoki R, Elliott EN, Schug J, Burger L, Schübeler D, Kaestner KH. DNA methylation is required for the control of stem cell differentiation in the small intestine. Genes Dev 2014; 28:652-64. [PMID: 24637118 PMCID: PMC3967052 DOI: 10.1101/gad.230318.113] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is a tight correlation between the epigenetic status of genes and expression changes during differentiation. Sheaffer et al. used acute deletion of Dnmt1 to reduce DNA methylation maintenance in the intestinal epithelium. This caused crypt expansion and decreased differentiation. DNA methylation was dynamic at enhancers during the rapid transition from stem to differentiated epithelial cells. These findings reveal that the loss of DNA methylation at intestinal stem cell gene enhancers causes inappropriate gene expression and delayed differentiation. The mammalian intestinal epithelium has a unique organization in which crypts harboring stem cells produce progenitors and finally clonal populations of differentiated cells. Remarkably, the epithelium is replaced every 3–5 d throughout adult life. Disrupted maintenance of the intricate balance of proliferation and differentiation leads to loss of epithelial integrity or barrier function or to cancer. There is a tight correlation between the epigenetic status of genes and expression changes during differentiation; however, the mechanism of how changes in DNA methylation direct gene expression and the progression from stem cells to their differentiated descendants is unclear. Using conditional gene ablation of the maintenance methyltransferase Dnmt1, we demonstrate that reducing DNA methylation causes intestinal crypt expansion in vivo. Determination of the base-resolution DNA methylome in intestinal stem cells and their differentiated descendants shows that DNA methylation is dynamic at enhancers, which are often associated with genes important for both stem cell maintenance and differentiation. We establish that the loss of DNA methylation at intestinal stem cell gene enhancers causes inappropriate gene expression and delayed differentiation.
Collapse
Affiliation(s)
- Karyn L Sheaffer
- Department of Genetics, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Ogaki S, Shiraki N, Kume K, Kume S. Wnt and Notch signals guide embryonic stem cell differentiation into the intestinal lineages. Stem Cells 2014; 31:1086-96. [PMID: 23378042 DOI: 10.1002/stem.1344] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 01/10/2013] [Indexed: 12/11/2022]
Abstract
The studies of differentiation of mouse or human embryonic stem cells (hESCs) into specific cell types of the intestinal cells would provide insights to the understanding of intestinal development and ultimately yield cells for the use in future regenerative medicine. Here, using an in vitro differentiation procedure of pluripotent stem cells into definitive endoderm (DE), inductive signal pathways' guiding differentiation into intestinal cells was investigated. We found that activation of Wnt/β-catenin and inhibition of Notch signaling pathways, by simultaneous application of 6-bromoindirubin-3'-oxime (BIO), a glycogen synthase kinase-3β inhibitor, and N-[(3,5-Difluorophenyl)acetyl]-L-alanyl-2-phenylglycine-1,1-dimethylethyl ester (DAPT), a known γ-secretase inhibitor, efficiently induced intestinal differentiation of ESCs cultured on feeder cell. BIO and DAPT patterned the DE at graded concentrations. Upon prolonged culture on feeder cells, all four intestinal differentiated cell types, the absorptive enterocytes and three types of secretory cells (goblet cells, enteroendocrine cells, and Paneth cells), were efficiently differentiated from mouse and hESC-derived intestinal epithelium cells. Further investigation revealed that in the mouse ESCs, fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) signaling act synergistically with BIO and DAPT to potentiate differentiation into the intestinal epithelium. However, in hESCs, FGF signaling inhibited, and BMP signaling did not affect differentiation into the intestinal epithelium. We concluded that Wnt and Notch signaling function to pattern the anterior-posterior axis of the DE and control intestinal differentiation.
Collapse
Affiliation(s)
- Soichiro Ogaki
- Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo, Kumamoto, Japan
| | | | | | | |
Collapse
|
89
|
Kandasamy J, Huda S, Ambalavanan N, Jilling T. Inflammatory signals that regulate intestinal epithelial renewal, differentiation, migration and cell death: Implications for necrotizing enterocolitis. ACTA ACUST UNITED AC 2014; 21:67-80. [PMID: 24533974 DOI: 10.1016/j.pathophys.2014.01.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Necrotizing enterocolitis is a disease entity with multiple proposed pathways of pathogenesis. Various combinations of these risk factors, perhaps based on genetic predisposition, possibly lead to the mucosal and epithelial injury that is the hallmark of NEC. Intestinal epithelial integrity is controlled by a tightly regulated balance between proliferation and differentiation of epithelium from intestinal epithelial stem cells and cellular loss by apoptosis. various signaling pathways play a key role in creating and maintaining this balance. The aim of this review article is to outline intestinal epithelial barrier development and structure and the impact of these inflammatory signaling and regulatory pathways as they pertain to the pathogenesis of NEC.
Collapse
Affiliation(s)
- Jegen Kandasamy
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Shehzad Huda
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Namasivayam Ambalavanan
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Tamas Jilling
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA.
| |
Collapse
|
90
|
Kobayashi T, Kageyama R. Expression Dynamics and Functions of Hes Factors in Development and Diseases. Curr Top Dev Biol 2014; 110:263-83. [DOI: 10.1016/b978-0-12-405943-6.00007-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
91
|
Jeon MK, Klaus C, Kaemmerer E, Gassler N. Intestinal barrier: Molecular pathways and modifiers. World J Gastrointest Pathophysiol 2013; 4:94-99. [PMID: 24244877 PMCID: PMC3829455 DOI: 10.4291/wjgp.v4.i4.94] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/28/2013] [Accepted: 09/03/2013] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract is frequently challenged by pathogens/antigens contained in food and water and the intestinal epithelium must be capable of rapid regeneration in the event of tissue damage. Disruption of the intestinal barrier leads to a number of immune-mediated diseases, including inflammatory bowel disease, food allergy, and celiac disease. The intestinal mucosa is composed of different types of epithelial cells in specific barrier functions. Epithelial cells control surface-associated bacterial populations without disrupting the intestinal microflora that is crucial for host health. They are also capable of modulating mucosal immune system, and are thus essential in maintaining homeostasis in the gut. Thus, the regulation of intestinal epithelial homeostasis is crucial for the maintenance of the structure of the mucosa and the defensive barrier functions. Recent studies have demonstrated that multiple molecular pathways are involved in the regulation of intestinal epithelial cell polarity. These include the Wnt, Notch, Hippo, transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) and Hedgehog pathways, most of which were identified in lower organisms where they play important roles during embryogenesis. These pathways are also used in adult organisms to regulate multiple self-renewing organs. Understanding the interactions between these molecular mechanisms and intestinal barrier function will therefore provide important insight into the pathogenesis of intestinal-based immune-mediated diseases.
Collapse
|
92
|
Abstract
The intestine has become a prime model system to study stem cell biology. Intestinal stem cells can be identified based on the expression of a unique marker gene, namely Lgr5. A transgenic mouse model expressing green fluorescent protein in intestinal stem cells has allowed their visualization, isolation, molecular characterization and use in generating organoids: small mini-guts that contain all cell types of the intestine. Detailing the behavior of intestinal stem cells has also led to new insights concerning the mechanism of self-renewal versus differentiation. Genes and pathways directing daughter cells of stem cells towards the differentiated lineages of the intestine are getting better defined. Of all differentiated cells, Paneth cells play a distinguished role: they emerged from pure bystanders to the guardians of the stem cell. Taken together, a detailed molecular picture emerges that describes the mechanisms of intestinal homeostatic self-renewal and outlines new therapeutic avenues.
Collapse
Affiliation(s)
- Daniel E Stange
- Department of General, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, University Dresden, Dresden, Germany
| |
Collapse
|
93
|
Poser SW, Park DM, Androutsellis-Theotokis A. The STAT3-Ser/Hes3 signaling axis: an emerging regulator of endogenous regeneration and cancer growth. Front Physiol 2013; 4:273. [PMID: 24101906 PMCID: PMC3787304 DOI: 10.3389/fphys.2013.00273] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 09/11/2013] [Indexed: 12/19/2022] Open
Abstract
Stem cells, by definition, are able to both self-renew (give rise to more cells of their own kind) and demonstrate multipotential (the ability to differentiate into multiple cell types). To accommodate this unique dual ability, stem cells interpret signal transduction pathways in specialized ways. Notable examples include canonical and non-canonical branches of the Notch signaling pathway, with each controlling different downstream targets (e.g., Hes1 vs. Hes3) and promoting either differentiation or self-renewal. Similarly, stem cells utilize STAT3 signaling uniquely. Most mature cells studied thus far rely on tyrosine phosphorylation (STAT3-Tyr) to promote survival and growth; in contrast, STAT3-Tyr induces the differentiation of neural stem cells (NSCs). NSCs use an alternative phosphorylation site, STAT3-Ser, to regulate survival and growth, a site that is largely redundant for this function in most other cell types. STAT3-Ser regulates Hes3, and together they form a convergence point for several signals, including Notch, Tie2, and insulin receptor activation. Disregulation and manipulation of the STAT3-Ser/Hes3 signaling pathway is important in both tumorigenesis and regenerative medicine, and worthy of extensive study.
Collapse
Affiliation(s)
- Steven W Poser
- Department of Medicine, University of Dresden Dresden, Germany
| | | | | |
Collapse
|
94
|
Regulation of intestinal stem cells by Wnt and Notch signalling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:175-86. [PMID: 23696357 DOI: 10.1007/978-94-007-6621-1_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mammalian intestine is lined by an epithelial cell layer that is constantly renewed via a population of stem cells that reside in a specialised niche within intestinal crypts. The recent development of tools that permit genetic manipulation and lineage tracing of cells in vivo combined with culture methods in vitro has made the intestine particularly amenable for the study of signals that regulate stem cell function. Both Wnt and Notch signalling are critical regulators of stem cell fate. Gene knockout and transgenic expression analysis combined with meticulous analysis of lineage tracing and molecular characterisation has contributed to the definition of the mechanisms by which these pathways act during normal homeostasis and in disease states.
Collapse
|
95
|
Chen G, Sun L, Yu M, Meng D, Wang W, Yang Y, Yang H. The Jagged-1/Notch-1/Hes-1 pathway is involved in intestinal adaptation in a massive small bowel resection rat model. Dig Dis Sci 2013; 58:2478-86. [PMID: 23595520 DOI: 10.1007/s10620-013-2680-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 04/04/2013] [Indexed: 12/27/2022]
Abstract
BACKGROUND Notch signaling is required for the maintenance of intestinal epithelial proliferation. Dysfunction of this signaling pathway is associated with the loss of proliferated crypt epithelial cells. AIM The aim of this study was to investigate the role of Notch signaling in small bowel resection (SBR)-associated crypt epithelial cell proliferation. METHODS Male Sprague-Dawley rats were subjected to sham operation (bowel transection and reanastomosis) or 70% mid-SBR. Intestinal tissue samples were collected at 0.5, 1, 6, 12, 24, 72, and 168 h after operation. The expression of Notch pathway mRNAs and proteins was analyzed using RT-PCR and Western blot. The expression of the Notch pathway proteins Jagged-1, NICD and Hes-1 was also determined through immunohistochemical staining using day 3 postoperative intestinal tissues. The degree of crypt epithelial cell proliferation was evaluated using the immunohistochemical staining of proliferating cell nuclear antigen (PCNA). Furthermore, IEC-6 cells were used to examine the function of the Jagged-1 signaling system. RESULTS SBR led to increased crypt epithelial cell proliferation and increased expression of Jagged-1 and Hes-1 mRNA and protein along with cleaved Notch-1. Immunohistochemical staining showed that Jagged-1, cleaved Notch-1 and Hes-1 colocalized in the same proliferated crypt epithelial cell population. Recombinant Jagged-1 significantly stimulated the proliferation of IEC-6 cells. Transient upregulation of Jagged-2 expression was found 1 h after SBR, and it was accompanied by cleaved Notch-1 and Hes-1 upregulation. CONCLUSION The Jagged-1/Notch-1/Hes-1 signaling pathway is involved in intestinal adaptation through increasing crypt epithelial cell proliferation.
Collapse
Affiliation(s)
- Guoqing Chen
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | | | | | | | | | | | | |
Collapse
|
96
|
Candy PA, Phillips MR, Redfern AD, Colley SM, Davidson JA, Stuart LM, Wood BA, Zeps N, Leedman PJ. Notch-induced transcription factors are predictive of survival and 5-fluorouracil response in colorectal cancer patients. Br J Cancer 2013; 109:1023-30. [PMID: 23900217 PMCID: PMC3749585 DOI: 10.1038/bjc.2013.431] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/29/2013] [Accepted: 07/04/2013] [Indexed: 12/15/2022] Open
Abstract
Background: The purpose of this study was to evaluate the expression of Notch-induced transcription factors (NTFs) HEY1, HES1 and SOX9 in colorectal cancer (CRC) patients to determine their clinicopathologic and prognostic significance. Methods: Levels of HEY1, HES1 and SOX9 protein were measured by immunohistochemistry in a nonmalignant and malignant tissue microarray of 441 CRC patients, and the findings correlated with pathologic, molecular and clinical variables. Results: The NTFs HEY1, HES1 and SOX9 were overexpressed in tumours relative to colonic mucosa (OR=3.44, P<0.0001; OR=7.40, P<0.0001; OR=4.08 P<0.0001, respectively). HEY1 overexpression was a negative prognostic factor for all CRC patients (HR=1.29, P=0.023) and strongly correlated with perineural and vascular invasion and lymph node (LN) metastasis. In 5-fluorouracil (5-FU)-treated patients, the tumour overexpression of SOX9 correlated with markedly poorer survival (HR=8.72, P=0.034), but had no predictive effect in untreated patients (HR=0.70, P=0.29). When HEY1, HES1 and SOX9 expression were combined to predict survival with chemotherapy, in treated patients there was an additive increase in the risk of death with each NTF overexpressed (HR=2.09, P=0.01), but no prognostic import in the untreated patient group (HR=0.74, P=0.19). Conclusion: The present study is the first to discover that HEY1 overexpression correlates with poorer outcome in CRC, and NTF expression is predictive of CRC patient survival with 5-FU chemotherapy. If confirmed in future studies, testing of NTF expression has the potential to enter routine pathological practice for the selection of patients to undergo chemotherapy alone or in combination with Notch inhibitors.
Collapse
Affiliation(s)
- P A Candy
- Laboratory for Cancer Medicine, University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Western Australia 6000, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Affiliation(s)
- Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands.
| |
Collapse
|
98
|
Abstract
Notch signaling has been shown over the past few decades to play fundamental roles in a plethora of developmental processes in an evolutionarily conserved fashion. Notch-mediated cell-to-cell signaling is involved in many aspects of embryonic development and control of tissue homeostasis in a variety of adult tissues, and regulates stem cell maintenance, cell differentiation and cellular homeostasis. The focus of this Review is the role of Notch signaling in stem cells, comparing insights from flies, fish and mice to highlight similarities, as well as differences, between species, tissues and stem cell compartments.
Collapse
Affiliation(s)
- Ute Koch
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Science, SwissInstitute for Experimental Cancer Research (ISREC), Station 19, 1015 Lausanne, Switzerland.
| | | | | |
Collapse
|
99
|
Manosalva I, González A, Kageyama R. Hes1 in the somatic cells of the murine ovary is necessary for oocyte survival and maturation. Dev Biol 2013; 375:140-51. [DOI: 10.1016/j.ydbio.2012.12.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 01/06/2023]
|
100
|
Abstract
The small and large intestines are tubular organs composed of several tissue types. The columnar epithelium that lines the inner surface of the intestines distinguishes the digestive physiology of each region of the intestine and consists of several distinct cell types that are rapidly and continually renewed by intestinal stem cells that reside near the base of the crypts of Lieberkühn. Notch signaling controls the fate of intestinal stem cells by regulating the expression of Hes genes and by repressing Atoh1. Alternate models of Notch pathway control of cell fate determination are presented. Roles for Notch signaling in development of the intestine, including mesenchymal and neural cells, are discussed. The oncogenic activities of Notch in colorectal cancer, as well as the tumor suppressive activities of Atoh1, are reviewed. Therapeutic targeting of the Notch pathway in colorectal cancers is discussed, along with potential caveats.
Collapse
Affiliation(s)
- Taeko K Noah
- Division of Gastroenterology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | |
Collapse
|