51
|
Abstract
Colorectal cancer is one of the so-called westernized diseases and the second leading cause of cancer death worldwide. On the basis of global epidemiological and scientific studies, evidence suggests that the risk of colorectal cancer is increased by processed and unprocessed meat consumption but suppressed by fibre, and that food composition affects colonic health and cancer risk via its effects on colonic microbial metabolism. The gut microbiota can ferment complex dietary residues that are resistant to digestion by enteric enzymes. This process provides energy for the microbiota but culminates in the release of short-chain fatty acids including butyrate, which are utilized for the metabolic needs of the colon and the body. Butyrate has a remarkable array of colonic health-promoting and antineoplastic properties: it is the preferred energy source for colonocytes, it maintains mucosal integrity and it suppresses inflammation and carcinogenesis through effects on immunity, gene expression and epigenetic modulation. Protein residues and fat-stimulated bile acids are also metabolized by the microbiota to inflammatory and/or carcinogenic metabolites, which increase the risk of neoplastic progression. This Review will discuss the mechanisms behind these microbial metabolite effects, which could be modified by diet to achieve the objective of preventing colorectal cancer in Western societies.
Collapse
|
52
|
Xu LM, Chen L, Li F, Zhang R, Li ZY, Chen FF, Jiang XD. Over-expression of the long non-coding RNA HOTTIP inhibits glioma cell growth by BRE. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:162. [PMID: 27733185 PMCID: PMC5062847 DOI: 10.1186/s13046-016-0431-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/20/2016] [Indexed: 12/14/2022]
Abstract
Background Gliomas are the most common type of primary brain tumour in the central nervous system of adults. The long non-coding RNA (lncRNA) HOXA transcript at the distal tip (HOTTIP) is transcribed from the 5′ tip of the HOXA locus. HOTTIP has recently been shown to be dysregulated and play an important role in the progression of several cancers. However, little is known about whether and how HOTTIP regulates glioma development. Methods In this study, we assayed the expression of HOTTIP in glioma tissue samples and glioma cell lines using real-time polymerase chain reaction and defined the biological functions of HOTTIP using the CCK-8 assay, flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL assay) and tumour formation assay in a nude mouse model. Finally, we discovered the underlying mechanism using the Apoptosis PCR 384HT Array, Western blot, RNA immunoprecipitation (RIP) and luciferase reporter assay. Results HOTTIP was aberrantly down-regulated in glioma tissues and glioma cell lines (U87-MG, U118-MG, U251 and A172), and over-expression of HOTTIP inhibited the growth of glioma cell lines in vitro and in vivo. Furthermore, HOTTIP could directly bind to the brain and reproductive expression (BRE) gene and down-regulate BRE gene expression. In addition, we further verified that over-expression of the BRE gene promoted the growth of glioma cell lines in vitro. Finally, over-expression of HOTTIP significantly suppressed the expression of the cyclin A and CDK2 proteins and increased the expression of the P53 protein. However, we found that the over-expression of BRE significantly increased the expression of the cyclin A and CDK2 proteins and suppressed the expression of the P53 protein. Taken together, these findings suggested that high levels of HOTTIP reduced glioma cell growth. Additionally, the mechanism of HOTTIP-mediated reduction of glioma cell growth may involve the suppression of cyclin A and CDK2 protein expression, which increases P53 protein expression via the down-regulation of BRE. Conclusions Our studies demonstrated that over-expression of HOTTIP promotes cell apoptosis and inhibits cell growth in U118-MG and U87-MG human glioma cell lines by down-regulating BRE expression to regulate the expression of P53, CDK2 and Cyclin A proteins. The data described in this study indicate that HOTTIP is an interesting candidate for further functional studies in glioma and demonstrate the potential application of HOTTIP in glioma therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0431-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Min Xu
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Lei Chen
- Department of Neurosurgery, Shenzhen Second People' s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518029, China
| | - Feng Li
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Run Zhang
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Zong-Yang Li
- Department of Neurosurgery, Shenzhen Second People' s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518029, China
| | - Fan-Fan Chen
- Department of Neurosurgery, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Xiao-Dan Jiang
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
53
|
Chen HJ, Zheng Y, Wei Z. Advancements in Modeling Colorectal Cancer in Rodents. CURRENT COLORECTAL CANCER REPORTS 2016. [DOI: 10.1007/s11888-016-0334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
54
|
Hanley MP, Kadaveru K, Perret C, Giardina C, Rosenberg DW. Dietary Methyl Donor Depletion Suppresses Intestinal Adenoma Development. Cancer Prev Res (Phila) 2016; 9:812-820. [PMID: 27530130 DOI: 10.1158/1940-6207.capr-16-0042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 08/01/2016] [Indexed: 01/07/2023]
Abstract
The role of folate one-carbon metabolism in colorectal cancer development is controversial, with nutritional intervention studies producing conflicting results. It has been reported that ApcMin/+ mice maintained on a diet deficient in the methyl donors folic acid, methionine, choline, and vitamin B12, and supplemented with homocysteine, show a greater than 95% reduction in intestinal tumor development. The present study extends these findings and shows that tumor protection afforded by dietary methyl donor deficiency (MDD) is long-lasting. After 11 weeks of MDD, tumor protection persisted for at least an additional 7 weeks of methyl donor repletion (22.2 ± 3.5 vs. 70.2 ± 4.6 tumors per mouse; P < 0.01). Sustained tumor protection was associated with a reduction in intestinal crypt length (26%, P < 0.01), crypt cell division and crypt fission, and an increase in apoptosis of both normal crypts and tumors (4.9- and 3.2-fold, respectively, P < 0.01). MDD also caused a significant reduction in the number of Dclk1-positive cells in the intestine (62%, P < 0.01), a long-lived crypt cell with cancer stem cell potential. Several undesirable effects associated with methyl donor restriction (e.g., reduced body weight gain) were shown to be transient and readily reversible following methyl donor repletion. Taken together, these results indicate that even temporary dietary methyl donor restriction in adenoma-prone mice can induce persistent changes to the intestinal epithelium and provide long-lasting tumor protection. These data also suggest that transient reductions in dietary methyl donor consumption should be considered when studying the impact of folate on colon cancer risk in humans. Cancer Prev Res; 9(10); 812-20. ©2016 AACR.
Collapse
Affiliation(s)
- Matthew P Hanley
- Center for Molecular Medicine, Neag Comprehensive Cancer Center, School of Medicine, UConn Health, Farmington, Connecticut
| | | | - Christine Perret
- Institut Cochin de Génétique Moléculaire INSERM U129, Paris, France
| | - Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Daniel W Rosenberg
- Center for Molecular Medicine, Neag Comprehensive Cancer Center, School of Medicine, UConn Health, Farmington, Connecticut.
| |
Collapse
|
55
|
Sarkar A, Huebner AJ, Sulahian R, Anselmo A, Xu X, Flattery K, Desai N, Sebastian C, Yram MA, Arnold K, Rivera M, Mostoslavsky R, Bronson R, Bass AJ, Sadreyev R, Shivdasani RA, Hochedlinger K. Sox2 Suppresses Gastric Tumorigenesis in Mice. Cell Rep 2016; 16:1929-41. [PMID: 27498859 DOI: 10.1016/j.celrep.2016.07.034] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 03/22/2016] [Accepted: 07/14/2016] [Indexed: 01/10/2023] Open
Abstract
Sox2 expression marks gastric stem and progenitor cells, raising important questions regarding the genes regulated by Sox2 and the role of Sox2 itself during stomach homeostasis and disease. By using ChIP-seq analysis, we have found that the majority of Sox2 targets in gastric epithelial cells are tissue specific and related to functions such as endoderm development, Wnt signaling, and gastric cancer. Unexpectedly, we found that Sox2 itself is dispensable for gastric stem cell and epithelial self-renewal, yet Sox2(+) cells are highly susceptible to tumorigenesis in an Apc/Wnt-driven mouse model. Moreover, Sox2 loss enhances, rather than impairs, tumor formation in Apc-deficient gastric cells in vivo and in vitro by inducing Tcf/Lef-dependent transcription and upregulating intestinal metaplasia-associated genes, providing a mechanistic basis for the observed phenotype. Together, these data identify Sox2 as a context-dependent tumor suppressor protein that is dispensable for normal tissue regeneration but restrains stomach adenoma formation through modulation of Wnt-responsive and intestinal genes.
Collapse
Affiliation(s)
- Abby Sarkar
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA
| | - Aaron J Huebner
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA
| | - Rita Sulahian
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Anthony Anselmo
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xinsen Xu
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Kyle Flattery
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA
| | - Niyati Desai
- Division of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Carlos Sebastian
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mary Anna Yram
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA
| | - Katrin Arnold
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA
| | - Miguel Rivera
- Division of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Raul Mostoslavsky
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Roderick Bronson
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Adam J Bass
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ramesh A Shivdasani
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Konrad Hochedlinger
- Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute and Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
56
|
Mcilhatton MA, Boivin GP, Groden J. Manipulation of DNA Repair Proficiency in Mouse Models of Colorectal Cancer. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1414383. [PMID: 27413734 PMCID: PMC4931062 DOI: 10.1155/2016/1414383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/09/2016] [Indexed: 12/20/2022]
Abstract
Technical and biological innovations have enabled the development of more sophisticated and focused murine models that increasingly recapitulate the complex pathologies of human diseases, in particular cancer. Mouse models provide excellent in vivo systems for deciphering the intricacies of cancer biology within the context of precise experimental settings. They present biologically relevant, adaptable platforms that are amenable to continual improvement and refinement. We discuss how recent advances in our understanding of tumorigenesis and the underlying deficiencies of DNA repair mechanisms that drive it have been informed by using genetically engineered mice to create defined, well-characterized models of human colorectal cancer. In particular, we focus on how mechanisms of DNA repair can be manipulated precisely to create in vivo models whereby the underlying processes of tumorigenesis are accelerated or attenuated, dependent on the composite alleles carried by the mouse model. Such models have evolved to the stage where they now reflect the initiation and progression of sporadic cancers. The review is focused on mouse models of colorectal cancer and how insights from these models have been instrumental in shaping our understanding of the processes and potential therapies for this disease.
Collapse
Affiliation(s)
- Michael A. Mcilhatton
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12th Avenue, Columbus, OH 43210, USA
| | - Gregory P. Boivin
- Department of Pathology, Boonshoft School of Medicine, Wright State University, Health Sciences Building 053, 3640 Colonel Glenn Highway, Dayton, OH 45435, USA
| | - Joanna Groden
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
57
|
SLAP displays tumour suppressor functions in colorectal cancer via destabilization of the SRC substrate EPHA2. Nat Commun 2016; 5:3159. [PMID: 24457997 DOI: 10.1038/ncomms4159] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 12/19/2013] [Indexed: 12/11/2022] Open
Abstract
The adaptor SLAP is a negative regulator of receptor signalling in immune cells but its role in human cancer is ill defined. Here we report that SLAP is abundantly expressed in healthy epithelial intestine but strongly downregulated in 50% of colorectal cancer. SLAP overexpression suppresses cell tumorigenicity and invasiveness while SLAP silencing enhances these transforming properties. Mechanistically, SLAP controls SRC/EPHA2/AKT signalling via destabilization of the SRC substrate and receptor tyrosine kinase EPHA2. This activity is independent from CBL but requires SLAP SH3 interaction with the ubiquitination factor UBE4A and SLAP SH2 interaction with pTyr594-EPHA2. SRC phosphorylates EPHA2 on Tyr594, thus creating a feedback loop that promotes EPHA2 destruction and thereby self-regulates its transforming potential. SLAP silencing enhances SRC oncogenicity and sensitizes colorectal tumour cells to SRC inhibitors. Collectively, these data establish a tumour-suppressive role for SLAP in colorectal cancer and a mechanism of SRC oncogenic induction through stabilization of its cognate substrates.
Collapse
|
58
|
Beyaz S, Mana MD, Roper J, Kedrin D, Saadatpour A, Hong SJ, Bauer-Rowe KE, Xifaras ME, Akkad A, Arias E, Pinello L, Katz Y, Shinagare S, Abu-Remaileh M, Mihaylova MM, Lamming DW, Dogum R, Guo G, Bell GW, Selig M, Nielsen GP, Gupta N, Ferrone CR, Deshpande V, Yuan GC, Orkin SH, Sabatini DM, Yilmaz ÖH. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 2016; 531:53-8. [PMID: 26935695 PMCID: PMC4846772 DOI: 10.1038/nature17173] [Citation(s) in RCA: 547] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 01/28/2016] [Indexed: 12/27/2022]
Abstract
Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.
Collapse
Affiliation(s)
- Semir Beyaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Miyeko D Mana
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| | - Jatin Roper
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Division of Gastroenterology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts 02111, USA
| | - Dmitriy Kedrin
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Assieh Saadatpour
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Sue-Jean Hong
- Whitehead Institute for Biomedical Research, Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, Massachusetts 02142, USA
| | - Khristian E Bauer-Rowe
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| | - Michael E Xifaras
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| | - Adam Akkad
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| | - Erika Arias
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| | - Luca Pinello
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Yarden Katz
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Shweta Shinagare
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| | - Monther Abu-Remaileh
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Whitehead Institute for Biomedical Research, Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, Massachusetts 02142, USA
| | - Maria M Mihaylova
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Whitehead Institute for Biomedical Research, Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, Massachusetts 02142, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Rizkullah Dogum
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
| | - Guoji Guo
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - George W Bell
- Whitehead Institute for Biomedical Research, Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, Massachusetts 02142, USA
| | - Martin Selig
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - G Petur Nielsen
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Nitin Gupta
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, Missisippi 39216, USA
| | - Cristina R Ferrone
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Vikram Deshpande
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Stuart H Orkin
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - David M Sabatini
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Whitehead Institute for Biomedical Research, Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, Massachusetts 02142, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, Massachusetts 02139, USA
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
59
|
Dingemanse C, Belzer C, van Hijum SAFT, Günthel M, Salvatori D, den Dunnen JT, Kuijper EJ, Devilee P, de Vos WM, van Ommen GB, Robanus-Maandag EC. Akkermansia muciniphila and Helicobacter typhlonius modulate intestinal tumor development in mice. Carcinogenesis 2015; 36:1388-96. [PMID: 26320104 DOI: 10.1093/carcin/bgv120] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 08/13/2015] [Indexed: 12/17/2022] Open
Abstract
Gastrointestinal tumor growth is thought to be promoted by gastrointestinal bacteria and their inflammatory products. We observed that intestine-specific conditional Apc mutant mice (FabplCre;Apc (15lox/+)) developed many more colorectal tumors under conventional than under pathogen-low housing conditions. Shotgun metagenomic sequencing plus quantitative PCR analysis of feces DNA revealed the presence of two bacterial species in conventional mice, absent from pathogen-low mice. One, Helicobacter typhlonius, has not been associated with cancer in man, nor in immune-competent mice. The other species, mucin-degrading Akkermansia muciniphila, is abundantly present in healthy humans, but reduced in patients with inflammatory gastrointestinal diseases and in obese and type 2 diabetic mice. Eradication of H.typhlonius in young conventional mice by antibiotics decreased the number of intestinal tumors. Additional presence of A.muciniphila prior to the antibiotic treatment reduced the tumor number even further. Colonization of pathogen-low FabplCre;Apc (15lox/+) mice with H.typhlonius or A.muciniphila increased the number of intestinal tumors, the thickness of the intestinal mucus layer and A.muciniphila colonization without H.typhlonius increased the density of mucin-producing goblet cells. However, dual colonization with H.typhlonius and A.muciniphila significantly reduced the number of intestinal tumors, the mucus layer thickness and goblet cell density to that of control mice. By global microbiota composition analysis, we found a positive association of A.muciniphila, and of H.typhlonius, and a negative association of unclassified Clostridiales with increased tumor burden. We conclude that A.muciniphila and H.typhlonius can modulate gut microbiota composition and intestinal tumor development in mice.
Collapse
Affiliation(s)
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University 6703 HB, Wageningen, The Netherlands
| | - Sacha A F T van Hijum
- Centre for Molecular and Biomolecular Informatics Bacterial Genomics, Radboud University Medical Centre 6525 GA, Nijmegen, The Netherlands, NIZO Food Research BV 6718 ZB, Ede, The Netherlands
| | | | | | | | - Ed J Kuijper
- Department of Medical Microbiology, Leiden University Medical Center 2300 RC, Leiden, The Netherlands and
| | | | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University 6703 HB, Wageningen, The Netherlands, Department of Veterinary Biosciences, University of Helsinki 00014, Helsinki, Finland
| | | | | |
Collapse
|
60
|
Golovko D, Kedrin D, Yilmaz ÖH, Roper J. Colorectal cancer models for novel drug discovery. Expert Opin Drug Discov 2015; 10:1217-29. [PMID: 26295972 DOI: 10.1517/17460441.2015.1079618] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Despite increased screening rates and advances in targeted therapy, colorectal cancer (CRC) remains the third leading cause of cancer-related mortality. CRC models that recapitulate key features of human disease are essential to the development of novel and effective therapeutics. Classic methods of modeling CRC such as human cell lines and xenograft mice, while useful for many applications, carry significant limitations. Recently developed in vitro and in vivo models overcome some of these deficiencies and thus can be utilized to better model CRC for mechanistic and translational research. AREAS COVERED The authors review established models of in vitro cell culture and describe advances in organoid culture for studying normal and malignant intestine. They also discuss key features of classic xenograft models and describe other approaches for in vivo CRC research, including patient-derived xenograft, carcinogen-induced, orthotopic transplantation and transgenic mouse models. We also describe mouse models of metastatic CRC. EXPERT OPINION No single model is optimal for drug discovery in CRC. Genetically engineered models overcome many limitations of xenograft models. Three-dimensional organoids can be efficiently derived from both normal and malignant tissue for large-scale in vitro and in vivo (transplantation) studies and are thus a significant advance in CRC drug discovery.
Collapse
Affiliation(s)
- Daniel Golovko
- a 1 Tufts Medical Center, Division of Gastroenterology and Molecular Oncology Research Institute , Boston, MA 02111, USA
| | - Dmitriy Kedrin
- b 2 MIT, The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology , Cambridge, MA 02139, USA.,c 3 Massachusetts General Hospital and Harvard Medical School, Division of Gastroenterology , Boston, MA 02114, USA
| | - Ömer H Yilmaz
- b 2 MIT, The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology , Cambridge, MA 02139, USA.,d 4 Massachusetts General Hospital and Harvard Medical School, Department of Pathology , Boston, MA 02114, USA
| | - Jatin Roper
- a 1 Tufts Medical Center, Division of Gastroenterology and Molecular Oncology Research Institute , Boston, MA 02111, USA .,b 2 MIT, The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology , Cambridge, MA 02139, USA
| |
Collapse
|
61
|
Zigmond E, Ya’acov AB, Lee H, Lichtenstein Y, Shalev Z, Smith Y, Zolotarov L, Ziv E, Kalman R, Le HV, Lu H, Silverman RB, Ilan Y. Suppression of Hepatocellular Carcinoma by Inhibition of Overexpressed Ornithine Aminotransferase. ACS Med Chem Lett 2015; 6:840-4. [PMID: 26288681 DOI: 10.1021/acsmedchemlett.5b00153] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 05/29/2015] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma is the second leading cause of cancer death worldwide. DNA microarray analysis identified the ornithine aminotransferase (OAT) gene as a prominent gene overexpressed in hepatocellular carcinoma (HCC) from Psammomys obesus. In vitro studies demonstrated inactivation of OAT by gabaculine (1), a neurotoxic natural product, which suppressed in vitro proliferation of two HCC cell lines. Alpha-fetoprotein (AFP) secretion, a biomarker for HCC, was suppressed by gabaculine in both cell lines, but not significantly. Because of the active site similarity between GABA aminotransferase (GABA-AT) and OAT, a library of 24 GABA-AT inhibitors was screened to identify a more selective inhibitor of OAT. (1S,3S)-3-Amino-4-(hexafluoropropan-2-ylidene)cyclopentane-1-carboxylic acid (2) was found to be an inactivator of OAT that only weakly inhibits GABA-AT, l-aspartate aminotransferase, and l-alanine aminotransferase. In vitro administration of 2 significantly suppressed AFP secretion in both Hep3B and HepG2 HCC cells; in vivo, 2 significantly suppressed AFP serum levels and tumor growth in HCC-harboring mice, even at 0.1 mg/kg. Overexpression of the OAT gene in HCC and the ability to block the growth of HCC by OAT inhibitors support the role of OAT as a potential therapeutic target to inhibit HCC growth. This is the first demonstration of suppression of HCC by an OAT inactivator.
Collapse
Affiliation(s)
| | | | - Hyunbeom Lee
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | | | | | | | | | | | | | - Hoang V. Le
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Hejun Lu
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Richard B. Silverman
- Department of Chemistry,
Department of Molecular Biosciences, Chemistry of Life Processes Institute,
and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | | |
Collapse
|
62
|
Lévy J, Cacheux W, Bara MA, L'Hermitte A, Lepage P, Fraudeau M, Trentesaux C, Lemarchand J, Durand A, Crain AM, Marchiol C, Renault G, Dumont F, Letourneur F, Delacre M, Schmitt A, Terris B, Perret C, Chamaillard M, Couty JP, Romagnolo B. Intestinal inhibition of Atg7 prevents tumour initiation through a microbiome-influenced immune response and suppresses tumour growth. Nat Cell Biol 2015; 17:1062-73. [PMID: 26214133 DOI: 10.1038/ncb3206] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/18/2015] [Indexed: 02/07/2023]
Abstract
Here, we show that autophagy is activated in the intestinal epithelium in murine and human colorectal cancer and that the conditional inactivation of Atg7 in intestinal epithelial cells inhibits the formation of pre-cancerous lesions in Apc(+/-) mice by enhancing anti-tumour responses. The antibody-mediated depletion of CD8(+) T cells showed that these cells are essential for the anti-tumoral responses mediated by the inhibition of autophagy. We show that Atg7 deficiency leads to intestinal dysbiosis and that the microbiota is required for anticancer responses. In addition, Atg7 deficiency resulted in a stress response accompanied by metabolic defects, AMPK activation and p53-mediated cell-cycle arrest in tumour cells but not in normal tissue. This study reveals that the inhibition of autophagy within the epithelium may prevent the development and progression of colorectal cancer in genetically predisposed patients.
Collapse
Affiliation(s)
- Jonathan Lévy
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Wulfran Cacheux
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France [3] Department of Medical Oncology, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France [4] Pharmacogenomics Unit, Department of Genetics, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Medhi Ait Bara
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Antoine L'Hermitte
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Patricia Lepage
- 1] Institut National de la Recherche Agronomique, Micalis UMR1319, Jouy-en-Josas 78352, France [2] AgroParisTech, Micalis UMR1319, 78350 Jouy-en-Josas, France
| | - Marie Fraudeau
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Coralie Trentesaux
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Julie Lemarchand
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Aurélie Durand
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Anne-Marie Crain
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France [3] Université Paris Diderot, UFR Sciences du Vivant, Sorbonne Paris Cité, Paris 75013, France
| | - Carmen Marchiol
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Gilles Renault
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Florent Dumont
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Franck Letourneur
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Myriam Delacre
- 1] Université Lille Nord de France, Lille 59000, France [2] Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille 59800, France [3] Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Lille 59046, France [4] Institut National de la Santé et de la Recherche Médicale, Lille 59045, France
| | - Alain Schmitt
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Benoit Terris
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France [3] Service d'Anatomie et Cytologie Pathologiques, AP-HP, Hôpital Cochin, Université Paris Descartes, Paris 75014, France
| | - Christine Perret
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| | - Mathias Chamaillard
- 1] Université Lille Nord de France, Lille 59000, France [2] Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille 59800, France [3] Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Lille 59046, France [4] Institut National de la Santé et de la Recherche Médicale, Lille 59045, France
| | - Jean-Pierre Couty
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France [3] Université Paris Diderot, UFR Sciences du Vivant, Sorbonne Paris Cité, Paris 75013, France
| | - Béatrice Romagnolo
- 1] Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS), UMR8104, Paris 75014, France [2] Institut National de la Sante et de la Recherche Médicale (INSERM), U1016, Paris 75014, France
| |
Collapse
|
63
|
Cai J, Maitra A, Anders RA, Taketo MM, Pan D. β-Catenin destruction complex-independent regulation of Hippo-YAP signaling by APC in intestinal tumorigenesis. Genes Dev 2015; 29:1493-506. [PMID: 26193883 PMCID: PMC4526734 DOI: 10.1101/gad.264515.115] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/19/2015] [Indexed: 01/16/2023]
Abstract
Cai et al. show that, besides its role in Wnt–β-catenin signaling, APC functions as a scaffold protein that facilitates the Hippo kinase cascade by interacting with Sav1 and Lats1. They also find that YAP is absolutely required for the development of APC-deficient adenomas. Mutations in Adenomatous polyposis coli (APC) underlie familial adenomatous polyposis (FAP), an inherited cancer syndrome characterized by the widespread development of colorectal polyps. APC is best known as a scaffold protein in the β-catenin destruction complex, whose activity is antagonized by canonical Wnt signaling. Whether other effector pathways mediate APC's tumor suppressor function is less clear. Here we report that activation of YAP, the downstream effector of the Hippo signaling pathway, is a general hallmark of tubular adenomas from FAP patients. We show that APC functions as a scaffold protein that facilitates the Hippo kinase cascade by interacting with Sav1 and Lats1. Consistent with the molecular link between APC and the Hippo signaling pathway, genetic analysis reveals that YAP is absolutely required for the development of APC-deficient adenomas. These findings establish Hippo–YAP signaling as a critical effector pathway downstream from APC, independent from its involvement in the β-catenin destruction complex.
Collapse
Affiliation(s)
- Jing Cai
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Anirban Maitra
- Sheikh Ahmed Bin Zayed Al Nahyan Center for Pancreatic Cancer Research, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Robert A Anders
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Makoto M Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Yoshida-Konoé-cho, Sakyo, Kyoto 606-8501, Japan
| | - Duojia Pan
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
64
|
Giardina C, Nakanishi M, Khan A, Kuratnik A, Xu W, Brenner B, Rosenberg DW. Regulation of VDR Expression in Apc-Mutant Mice, Human Colon Cancers and Adenomas. Cancer Prev Res (Phila) 2015; 8:387-99. [PMID: 25873367 DOI: 10.1158/1940-6207.capr-14-0371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/20/2015] [Indexed: 02/06/2023]
Abstract
One variable that may affect the ability of vitamin D to reduce colon cancer risk is the expression of its high-affinity receptor, VDR. Here, we show that vitamin D does not reduce tumor formation in Apc(Δ14/+) mice and that VDR expression is lost in the majority of the colon tumor cells. The extent of VDR loss corresponded inversely to the level of β-catenin nuclear localization and could be observed in early lesions composed of just a few crypts. Analysis of reported VDR regulators showed that the repressing class I histone deacetylases (HDAC) were significantly elevated in the tumors (up to 4-fold), whereas the VDR-activating retinoid X receptors (RXR) were downregulated (∼50%). Expression of the Slug repressor was also increased, but was found primarily in stromal cells. Analysis of epigenetically active compounds on colon cell lines and intestinal organoids showed that HDAC inhibitors were particularly adept at stimulating VDR expression. Treatment of tumor-bearing Apc(Δ14/+) mice with the HDAC inhibitor panobinostat increased VDR expression in the tumors and normal mucosa. The RXR agonist bexarotene failed to activate VDR expression, indicating that RXR ligands were not limiting. Analysis of human microarray data indicated that VDR mRNA is frequently downregulated in colon adenomas, which correlated positively with RXRA expression and inversely with HDAC 2 and 8 expression. Human adenomas showed variable VDR protein expression levels, both between and within individual lesions. Determining the mechanisms of VDR regulation in colon neoplasms may significantly enhance our ability to use vitamin D as a cancer prevention agent.
Collapse
Affiliation(s)
- Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut.
| | - Masako Nakanishi
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Awaad Khan
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Anton Kuratnik
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Wanli Xu
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Bruce Brenner
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Daniel W Rosenberg
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
65
|
Nakanishi M, Perret C, Meuillet EJ, Rosenberg DW. Non-cell autonomous effects of targeting inducible PGE2 synthesis during inflammation-associated colon carcinogenesis. Carcinogenesis 2015; 36:478-86. [PMID: 25634334 DOI: 10.1093/carcin/bgv004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/14/2015] [Indexed: 12/21/2022] Open
Abstract
Microsomal PGE2 synthase-1 (mPGES-1), the terminal enzyme in the formation of inducible PGE2, represents a potential target for cancer chemoprevention. We have previously shown that genetic abrogation of mPGES-1 significantly suppresses tumorigenesis in two preclinical models of intestinal cancer. In this study, we examined the role of mPGES-1 during colon tumorigenesis in the presence of dextran sulfate sodium (DSS)-induced inflammatory microenvironment. Using Apc (Δ14/+) in which the mPGES-1 gene is either wild-type (D14:WT) or deleted (D14:KO), we report that mPGES-1 deficiency enhances sensitivity to acute mucosal injury. As a result of the increased epithelial damage, protection against adenoma formation is unexpectedly compromised in the D14:KO mice. Examining the DSS-induced acute injury, cryptal structures are formed within inflamed areas of colonic mucosa of both genotypes that display the hallmarks of early neoplasia. When acute epithelial injury is balanced by titration of DSS exposures, however, these small cryptal lesions progress rapidly to adenomas in the D14:WT mice. Given that mPGES-1 is highly expressed within the intestinal stroma under the inflammatory conditions of DSS-induced ulceration, we propose a complex and dual role for inducible PGE2 synthesis within the colonic mucosa. Our data suggest that inducible PGE2 is critical for the maintenance of an intact colonic epithelial barrier, while promoting epithelial regeneration. This function is exploited during neoplastic transformation in Apc (Δ14/+) mice as PGE2 contributes to the growth and expansion of the early initiated cryptal structures. Taken together, inducible PGE2 plays a complex role in inflammation-associated cancers that requires further analysis. Inducible PGE2 production by mPGES-1 is critical for the colonic mucosal homeostasis. This function is exploited in the presence of the neoplastic transformation in Apc (Δ14/+) mice as PGE2 contributes to the growth and expansion of the early cryptal structures.
Collapse
Affiliation(s)
- Masako Nakanishi
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, CT, USA, Inserm, U1016, département endocrinologie métabolisme et cancer, Institut Cochin, Paris, France and The University of Arizona Cancer Center, Department of Molecular and Cell Biology, Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA. Tucson, AZ, USA
| | - Christine Perret
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, CT, USA, Inserm, U1016, département endocrinologie métabolisme et cancer, Institut Cochin, Paris, France and
| | - Emmanuelle J Meuillet
- The University of Arizona Cancer Center, Department of Molecular and Cell Biology, Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA. Tucson, AZ, USA
| | - Daniel W Rosenberg
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, CT, USA, Inserm, U1016, département endocrinologie métabolisme et cancer, Institut Cochin, Paris, France and The University of Arizona Cancer Center, Department of Molecular and Cell Biology, Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA. Tucson, AZ, USA
| |
Collapse
|
66
|
Funato Y, Yamazaki D, Mizukami S, Du L, Kikuchi K, Miki H. Membrane protein CNNM4-dependent Mg2+ efflux suppresses tumor progression. J Clin Invest 2014; 124:5398-410. [PMID: 25347473 DOI: 10.1172/jci76614] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/25/2014] [Indexed: 12/25/2022] Open
Abstract
Intracellular Mg(2+) levels are strictly regulated; however, the biological importance of intracellular Mg(2+) levels and the pathways that regulate them remain poorly understood. Here, we determined that intracellular Mg(2+) is important in regulating both energy metabolism and tumor progression. We determined that CNNM4, a membrane protein that stimulates Mg(2+) efflux, binds phosphatase of regenerating liver (PRL), which is frequently overexpressed in malignant human cancers. Biochemical analyses of cultured cells revealed that PRL prevents CNNM4-dependent Mg(2+) efflux and that regulation of intracellular Mg(2+) levels by PRL and CNNM4 is linked to energy metabolism and AMPK/mTOR signaling. Indeed, treatment with the clinically available mTOR inhibitor rapamycin suppressed the growth of cancer cells in which PRL was overexpressed. In ApcΔ(14/+) mice, which spontaneously form benign polyps in the intestine, deletion of Cnnm4 promoted malignant progression of intestinal polyps to adenocarcinomas. IHC analyses of tissues from patients with colon cancer demonstrated an inverse relationship between CNNM4 expression and colon cancer malignancy. Together, these results indicate that CNNM4-dependent Mg(2+) efflux suppresses tumor progression by regulating energy metabolism.
Collapse
|
67
|
Femia AP, Luceri C, Soares PV, Lodovici M, Caderni G. Multiple mucin depleted foci, high proliferation and low apoptotic response in the onset of colon carcinogenesis of the PIRC rat, mutated inApc. Int J Cancer 2014; 136:E488-95. [DOI: 10.1002/ijc.29232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/26/2014] [Accepted: 09/17/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Angelo Pietro Femia
- Section of Pharmacology and Toxicology; NEUROFARBA Department, University of Florence; 6 Viale Pieraccini 50139 Florence Italy
| | - Cristina Luceri
- Section of Pharmacology and Toxicology; NEUROFARBA Department, University of Florence; 6 Viale Pieraccini 50139 Florence Italy
| | - Paulo Victoria Soares
- Department of Pathology and Legal Medicine; Faculty of Medicine of Ribeirão Preto; University of São Paulo Brasil
| | - Maura Lodovici
- Section of Pharmacology and Toxicology; NEUROFARBA Department, University of Florence; 6 Viale Pieraccini 50139 Florence Italy
| | - Giovanna Caderni
- Section of Pharmacology and Toxicology; NEUROFARBA Department, University of Florence; 6 Viale Pieraccini 50139 Florence Italy
| |
Collapse
|
68
|
Fleet JC. Animal models of gastrointestinal and liver diseases. New mouse models for studying dietary prevention of colorectal cancer. Am J Physiol Gastrointest Liver Physiol 2014; 307:G249-59. [PMID: 24875098 PMCID: PMC4121636 DOI: 10.1152/ajpgi.00019.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Colorectal cancer is a heterogeneous disease that is one of the major causes of cancer death in the U.S. There is evidence that lifestyle factors like diet can modulate the course of this disease. Demonstrating the benefit and mechanism of action of dietary interventions against colon cancer will require studies in preclinical models. Many mouse models have been developed to study colon cancer but no single model can reflect all types of colon cancer in terms of molecular etiology. In addition, many models develop only low-grade cancers and are confounded by development of the disease outside of the colon. This review will discuss how mice can be used to model human colon cancer and it will describe a variety of new mouse models that develop colon-restricted cancer as well as more advanced phenotypes for studies of late-state disease.
Collapse
Affiliation(s)
- James C. Fleet
- 1Department of Nutrition Science, Purdue University, West Lafayette, Indiana; and ,2Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|
69
|
Powell AE, Vlacich G, Zhao ZY, McKinley ET, Washington MK, Manning HC, Coffey RJ. Inducible loss of one Apc allele in Lrig1-expressing progenitor cells results in multiple distal colonic tumors with features of familial adenomatous polyposis. Am J Physiol Gastrointest Liver Physiol 2014; 307:G16-23. [PMID: 24833705 PMCID: PMC4080164 DOI: 10.1152/ajpgi.00358.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Individuals with familial adenomatous polyposis (FAP) harbor a germline mutation in adenomatous polyposis coli (APC). The major clinical manifestation is development of multiple colonic tumors at a young age due to stochastic loss of the remaining APC allele. Extracolonic features, including periampullary tumors, gastric abnormalities, and congenital hypertrophy of the retinal pigment epithelium, may occur. The objective of this study was to develop a mouse model that simulates these features of FAP. We combined our Lrig1-CreERT2/+ mice with Apcfl/+ mice, eliminated one copy of Apc in leucine-rich repeats and immunoglobulin-like domains protein 1 (Lrig1)-positive (Lrig1(+)) progenitor cells with tamoxifen injection, and monitored tumor formation in the colon by colonoscopy and PET. Initial loss of one Apc allele in Lrig1(+) cells results in a predictable pattern of preneoplastic changes, culminating in multiple distal colonic tumors within 50 days of induction, as well as the extracolonic manifestations of FAP mentioned above. We show that tumor formation can be monitored by noninvasive PET imaging. This inducible stem cell-driven model recapitulates features of FAP and offers a tractable platform on which therapeutic interventions can be monitored over time by colonoscopy and noninvasive imaging.
Collapse
Affiliation(s)
- Anne E. Powell
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Gregory Vlacich
- 2Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Zhen-Yang Zhao
- 3Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Eliot T. McKinley
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - M. Kay Washington
- 4Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - H. Charles Manning
- 5Vanderbilt Institute for Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Robert J. Coffey
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; ,6Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
70
|
Mutoh H, Sashikawa M, Sakamoto H, Tateno T. Cyclooxygenase 2 in gastric carcinoma is expressed in doublecortin- and CaM kinase-like-1-positive tuft cells. Gut Liver 2014; 8:508-18. [PMID: 25228975 PMCID: PMC4164254 DOI: 10.5009/gnl13237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 09/04/2013] [Accepted: 09/05/2013] [Indexed: 12/13/2022] Open
Abstract
Background/Aims Doublecortin and CaM kinase-like-1 (DCAMKL1) is a marker of stem cells expressed predominantly in the crypt base in the intestine. However, DCAMKL1-positive cells have been shown to be differentiated tuft cells rather than quiescent progenitors. Tuft cells are the only epithelial cells that express cyclooxygenase 2 (COX-2) in the normal intestinal epithelium. We previously generated Cdx2-transgenic mice as model mice for intestinal metaplasia and gastric carcinoma. In the current study, we investigated the association between COX-2 and DCAMKL1 in gastric carcinoma. Methods We examined the association between COX-2 and DCAMKL1 expression in gastric carcinomas in clinical samples (early gastric well-differentiated adenocarcinoma) and Cdx2-transgenic mice; and the DCAMKL1-transgenic mouse stomach using immunohistochemistry and quantitative real-time polymerase chain reaction. Results The COX-2-expressing cells were scattered, not diffusely expressed, in gastric carcinomas from humans and Cdx2-transgenic mice. DCAMKL1-positive cells were also scattered in the gastric carcinomas, indicating that tuft cells could still be present in gastric carcinoma. COX-2 was expressed in DCAMKL1-positive tuft cells in Cdx2- and DCAMKL1-transgenic mouse stomachs, whereas the Sox9 transcription factor was ubiquitously expressed in gastric carcinomas, including COX-2-positive cells. Conclusions COX-2 is expressed in DCAMKL1-expressing quiescent tuft cells in gastric carcinoma.
Collapse
Affiliation(s)
- Hiroyuki Mutoh
- Division of Gastroenterology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Miho Sashikawa
- Division of Gastroenterology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Hirotsugu Sakamoto
- Division of Gastroenterology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tomoko Tateno
- Division of Gastroenterology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
71
|
Lapierre M, Bonnet S, Bascoul-Mollevi C, Ait-Arsa I, Jalaguier S, Del Rio M, Plateroti M, Roepman P, Ychou M, Pannequin J, Hollande F, Parker M, Cavailles V. RIP140 increases APC expression and controls intestinal homeostasis and tumorigenesis. J Clin Invest 2014; 124:1899-913. [PMID: 24667635 DOI: 10.1172/jci65178] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 01/23/2014] [Indexed: 12/14/2022] Open
Abstract
Deregulation of the Wnt/APC/β-catenin signaling pathway is an important consequence of tumor suppressor APC dysfunction. Genetic and molecular data have established that disruption of this pathway contributes to the development of colorectal cancer. Here, we demonstrate that the transcriptional coregulator RIP140 regulates intestinal homeostasis and tumorigenesis. Using Rip140-null mice and mice overexpressing human RIP140, we found that RIP140 inhibited intestinal epithelial cell proliferation and apoptosis. Interestingly, following whole-body irradiation, mice lacking RIP140 exhibited improved regenerative capacity in the intestine, while mice overexpressing RIP140 displayed reduced recovery. Enhanced RIP140 expression strongly repressed human colon cancer cell proliferation in vitro and after grafting onto nude mice. Moreover, in murine tissues and human cancer cells, RIP140 stimulated APC transcription and inhibited β-catenin activation and target gene expression. Finally, RIP140 mRNA and RIP140 protein levels were decreased in human colon cancers compared with those in normal mucosal tissue, and low levels of RIP140 expression in adenocarcinomas from patients correlated with poor prognosis. Together, these results support a tumor suppressor role for RIP140 in colon cancer.
Collapse
|
72
|
Abstract
Colorectal cancer is a heterogeneous disease that afflicts a large number of people in the USA. The use of animal models has the potential to increase our understanding of carcinogenesis, tumor biology, and the impact of specific molecular events on colon biology. In addition, animal models with features of specific human colorectal cancers can be used to test strategies for cancer prevention and treatment. In this review, we provide an overview of the mechanisms driving human cancer, we discuss the approaches one can take to model colon cancer in animals, and we describe a number of specific animal models that have been developed for the study of colon cancer. We believe that there are many valuable animal models to study various aspects of human colorectal cancer. However, opportunities for improving upon these models exist.
Collapse
|
73
|
Li L, Sun R, Liang Y, Pan X, Li Z, Bai P, Zeng X, Zhang D, Zhang L, Gao L. Association between polymorphisms in long non-coding RNA PRNCR1 in 8q24 and risk of colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:104. [PMID: 24330491 PMCID: PMC4029281 DOI: 10.1186/1756-9966-32-104] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 12/05/2013] [Indexed: 01/12/2023]
Abstract
BACKGROUND Genome-wide association studies have identified that genetic variants in 8q24 confer susceptibility to colorectal cancer (CRC). Recently, a novel lncRNA (PRNCR1) that located in the 8q24 was discovered. Single nucleotide polymorphisms (SNPs) in the lncRNAs may influence the process of splicing and stability of mRNA conformation, resulting in the modification of its interacting partners. We hypothesized that SNPs in the lncRNA PRNCR1 may be related to the risk of CRC. METHODS We conducted a case-control study and genotyped five tag SNPs in the lncRNA PRNCR1 in 908 subjects including 313 cases with CRC and 595 control subjects using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assay. RESULTS In overall analyses, we found that the rs13252298 and rs1456315 were associated with significantly decreased risks of CRC. In stratification analyses, we found that CRC patients carrying the rs1456315G were likely to have a tumor size of greater than 5 cm (G vs. A: adjusted OR = 1.56, 95% CI: 1.10-2.23). Additionally, patients with the rs7007694C and rs16901946G had decreased risks to develop poorly differentiated CRC, whereas patients with the rs1456315G had an increased risk to develop poorly differentiated CRC. CONCLUSION These findings suggest that SNPs in the lncRNA PRNCR1 may contribute to susceptibility to CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lin Zhang
- Laboratory of Molecular and Translational Medicine, West China Institute of Women and Children's Health; Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P,R, China.
| | | |
Collapse
|
74
|
Bmi1 is required for tumorigenesis in a mouse model of intestinal cancer. Oncogene 2013; 33:3742-7. [PMID: 23955081 DOI: 10.1038/onc.2013.333] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 06/21/2013] [Accepted: 06/24/2013] [Indexed: 12/19/2022]
Abstract
The epigenetic regulator BMI1 is upregulated progressively in a wide variety of human tumors including colorectal cancer. In this study, we assessed the requirement for Bmi1 in intestinal tumorigenesis using an autochthonous mouse model in which Apc was conditionally ablated in the intestinal epithelium. Germline mutation of Bmi1 significantly reduced both the number and size of small intestinal adenomas arising in this model, and it acted in a dose-dependent manner. Moreover, in contrast to wild-type controls, Bmi1(-/-) mice showed no increase in median tumor size, and a dramatic decrease in tumor number, between 3 and 4 months of age. Thus, Bmi1 is required for both progression and maintenance of small intestinal adenomas. Importantly, Bmi1 deficiency did not disrupt oncogenic events arising from Apc inactivation. Instead, the Arf tumor suppressor, a known target of Bmi1 epigenetic silencing, was upregulated in Bmi1 mutant tumors. This was accompanied by significant upregulation of p53, which was confirmed by sequencing to be wild-type, and also elevated apoptosis within the smallest Bmi1(-/-) adenomas. By crossing Arf into this cancer model, we showed that Arf is required for the induction of both p53 and apoptosis, and it is a key determinant of the ability of Bmi1 deficiency to suppress intestinal tumorigenesis. Finally, a conditional Bmi1 mutant strain was generated and used to determine the consequences of deleting Bmi1 specifically within the intestinal epithelium. Strikingly, intestinal-specific Bmi1 deletion suppressed small intestinal adenomas in a manner that was indistinguishable from germline Bmi1 deletion. Thus, we conclude that Bmi1 deficiency impairs the progression and maintenance of small intestinal tumors in a cell autonomous and highly Arf-dependent manner.
Collapse
|
75
|
Toki H, Inoue M, Motegi H, Minowa O, Kanda H, Yamamoto N, Ikeda A, Karashima Y, Matsui J, Kaneda H, Miura I, Suzuki T, Wakana S, Masuya H, Gondo Y, Shiroishi T, Akiyama T, Yao R, Noda T. Novel mouse model for Gardner syndrome generated by a large-scale N-ethyl-N-nitrosourea mutagenesis program. Cancer Sci 2013; 104:937-44. [PMID: 23551873 PMCID: PMC7657124 DOI: 10.1111/cas.12161] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Revised: 03/21/2013] [Accepted: 03/27/2013] [Indexed: 12/26/2022] Open
Abstract
Mutant mouse models are indispensable tools for clarifying the functions of genes and elucidating the underlying pathogenic mechanisms of human diseases. We carried out large-scale mutagenesis using the chemical mutagen N-ethyl-N-nitrosourea. One specific aim of our mutagenesis project was to generate novel cancer models. We screened 7012 animals for dominant traits using a necropsy test and thereby established 17 mutant lines predisposed to cancer. Here, we report on a novel cancer model line that developed osteoma, trichogenic tumor, and breast cancer. Using fine mapping and genomic sequencing, we identified a point mutation in the adenomatous polyposis coli (Apc) gene. The Apc1576 mutants bear a nonsense mutation at codon 1576 in the Apc gene. Although most Apc mutant mice established thus far have multifocal intestinal tumors, mice that are heterozygous for the Apc1576 mutation do not develop intestinal tumors; instead, they develop multifocal breast cancers and trichogenic tumors. Notably, the osteomas that develop in the Apc1576 mutant mice recapitulate the lesion observed in Gardner syndrome, a clinical variant of familial adenomatous polyposis. Our Apc1576 mutant mice will be valuable not only for understanding the function of the Apc gene in detail but also as models of human Gardner syndrome.
Collapse
Affiliation(s)
- Hideaki Toki
- Team for Advanced Development and Evaluation of Human Disease Models, Riken BioResource Center, Tsukuba, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Liu KH, Huynh N, Patel O, Shulkes A, Baldwin G, He H. P21-activated kinase 1 promotes colorectal cancer survival by up-regulation of hypoxia-inducible factor-1α. Cancer Lett 2013; 340:22-9. [PMID: 23811286 DOI: 10.1016/j.canlet.2013.06.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 06/18/2013] [Accepted: 06/20/2013] [Indexed: 12/16/2022]
Abstract
P21 activated kinase 1 (PAK1) enhances colorectal cancer (CRC) progression by stimulating Wnt/β-catenin and Ras oncogene, which promote CRC survival via stimulation of hypoxia-inducible factor 1α (HIF-1α). The aim of this study was to assess the mechanism involved in the stimulation by PAK1 of CRC survival. PAK1 promoted CRC cell survival by up-regulation of HIF-1α. PAK1 was over-expressed and hyper-activated in tumors of ApcΔ(14/+) mice, which was correlated with over-expression of HIF-1α and β-catenin. Inhibition of PAK1 decreased tumor growth and the expression of HIF-1α and β-catenin in tumors of ApcΔ(14/+) mice, and suppressed xenograft tumor survival in SCID mice. These findings indicate that PAK1 stimulates CRC survival by up-regulation of HIF-1α.
Collapse
Affiliation(s)
- Kevin H Liu
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | | | | | | | | | | |
Collapse
|
77
|
Inoue K, Fry EA, Taneja P. Recent progress in mouse models for tumor suppressor genes and its implications in human cancer. Clin Med Insights Oncol 2013; 7:103-22. [PMID: 23843721 PMCID: PMC3682694 DOI: 10.4137/cmo.s10358] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Gain-of-function mutations in oncogenes and loss-of-function mutations in tumor suppressor genes (TSG) lead to cancer. In most human cancers, these mutations occur in somatic tissues. However, hereditary forms of cancer exist for which individuals are heterozygous for a germline mutation in a TSG locus at birth. The second allele is frequently inactivated by gene deletion, point mutation, or promoter methylation in classical TSGs that meet Knudson's two-hit hypothesis. Conversely, the second allele remains as wild-type, even in tumors in which the gene is haplo-insufficient for tumor suppression. This article highlights the importance of PTEN, APC, and other tumor suppressors for counteracting aberrant PI3K, β-catenin, and other oncogenic signaling pathways. We discuss the use of gene-engineered mouse models (GEMM) of human cancer focusing on Pten and Apc knockout mice that recapitulate key genetic events involved in initiation and progression of human neoplasia. Finally, the therapeutic potential of targeting these tumor suppressor and oncogene signaling networks is discussed.
Collapse
Affiliation(s)
- Kazushi Inoue
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Elizabeth A. Fry
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Pankaj Taneja
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|
78
|
Dionne S, Elimrani I, Roy MJ, Qureshi IA, Sarma DR, Levy E, Seidman EG. Studies on the chemopreventive effect of carnitine on tumorigenesis in vivo, using two experimental murine models of colon cancer. Nutr Cancer 2013; 64:1279-87. [PMID: 23163856 DOI: 10.1080/01635581.2012.722247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Carnitine is known for its essential role in intermediary metabolism. In vitro studies suggest that its antioxidant and anti-inflammatory properties are potentially beneficial toward cancer prevention. This study tested effects of carnitine on the development of colon cancer in vivo using 2 murine models: azoxymethane (AOM) treatment as a model of carcinogen-induced colon cancer and a genetically induced model using Apc (Min/+) mice. AOM and Apc (Min/+) mice divided into dietary groups varying in lipid content, with or without carnitine supplementation (0.08%). AOM-exposed mice on a high butterfat diet had significantly increased aberrant crypts (ACF) (9.3 ± 0.88 vs. 6.3 ± 0.65), and macroscopic tumors (3.8 ± 0.95 vs. 2.0 ± 0.25) compared to mice on a control diet. In AOM mice fed the high butterfat diet, carnitine supplementation inhibited ACF (4.9 ± 0.7 vs. 9.3 ± 0.88, P < 0.001), crypt multiciplicity (1.6 ± 0.08 vs. 1.92 ± 0.1, P < 0.01) and tumors (1.5 ± 0.38 vs. 3.8 ± 0.95, P < 0.001). Carnitine supplementation resulted in significantly increased tissue carnitine and acylcarnitine levels. Carnitine inhibited the development of precancerous lesions and macroscopic colonic tumors in AOM-treated mice. However, carnitine did not exert protective effects on intestinal tumors in Apc (Min/+) mice.
Collapse
Affiliation(s)
- Serge Dionne
- Division of Gastroenterology, Research Institute, McGill University Health Center, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
Adenomatous polyposis coli (APC) is best known for its crucial role in colorectal cancer suppression. Rodent models with various Apc mutations have enabled experimental validation of different Apc functions in tumors and normal tissues. Since the development of the first mouse model with a germline Apc mutation in the early 1990s, 20 other Apc mouse and rat models have been generated. This article compares and contrasts currently available Apc rodent models with particular emphasis on providing potential explanations for their reported variation in three areas: (i) intestinal polyp multiplicity, (ii) intestinal polyp distribution, and (iii) extraintestinal phenotypes.
Collapse
Affiliation(s)
- Maged Zeineldin
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Ave. Lawrence, KS 66045 USA
| | - Kristi L. Neufeld
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Ave. Lawrence, KS 66045 USA
| |
Collapse
|
80
|
Abstract
Colorectal cancer is one of the most common malignancies in the world. Many mouse models have been developed to evaluate features of colorectal cancer in humans. These can be grouped into genetically-engineered, chemically-induced, and inoculated models. However, none recapitulates all of the characteristics of human colorectal cancer. It is critical to use a specific mouse model to address a particular research question. Here, we review commonly used mouse models for human colorectal cancer.
Collapse
Affiliation(s)
- Yunguang Tong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | |
Collapse
|
81
|
Luo M, Li Z, Wang W, Zeng Y, Liu Z, Qiu J. Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett 2013; 333:213-21. [PMID: 23354591 DOI: 10.1016/j.canlet.2013.01.033] [Citation(s) in RCA: 449] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/14/2013] [Accepted: 01/18/2013] [Indexed: 12/20/2022]
Abstract
lncRNA H19 is essential for human tumor growth. However, little is known about whether H19 regulates bladder cancer metastasis. Here we found that H19 levels are remarkably increased in bladder cancer tissues, and upregulated H19 promotes bladder cancer cell migration in vitro and in vivo. H19 is associated with enhancer of zeste homolog 2 (EZH2), and that this association results in Wnt/β-catenin activation and subsequent downregulation of E-cadherin. A significant negative correlation is also observed between H19 levels and E-cad levels in vivo. These data suggest that upregulated H19 enhances bladder cancer metastasis by associating with EZH2 and inhibiting E-cad expression.
Collapse
Affiliation(s)
- Ming Luo
- Department of Urology, The Tenth People's Hospital Affiliated to Tongji University, Shanghai 200072, China
| | | | | | | | | | | |
Collapse
|
82
|
Zeineldin M, Neufeld KL. More than two decades of Apc modeling in rodents. Biochim Biophys Acta Rev Cancer 2013; 1836:80-9. [PMID: 23333833 DOI: 10.1016/j.bbcan.2013.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/31/2012] [Accepted: 01/03/2013] [Indexed: 02/07/2023]
Abstract
Mutation of tumor suppressor gene adenomatous polyposis coli (APC) is an initiating step in most colon cancers. This review summarizes Apc models in mice and rats, with particular concentration on those most recently developed, phenotypic variation among different models, and genotype/phenotype correlations.
Collapse
Affiliation(s)
- Maged Zeineldin
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Ave., Lawrence, KS 66045, USA
| | | |
Collapse
|
83
|
Barry ER, Morikawa T, Butler BL, Shrestha K, de la Rosa R, Yan KS, Fuchs CS, Magness ST, Smits R, Ogino S, Kuo CJ, Camargo FD. Restriction of intestinal stem cell expansion and the regenerative response by YAP. Nature 2012. [PMID: 23178811 PMCID: PMC3536889 DOI: 10.1038/nature11693] [Citation(s) in RCA: 434] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A remarkable feature of regenerative processes is their ability to halt proliferation once an organ’s structure has been restored. The Wnt signaling pathway is the major driving force for homeostatic self-renewal and regeneration in the mammalian intestine. The mechanisms that counterbalance Wnt-driven proliferation are poorly understood. We demonstrate here that YAP, a protein known for its powerful growth-inducing and oncogenic properties1-2, has an unexpected growth-suppressive function restricting Wnt signals during intestinal regeneration. Transgenic expression of YAP reduces Wnt target gene expression and results in the rapid loss of intestinal crypts. In addition, loss of YAP results in Wnt hypersensitivity during regeneration, leading to hyperplasia, expansion of intestinal stem cells (ISCs) and niche cells, and formation of ectopic crypts and microadenomas. We find that cytoplasmic YAP restricts elevated Wnt signaling independently of the APC/Axin/GSK3β complex partly by limiting the activity of Dishevelled (DVL). DVL signals in the nucleus of ISCs and its forced expression leads to enhanced Wnt signaling in crypts. YAP dampens Wnt signals by restricting DVL nuclear translocation during regenerative growth. Finally, we provide evidence that YAP is silenced in a subset of highly aggressive and undifferentiated human colorectal carcinomas (CRC) and its expression can restrict the growth of CRC xenografts. Collectively, our work describes a novel mechanistic paradigm for how proliferative signals are counterbalanced in regenerating tissues. Additionally, our findings have important implications for the targeting of YAP in human malignancies.
Collapse
Affiliation(s)
- Evan R Barry
- Stem Cell Program and Department of Hematology/Oncology, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Nakanishi M, Ménoret A, Belinsky GS, Giardina C, Godman CA, Vella AT, Rosenberg DW. Utilizing endoscopic technology to reveal real-time proteomic alterations in response to chemoprevention. Proteomics Clin Appl 2012; 1:1660-6. [PMID: 21136663 DOI: 10.1002/prca.200700545] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer chemoprevention approaches use either pharmacological or dietary agents to impede, arrest or reverse the carcinogenic process. Although several agents have shown effectiveness against colon cancer, present intervention strategies provide only partial reduction. In this study, we utilized high-resolution endoscopy to obtain colon tumor biopsy specimens from Apc mutant mice before and after 2-wk sulindac intervention. To acquire information beyond genomics, proteome analysis using the ProteomeLab PF2D platform was implemented to generate 2-D protein expression maps from biopsies. Chromatograms produced common signature profiles between sulindac and nonsulindac treated samples, and contrasting profiles termed "fingerprints". We selected a double peak that appeared in tumor biopsies from sulindac-treated mice. Further analyses using MS sequencing identified this protein as histone H2B. The location of H2B in the 1(st) dimension strongly suggested PTM, consistent with identification of two oxidized methionines. While further studies on sulindac proteomic fingerprints are underway, this study demonstrates the feasibility and advantages of "real-time" proteomic analysis for obtaining information on biomarker discovery and drug activity that would not be revealed by a genetic assay. This approach should be broadly applicable for assessing lesion responsiveness in a wide range of translational and human clinical studies.
Collapse
Affiliation(s)
- Masako Nakanishi
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | | | | | | | |
Collapse
|
85
|
Wang P, Ren Z, Sun P. Overexpression of the long non-coding RNA MEG3 impairs in vitro glioma cell proliferation. J Cell Biochem 2012; 113:1868-74. [PMID: 22234798 DOI: 10.1002/jcb.24055] [Citation(s) in RCA: 281] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gliomas are the most common type of primary brain tumor in the central nervous system of adults. Maternally Expressed Gene 3 (MEG3) is an imprinted gene located at 14q32 that encodes a non-coding RNA (ncRNA) associated with tumorigenesis. However, little is known about whether and how MEG3 regulates glioma development. In the present study we assayed the expression of MEG3 in glioma tissue samples by real-time polymerase chain reaction assay, and defined the biological functions and target genes by CCK-8 assay, flow cytometry, and RNA immunoprecipitation. We first demonstrated that MEG3 expression was markedly decreased in glioma tissues compared with adjacent normal tissues. Moreover, ectopic expression of MEG3 inhibited cell proliferation and promoted cell apoptosis in U251 and U87 MG human glioma cell lines. We further verified that MEG3 was associated with p53 and that this association was required for p53 activation. These data suggest an important role of MEG3 in the molecular etiology of glioma and implicate the potential application of MEG3 in glioma therapy.
Collapse
Affiliation(s)
- Pengjun Wang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | | | | |
Collapse
|
86
|
Saandi T, Baraille F, Derbal-Wolfrom L, Cattin AL, Benahmed F, Martin E, Cardot P, Duclos B, Ribeiro A, Freund JN, Duluc I. Regulation of the tumor suppressor homeogene Cdx2 by HNF4α in intestinal cancer. Oncogene 2012; 32:3782-8. [PMID: 22986531 DOI: 10.1038/onc.2012.401] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 12/21/2022]
Abstract
The gut-specific homeotic transcription factor Cdx2 is a crucial regulator of intestinal development and homeostasis, which is downregulated in colorectal cancers (CRC) and exhibits a tumor suppressor function in the colon. We have previously established that several endodermal transcription factors, including HNF4α and GATA6, are involved in Cdx2 regulation in the normal gut. Here we have studied the role of HNF4α in the mechanism of deregulation of Cdx2 in colon cancers. Crossing Apc(Δ14/+) mice prone to spontaneous intestinal tumor development with pCdx2-9LacZ transgenic mice containing the LacZ reporter under the control of the 9.3-kb Cdx2 promoter showed that this promoter segment contains sequences recapitulating the decrease of Cdx2 expression in intestinal cancers. Immunohistochemistry revealed that HNF4α, unlike GATA6, exhibited a similar decrease to Cdx2 in genetic (Apc(min/+) and Apc(Δ14/+)) and chemically induced (Azoxymethane (AOM) treatment) models of intestinal tumors in mice. HNF4α and Cdx2 also exhibited a comparable deregulated pattern in human CRC. Correlated patterns were observed between HNF4α and Cdx2 in several experimental models of human colon cancer cell lines: xenografts in nude mice, wound healing and glucose starvation. Furthermore, Cdx2 decreased by knocking down HNF4α in human colon cancer cells using siRNA and in the colon of mice conditionally knocked out for the Hnf4α gene in the adult intestine (Hnf4α(f/f);VilCre(ERT2) mice). Finally, the conditionally knocked out mice Hnf4α(f/f);VilCre(ERT2) treated with the carcinogen AOM developed colorectal tumors earlier than wild-type mice, as previously reported for mice with a reduced Cdx2 expression. In conclusion, this study provides evidence that the downregulation of HNF4α is an important determinant of the reduced expression of the Cdx2 tumor suppressor gene in intestinal cancers. Consistently, similar to Cdx2, HNF4α exerts a tumor suppressor function in the colon in that its loss of function facilitates tumor progression.
Collapse
Affiliation(s)
- T Saandi
- Inserm, Unité 682, Strasbourg, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Roper J, Hung KE. Priceless GEMMs: genetically engineered mouse models for colorectal cancer drug development. Trends Pharmacol Sci 2012; 33:449-55. [DOI: 10.1016/j.tips.2012.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/11/2012] [Accepted: 05/02/2012] [Indexed: 12/13/2022]
|
88
|
Yang F, Bi J, Xue X, Zheng L, Zhi K, Hua J, Fang G. Up-regulated long non-coding RNA H19 contributes to proliferation of gastric cancer cells. FEBS J 2012; 279:3159-65. [PMID: 22776265 DOI: 10.1111/j.1742-4658.2012.08694.x] [Citation(s) in RCA: 373] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been shown to have important regulatory roles in cancer biology, and the lncRNA H19 is up-regulated in hypoxic stress and in some tumors. However, the contributions of H19 to gastric cancer remain largely unknown. In this study, we assayed the H19 expression level in gastric cancer tissues by real-time PCR, and defined the biological functions by flow cytometry and RNA immunoprecipitation. We demonstrated that H19 levels were markedly increased in gastric cancer cells and gastric cancer tissues compared with normal controls. Moreover, ectopic expression of H19 increased cell proliferation, whereas H19 siRNA treatment contributed to cell apoptosis in AGS cell line. We further verified that H19 was associated with p53, and that this association resulted in partial p53 inactivation. These data suggest an important role for H19 in the molecular etiology of gastric cancer and potential application of H19 in gastric cancer therapy.
Collapse
Affiliation(s)
- Feng Yang
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
89
|
Bellis J, Duluc I, Romagnolo B, Perret C, Faux MC, Dujardin D, Formstone C, Lightowler S, Ramsay RG, Freund JN, De Mey JR. The tumor suppressor Apc controls planar cell polarities central to gut homeostasis. ACTA ACUST UNITED AC 2012; 198:331-41. [PMID: 22851318 PMCID: PMC3413367 DOI: 10.1083/jcb.201204086] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Asymmetric stem cell divisions controlled by Apc in the intestinal crypt result in regulated, anisotropic movement of daughter cells away from the niche. The stem cells (SCs) at the bottom of intestinal crypts tightly contact niche-supporting cells and fuel the extraordinary tissue renewal of intestinal epithelia. Their fate is regulated stochastically by populational asymmetry, yet whether asymmetrical fate as a mode of SC division is relevant and whether the SC niche contains committed progenitors of the specialized cell types are under debate. We demonstrate spindle alignments and planar cell polarities, which form a novel functional unit that, in SCs, can yield daughter cell anisotropic movement away from niche-supporting cells. We propose that this contributes to SC homeostasis. Importantly, we demonstrate that some SC divisions are asymmetric with respect to cell fate and provide data suggesting that, in some SCs, mNumb displays asymmetric segregation. Some of these processes were altered in apparently normal crypts and microadenomas of mice carrying germline Apc mutations, shedding new light on the first stages of progression toward colorectal cancer.
Collapse
Affiliation(s)
- Julien Bellis
- Laboratoire de Biophotonique et Pharmacologie, Unité Mixte de Recherche 7213, Centre National de la Recherche Scientifique, 67401 Illkirch, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Hinkel I, Duluc I, Martin E, Guenot D, Freund JN, Gross I. Cdx2 controls expression of the protocadherin Mucdhl, an inhibitor of growth and β-catenin activity in colon cancer cells. Gastroenterology 2012; 142:875-885.e3. [PMID: 22202456 DOI: 10.1053/j.gastro.2011.12.037] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 11/28/2011] [Accepted: 12/09/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The intestine-specific homeobox transcription factor Cdx2 is an important determinant of intestinal identity in the embryonic endoderm and regulates the balance between proliferation and differentiation in the adult intestinal epithelium. Human colon tumors often lose Cdx2 expression, and heterozygous inactivation of Cdx2 in mice increases colon tumorigenesis. We sought to identify Cdx2 target genes to determine how it contributes to intestinal homeostasis. METHODS We used expression profiling analysis to identify genes that are regulated by Cdx2 in colon cancer cells lines. Regulation and function of a potential target gene were further investigated using various cell assays. RESULTS In colon cancer cell lines, Cdx2 directly regulated the transcription of the gene that encodes the protocadherin Mucdhl. Mucdhl localized to the apex of differentiated cells in the intestinal epithelium, and its expression was reduced in most human colon tumors. Overexpression of Mucdhl inhibited low-density proliferation of colon cancer cells and reduced tumor formation in nude mice. One isoform of Mucdhl interacted with β-catenin and inhibited its transcriptional activity. CONCLUSIONS The transcription factor Cdx2 activates expression of the protocadherin Mucdhl, which interacts with β-catenin and regulates activities of intestinal cells. Loss of Cdx2 expression in colon cancer cells might reduce expression of Mucdhl and thereby lead to tumor formation.
Collapse
|
91
|
Powell AE, Wang Y, Li Y, Poulin EJ, Means AL, Washington MK, Higginbotham JN, Juchheim A, Prasad N, Levy SE, Guo Y, Shyr Y, Aronow BJ, Haigis KM, Franklin JL, Coffey RJ. The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell 2012; 149:146-58. [PMID: 22464327 PMCID: PMC3563328 DOI: 10.1016/j.cell.2012.02.042] [Citation(s) in RCA: 543] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 01/12/2012] [Accepted: 02/01/2012] [Indexed: 12/20/2022]
Abstract
Lineage mapping has identified both proliferative and quiescent intestinal stem cells, but the molecular circuitry controlling stem cell quiescence is incompletely understood. By lineage mapping, we show Lrig1, a pan-ErbB inhibitor, marks predominately noncycling, long-lived stem cells that are located at the crypt base and that, upon injury, proliferate and divide to replenish damaged crypts. Transcriptome profiling of Lrig1(+) colonic stem cells differs markedly from the profiling of highly proliferative, Lgr5(+) colonic stem cells; genes upregulated in the Lrig1(+) population include those involved in cell cycle repression and response to oxidative damage. Loss of Apc in Lrig1(+) cells leads to intestinal adenomas, and genetic ablation of Lrig1 results in heightened ErbB1-3 expression and duodenal adenomas. These results shed light on the relationship between proliferative and quiescent intestinal stem cells and support a model in which intestinal stem cell quiescence is maintained by calibrated ErbB signaling with loss of a negative regulator predisposing to neoplasia.
Collapse
Affiliation(s)
- Anne E. Powell
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yang Wang
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yina Li
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Emily J. Poulin
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Anna L. Means
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mary K. Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James N. Higginbotham
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alwin Juchheim
- Molecular Pathology Unit, Massachusetts General Hospital and Department of Pathology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nripesh Prasad
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806
| | - Shawn E. Levy
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806
| | - Yan Guo
- Department of Biostatistics, Vanderbilt University, Nashville, TN, 37232
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University, Nashville, TN, 37232
| | - Bruce J. Aronow
- Departments of Biomedical Informatics and Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kevin M. Haigis
- Molecular Pathology Unit, Massachusetts General Hospital and Department of Pathology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jeffrey L. Franklin
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert J. Coffey
- Departments of Medicine and Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Medical Center, Nashville, TN 37232, USA
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
92
|
Essential requirement for β-arrestin2 in mouse intestinal tumors with elevated Wnt signaling. Proc Natl Acad Sci U S A 2012; 109:3047-52. [PMID: 22315403 DOI: 10.1073/pnas.1109457109] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
β-Arrestins (Arrb) participate in the regulation of multiple signaling pathways, including Wnt/β-catenin, the major actor in human colorectal cancer initiation. To better understand the roles of Arrb in intestinal tumorigenesis, a reverse genetic approach (Arrb(-/-)) and in vivo siRNA treatment were used in Apc(Δ14/+) mice. Mice with Arrb2 depletion (knockout and siRNA) developed only 33% of the tumors detected in their Arrb2-WT littermates, whereas Arrb1 depletion remained without significant effect. These remaining tumors grow normally and are essentially Arrb2-independent. Unsupervised hierarchical clustering analysis showed that they clustered with 25% of Apc(Δ14/+);Arrb2(+/+) tumors. Genes overexpressed in this subset reflect a high interaction with the immune system, whereas those overexpressed in Arrb2-dependent tumors are predominantly involved in Wnt signaling, cell adhesion, migration, and extracellular matrix remodeling. The involvement of Arrb2 in intestinal tumor development via the regulation of the Wnt pathway is supported by ex vivo and in vitro experiments using either tumors from Apc(Δ14/+) mice or murine Apc(Min/+) cells. Indeed, Arrb2 siRNAs decreased the expression of Wnt target genes in cells isolated from 12 of 18 tumors from Apc(Δ14/+) mice. In Apc(Min/+) cells, Arrb2 siRNAs completely reversed the increased Wnt activity and colony formation in soft agar induced by Apc siRNA treatment, whereas they did not affect these parameters in basal conditions or in cells expressing constitutively active β-catenin. We demonstrate that Arrb2 is essential for the initiation and growth of intestinal tumors displaying elevated Wnt pathway activity and identify a previously unsuspected molecular heterogeneity among tumors induced by truncating Apc mutations.
Collapse
|
93
|
Ellatif SKA, Gutschner T, Diederichs S. Long Noncoding RNA Function and Expression in Cancer. REGULATORY RNAS 2012:197-226. [DOI: 10.1007/978-3-642-22517-8_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
94
|
Ellatif SKA, Gutschner T, Diederichs S. Long Noncoding RNA Function and Expression in Cancer. REGULATORY RNAS 2012:197-226. [DOI: 10.1007/978-3-662-45801-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
95
|
Lewis A, Davis H, Deheragoda M, Pollard P, Nye E, Jeffery R, Segditsas S, East P, Poulsom R, Stamp G, Wright N, Tomlinson I. The C-terminus of Apc does not influence intestinal adenoma development or progression. J Pathol 2011; 226:73-83. [PMID: 22009253 DOI: 10.1002/path.2972] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2011] [Accepted: 07/18/2011] [Indexed: 12/11/2022]
Abstract
Adenomatous polyposis coli (APC ) mutations are found in most colorectal tumours. These mutations are almost always protein-truncating, deleting both central domains that regulate Wnt signalling and C-terminal domains that interact with the cytoskeleton. The importance of Wnt dysregulation for colorectal tumourigenesis is well characterized. It is, however, unclear whether loss of C-terminal functions contributes to tumourigenesis, although this protein region has been implicated in cellular processes--including polarity, migration, mitosis, and chromosomal instability (CIN)—that have been postulated as critical for the development and progression of intestinal tumours. Since almost all APC mutations in human patients disrupt both central and C-terminal regions, we created a mouse model to test the role of the C-terminus of APC in intestinal tumourigenesis. This mouse (Apc(ΔSAMP)) carries an internal deletion within Apc that dysregulates Wnt by removing the beta-catenin-binding and SAMP repeats, but leaves the C-terminus intact. We compared Apc(ΔSAMP) mice with Apc(1322T) animals. The latter allele represented the most commonly found human APC mutation and was identical to Apc(ΔSAMP) except for absence of the entire C-terminus. Apc(ΔSAMP) mice developed numerous intestinal adenomas indistinguishable in number, location, and dysplasia from those seen in Apc(1322T) mice. No carcinomas were found in Apc(ΔSAMP) or Apc(1322T) animals. While similar disruption of the Wnt signalling pathway was observed in tumours from both mice, no evidence of differential C-terminus functions (such as cell migration, CIN, or localization of APC and EB1) was seen. We conclude that the C-terminus of APC does not influence intestinal adenoma development or progression.
Collapse
Affiliation(s)
- Annabelle Lewis
- Molecular and Population Genetics Laboratory, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
A knock-in mouse model reveals roles for nuclear Apc in cell proliferation, Wnt signal inhibition and tumor suppression. Oncogene 2011; 31:2423-37. [PMID: 21996741 PMCID: PMC3265630 DOI: 10.1038/onc.2011.434] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mutation of the tumor suppressor adenomatous polyposis coli (APC) is considered an initiating step in the genesis of the vast majority of colorectal cancers. APC inhibits the Wnt-signaling pathway by targeting the proto-oncogene β-catenin for destruction by cytoplasmic proteasomes. In the presence of a Wnt signal, or in the absence of functional APC, β-catenin can serve as a transcription cofactor for genes required for cell proliferation such as cyclin-D1 and c-Myc. In cultured cells, APC shuttles between the nucleus and the cytoplasm, with nuclear APC implicated in the inhibition of Wnt target gene expression. Adopting a genetic approach to evaluate the functions of nuclear APC in the context of a whole organism, we generated a mouse model with mutations that inactivate the nuclear localization signals (NLSs) of Apc (Apc(mNLS)). Apc(mNLS/mNLS) mice are viable and fractionation of mouse embryonic fibroblasts (MEFs) isolated from these mice revealed a significant reduction in nuclear Apc as compared with Apc(+/+) MEFs. The levels of Apc and β-catenin protein were not significantly altered in small intestinal epithelia from Apc(mNLS/mNLS) mice. Compared with Apc(+/+) mice, Apc(mNLS/mNLS) mice showed increased proliferation in epithelial cells from the jejunum, ileum and colon. These same tissues from Apc(mNLS/mNLS) mice showed more mRNA from three genes upregulated in response to canonical Wnt signal, c-Myc, axin-2 and cyclin-D1, and less mRNA from Hath-1, which is downregulated in response to Wnt. These observations suggest a role for nuclear Apc in the inhibition of canonical Wnt signaling and the control of epithelial proliferation in intestinal tissue. Furthermore, we found Apc(Min/+) mice, which harbor a mutation that truncates Apc, to have an increased polyp size and multiplicity if they also carry the Apc(mNLS) allele. Taken together, this analysis of the novel Apc(mNLS) mouse model supports a role for nuclear Apc in the control of Wnt target genes, intestinal epithelial cell proliferation and polyp formation.
Collapse
|
97
|
Tanwar PS, Zhang L, Teixeira JM. Adenomatous polyposis coli (APC) is essential for maintaining the integrity of the seminiferous epithelium. Mol Endocrinol 2011; 25:1725-39. [PMID: 21816903 DOI: 10.1210/me.2011-0057] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sertoli cells provide the microenvironment necessary for germ cell development and spermatogenesis; disruption of Sertoli cell morphology or function can lead to germ cell aplasia, which is observed in testicular dysgenesis syndrome. Mutation of the adenomatous polyposis coli (APC) gene has been associated with various human cancers, including testicular cancer, but its involvement in nonmalignant testicular pathologies has not been reported. We have developed a mouse model (APC(cko)) that expresses a truncated form of APC in Sertoli cells. Despite normal embryonic and early postnatal testicular development in APC(cko) mice, premature germ cell loss and Sertoli cell-only seminiferous tubules were observed in mutant testes without affecting Sertoli cell quiescence, apoptosis, or differentiation, which were confirmed by the absence of both proliferating cell nuclear antigen, DNA strand breaks, and anti-Müllerian hormone, respectively. We show that mutant Sertoli cells lose their apical extensions, which would normally enclose germ cells during various stages of spermatogenesis, and were unable to maintain the blood-testis barrier because of disrupted expression of junctional proteins. We also observed an up-regulation of Snail and Slug, markers suggestive of epithelial-mesenchymal transition in the Sertoli cells, but tumorigenesis was not observed. No comparable phenotype was observed with Sertoli cell-specific loss-of-function mutations in β-catenin, leading us to speculate that truncation of APC in Sertoli cells results in progressive degeneration of the seminiferous tubules by a mechanism that disrupts the integrity of Sertoli cell junctions independently of APC-regulated β-catenin activities and leads to development of a Sertoli cell-only phenotype.
Collapse
Affiliation(s)
- Pradeep S Tanwar
- Vincent Center for Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
98
|
Gerbe F, van Es JH, Makrini L, Brulin B, Mellitzer G, Robine S, Romagnolo B, Shroyer NF, Bourgaux JF, Pignodel C, Clevers H, Jay P. Distinct ATOH1 and Neurog3 requirements define tuft cells as a new secretory cell type in the intestinal epithelium. ACTA ACUST UNITED AC 2011; 192:767-80. [PMID: 21383077 PMCID: PMC3051826 DOI: 10.1083/jcb.201010127] [Citation(s) in RCA: 301] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tuft cells represent a fourth type of intestinal secretory cell that constitutes the primary source of endogenous intestinal opioids and are the only epithelial cell that constitutively express cyclooxygenases. The unique morphology of tuft cells was first revealed by electron microscopy analyses in several endoderm-derived epithelia. Here, we explore the relationship of these cells with the other cell types of the intestinal epithelium and describe the first marker signature allowing their unambiguous identification. We demonstrate that although mature tuft cells express DCLK1, a putative marker of quiescent stem cells, they are post-mitotic, short lived, derive from Lgr5-expressing epithelial stem cells, and are found in mouse and human tumors. We show that whereas the ATOH1/MATH1 transcription factor is essential for their differentiation, Neurog3, SOX9, GFI1, and SPDEF are dispensable, which distinguishes these cells from enteroendocrine, Paneth, and goblet cells, and raises from three to four the number of secretory cell types in the intestinal epithelium. Moreover, we show that tuft cells are the main source of endogenous intestinal opioids and are the only epithelial cells that express cyclooxygenase enzymes, suggesting important roles for these cells in the intestinal epithelium physiopathology.
Collapse
|
99
|
Regulation of homeostasis and oncogenesis in the intestinal epithelium by Ras. Exp Cell Res 2011; 317:2732-9. [PMID: 21741971 DOI: 10.1016/j.yexcr.2011.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/07/2011] [Accepted: 06/09/2011] [Indexed: 02/06/2023]
Abstract
Much of our current state of knowledge pertaining to the mechanisms controlling intestinal epithelial homeostasis derives from epidemiological, molecular genetic, cell biological, and biochemical studies of signaling pathways that are dysregulated during the process of colorectal tumorigenesis. Activating mutations in members of the RAS oncoprotein family play an important role in the progression of colorectal cancer (CRC) and, by extension, intestinal epithelial homeostasis. Mutations in K-RAS account for 90% of the RAS mutations found in CRC. As such, the study of RAS protein function in the intestinal epithelium is largely encompassed by the study of K-RAS function in CRC. In this review, we summarize the data available from genetically defined in vitro and in vivo models of CRC that aim to characterize the oncogenic properties of mutationally activated K-RAS. These studies paint a complex picture of a multi-functional oncoprotein that engages an array of downstream signaling pathways to influence cellular behaviors that are both pro- and anti-tumorigenic. While the complexity of K-RAS biology has thus far prevented a comprehensive understanding of its oncogenic properties, the work to date lays a foundation for the development of new therapeutic strategies to treat K-RAS mutant CRC.
Collapse
|
100
|
Gibb EA, Brown CJ, Lam WL. The functional role of long non-coding RNA in human carcinomas. Mol Cancer 2011; 10:38. [PMID: 21489289 PMCID: PMC3098824 DOI: 10.1186/1476-4598-10-38] [Citation(s) in RCA: 1314] [Impact Index Per Article: 101.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 04/13/2011] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are emerging as new players in the cancer paradigm demonstrating potential roles in both oncogenic and tumor suppressive pathways. These novel genes are frequently aberrantly expressed in a variety of human cancers, however the biological functions of the vast majority remain unknown. Recently, evidence has begun to accumulate describing the molecular mechanisms by which these RNA species function, providing insight into the functional roles they may play in tumorigenesis. In this review, we highlight the emerging functional role of lncRNAs in human cancer.
Collapse
Affiliation(s)
- Ewan A Gibb
- British Columbia Cancer Agency Research Centre, Vancouver, Canada.
| | | | | |
Collapse
|