Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Oncol ; 10: 1565, 2020.
Article in English | MEDLINE | ID: mdl-32850460

ABSTRACT

INTRODUCTION: Colorectal cancer (CRC) is the second leading cause of cancer death worldwide and most deaths result from metastases. We have analyzed animal models in which Apc, a gene that is frequently mutated during the early stages of colorectal carcinogenesis, was inactivated and human samples to try to identify novel potential biomarkers for CRC. MATERIALS AND METHODS: We initially compared the proteomic and transcriptomic profiles of the small intestinal epithelium of transgenic mice in which Apc and/or Myc had been inactivated. We then studied the mRNA and immunohistochemical expression of one protein that we identified to show altered expression following Apc inactivation, nucleosome assembly protein 1-like 1 (NAP1L1) in human CRC samples and performed a prognostic correlation between biomarker expression and survival in CRC patients. RESULTS: Nap1l1 mRNA expression was increased in mouse small intestine following Apc deletion in a Myc dependant manner and was also increased in human CRC samples. Immunohistochemical NAP1L1 expression was decreased in human CRC samples relative to matched adjacent normal colonic tissue. In a separate cohort of 75 CRC patients, we found a strong correlation between NAP1L1 nuclear expression and overall survival in those patients who had stage III and IV cancers. CONCLUSION: NAP1L1 expression is increased in the mouse small intestine following Apc inactivation and its expression is also altered in human CRC. Immunohistochemical NAP1L1 nuclear expression correlated with overall survival in a cohort of CRC patients. Further studies are now required to clarify the role of this protein in CRC.

2.
Sci Rep ; 10(1): 11169, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32612166

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
BMC Cancer ; 19(1): 677, 2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31291912

ABSTRACT

BACKGROUND: Canonical WNT signalling plays a critical role in the regulation of ovarian development; mis-regulation of this key pathway in the adult ovary is associated with subfertility and tumourigenesis. The roles of Adenomatous polyposis coli 2 (APC2), a little-studied WNT signalling pathway regulator, in ovarian homeostasis, fertility and tumourigenesis have not previously been explored. Here, we demonstrate essential roles of APC2 in regulating ovarian WNT signalling and ovarian homeostasis. METHODS: A detailed analysis of ovarian histology, gene expression, ovulation and hormone levels was carried out in 10 week old and in aged constitutive APC2-knockout (Apc2-/-) mice (mixed background). Statistical significance for qRT-PCR data was determined from 95% confidence intervals. Significance testing was performed using 2-tailed Student's t-test, when 2 experimental cohorts were compared. When more were compared, ANOVA test was used, followed by a post-hoc test (LSD or Games-Howell). P-values of < 0.05 were considered statistically significant. RESULTS: APC2-deficiency resulted in activation of ovarian WNT signalling and sub-fertility driven by intra-ovarian defects. Follicular growth was perturbed, resulting in a reduced rate of ovulation and corpora lutea formation, which could not be rescued by administration of gonadotrophins. Defects in steroidogenesis and follicular vascularity contributed to the subfertility phenotype. Tumour incidence was assessed in aged APC2-deficient mice, which also carried a hypomorphic Apc allele. APC2-deficiency in these mice resulted in predisposition to granulosa cell tumour (GCT) formation, accompanied by acute tumour-associated WNT-signalling activation and a histologic pattern and molecular signature seen in human adult GCTs. CONCLUSIONS: Our work adds APC2 to the growing list of WNT-signalling members that regulate ovarian homeostasis, fertility and suppress GCT formation. Importantly, given that the APC2-deficient mouse develops tumours that recapitulate the molecular signature and histological features of human adult GCTs, this mouse has excellent potential as a pre-clinical model to study ovarian subfertility and transitioning to GCT, tumour biology and for therapeutic testing.


Subject(s)
Carcinogenesis/metabolism , Cytoskeletal Proteins/metabolism , Fertility , Ovary/metabolism , Wnt Signaling Pathway , Analysis of Variance , Animals , Cytoskeletal Proteins/genetics , Female , Forkhead Box Protein O1/metabolism , Gene Knockout Techniques , Granulosa Cell Tumor/etiology , Granulosa Cell Tumor/metabolism , Homeostasis , Infertility/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Ovarian Follicle/growth & development , Ovarian Neoplasms/etiology , Ovarian Neoplasms/metabolism , beta Catenin/metabolism
4.
Sci Rep ; 8(1): 2735, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426940

ABSTRACT

While the Wnt/ß-catenin pathway plays a critical role in the maintenance of the zonation of ammonia metabolizing enzymes in the adult liver, the mechanisms responsible for inducing zonation in the embryo are not well understood. Herein we address the spatiotemporal role of the Wnt/ß-catenin pathway in the development of zonation in embryonic mouse liver by conditional deletion of Apc and ß-catenin at different stages of mouse liver development. In normal development, the ammonia metabolising enzymes carbamoylphosphate synthetase I (CPSI) and Glutamine synthetase (GS) begin to be expressed in separate hepatoblasts from E13.5 and E15.5 respectively and gradually increase in number thereafter. Restriction of GS expression occurs at E18 and becomes increasingly limited to the terminal perivenous hepatocytes postnatally. Expression of nuclear ß-catenin coincides with the restriction of GS expression to the terminal perivenous hepatocytes. Conditional loss of Apc resulted in the expression of nuclear ß-catenin throughout the developing liver and increased number of cells expressing GS. Conversely, conditional loss of ß-catenin resulted in loss of GS expression. These data suggest that the Wnt pathway is critical to the development of zonation as well as maintaining the zonation in the adult liver.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Glutamate-Ammonia Ligase/metabolism , Hepatocytes/metabolism , Liver/embryology , Wnt Signaling Pathway/physiology , beta Catenin/physiology , Adenomatous Polyposis Coli Protein/genetics , Ammonia/metabolism , Animals , Hepatocytes/cytology , Liver/metabolism , Loss of Function Mutation , Mice , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics
5.
Curr Colorectal Cancer Rep ; 12(5): 281-287, 2016.
Article in English | MEDLINE | ID: mdl-27656116

ABSTRACT

Modelling human diseases in in vitro systems is undisputedly an invaluable research tool, yet there are many limitations. Some of those limitations have been overcome through the introduction of organoid culture systems, which have revolutionised colorectal cancer research and enabled an array of new experimental techniques. This 3D system models the physiology, shape, dynamics and cell make-up of the intestinal epithelium producing a relevant and highly adaptable model system. The increased functional relevance of this model compared to the use of 2D cancer cell lines makes it an invaluable tool for both basic and translational research. As the limitations of this system are being overcome to make high-throughput assays possible, it is clear that organoids are becoming a mainstay of colorectal cancer research. This review aims to explore the advantages and limitations of this system and discusses the future directions enabled by this model.

6.
Oncotarget ; 7(32): 51665-51673, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27323825

ABSTRACT

BACKGROUND AND AIMS: Colorectal cancer (CRC) arises via multiple genetic changes. Mutation of the tumour suppressor gene APC, a key regulator of Wnt signalling, is recognised as a frequent early driving mutation in CRC. We have previously shown that conditional loss of Apc within the murine small intestine (Apcfloxmice) results in acute Wnt signalling activation, altered crypt-villus architecture and many hallmarks of neoplasia. Our transctipomic profiling (Affymetrix Microarrays) and proteomic profiling (iTRAQ-QSTAR) of Apc-deficient intestine inferred the involvement of High Mobility Group Box 1 (Hmgb1) in CRC pathogenesis. Here we assess the contribution of HMGB1 to the crypt progenitor phenotype seen following Apc loss. RESULTS: Elevated HMGB1 was confirmed in intestinal epithelia and serum following conditional loss of Apc. Treatment of Apcflox mice with anti-HMGB1 neutralising antibody significantly reduced many of the crypt progenitor phenotypes associated with Apc loss; proliferation and apoptosis levels were reduced, cell differentiation was restored and the expansion of stem cell marker expression was eradicated. METHODS: Hmgb1 levels in intestinal epithelia and serum in Apcflox and ApcMin mice were assessed using qRT-PCR, Western blot and ELISA assays. The functional importance of elevated extracellular Hmgb1 was assessed using an anti-HMGB1 neutralising antibody in Apcflox mice. CONCLUSIONS: HMGB1 is expressed and secreted from intestinal epithelial cells in response to Wnt signalling activation. This secreted HMGB1 is required to maintain nearly all aspects of the crypt progenitor phenotype observed following Apc loss and add to the body of accumulating evidence indicating that targeting HMGB1 may be a viable novel therapeutic approach.


Subject(s)
HMGB1 Protein/metabolism , Intestinal Mucosa/metabolism , Stem Cell Niche , Stem Cells/cytology , Wnt Proteins/metabolism , Animals , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, APC , HMGB1 Protein/genetics , Intestine, Small/metabolism , Mice , Mice, Transgenic , Microarray Analysis , Phenotype , Stem Cells/metabolism , Wnt Signaling Pathway
7.
EMBO J ; 34(18): 2321-33, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26240067

ABSTRACT

Wnt pathway deregulation is a common characteristic of many cancers. Only colorectal cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of the pancreas) have activating mutations in ß-catenin (CTNNB1). We have compared the dynamics and the potency of ß-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of ß-catenin took much longer to achieve Wnt deregulation and acquire a crypt-progenitor cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of ß-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of ß-catenin mutation to differentially transform the SI versus the colon correlated with higher expression of E-cadherin and a higher number of E-cadherin:ß-catenin complexes at the membrane. Reduction in E-cadherin synergised with an activating mutation of ß-catenin resulting in a rapid CPC phenotype within the SI and colon. Thus, there is a threshold of ß-catenin that is required to drive transformation, and E-cadherin can act as a buffer to sequester mutated ß-catenin.


Subject(s)
Cadherins/metabolism , Cell Transformation, Neoplastic , Colonic Neoplasms , Mutation , Neoplasm Proteins , Wnt Signaling Pathway , beta Catenin , Animals , Cadherins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
8.
BMC Cancer ; 15: 110, 2015 Mar 08.
Article in English | MEDLINE | ID: mdl-25881306

ABSTRACT

BACKGROUND: Conditional deletion of the tumour suppressor gene Apc within the murine intestine results in acute Wnt signalling activation. The associated over-expression of a myriad of Wnt signalling target genes yields phenotypic alterations that encompass many of the hallmarks of neoplasia. Previous transcriptomic analysis aimed at identifying genes that potentially play an important role in this process, inferred the Hormonally upregulated Neu-associated kinase (HUNK/Mak-v/Bstk1) gene as a possible candidate. Hunk is a SNF1 (sucrose non fermenting 1)-related serine/threonine kinase with a proposed association with many different tumour types, including colorectal cancer. METHODS: Here we describe the generation of a novel Hunk kinase deficient mouse which has been used to investigate the involvement of Hunk-kinase activity in intestinal homeostasis and tumourigenesis. RESULTS: We show that in the morphologically normal intestine, Hunk-kinase negatively regulates epithelial cell proliferation. However, the increase in cell proliferation observed in the Hunk kinase deficient intestine is counteracted by increased cell migration, thereby maintaining intestinal homeostasis. Using qRT-PCR, we further demonstrate that Hunk is significantly over-expressed in Apc deficient / Wnt-signalling activated intestinal tissue. Using the classical intestinal tumourigenesis Apc (Min) mouse model we show that loss of Hunk-kinase activity significantly reduced tumour initiation rates in the small intestine. However, an accompanying increase in the size of the tumours counteracts the impact this has on overall tumour burden or subsequently survival. CONCLUSIONS: In the intestinal setting we demonstrate that Hunk has a role in normal intestinal proliferation and homeostasis and, although it does not alter overall survival rates, activity of this kinase does impact on tumour initiation rates during the early stages in tumourigenesis in the small intestine.


Subject(s)
Intestinal Mucosa/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation , Gene Knockdown Techniques , Gene Targeting , Genetic Loci , Male , Mice , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Protein Kinases/deficiency , Protein Serine-Threonine Kinases , Tumor Burden , Up-Regulation , Wnt Signaling Pathway
9.
Biochem Biophys Res Commun ; 440(3): 364-70, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-23998936

ABSTRACT

Colorectal cancer (CRC) is the fourth most common cause of cancer-related death worldwide. Accurate non-invasive screening for CRC would greatly enhance a population's health. Adenomatous polyposis coli (Apc) gene mutations commonly occur in human colorectal adenomas and carcinomas, leading to Wnt signalling pathway activation. Acute conditional transgenic deletion of Apc in murine intestinal epithelium (AhCre(+)Apc(fl)(/)(fl)) causes phenotypic changes similar to those found during colorectal tumourigenesis. This study comprised a proteomic analysis of murine small intestinal epithelial cells following acute Apc deletion to identify proteins that show altered expression during human colorectal carcinogenesis, thus identifying proteins that may prove clinically useful as blood/serum biomarkers of colorectal neoplasia. Eighty-one proteins showed significantly increased expression following iTRAQ analysis, and validation of nine of these by Ingenuity Pathaway Analysis showed they could be detected in blood or serum. Expression was assessed in AhCre(+)Apc(fl)(/)(fl) small intestinal epithelium by immunohistochemistry, western blot and quantitative real-time PCR; increased nucelolin concentrations were also detected in the serum of AhCre(+)Apc(fl)(/)(fl) and Apc(Min)(/)(+) mice by ELISA. Six proteins; heat shock 60kDa protein 1, Nucleolin, Prohibitin, Cytokeratin 18, Ribosomal protein L6 and DEAD (Asp-Glu-Ala-Asp) box polypeptide 5,were selected for further investigation. Increased expression of 4 of these was confirmed in human CRC by qPCR. In conclusion, several novel candidate biomarkers have been identified from analysis of transgenic mice in which the Apc gene was deleted in the intestinal epithelium that also showed increased expression in human CRC. Some of these warrant further investigation as potential serum-based biomarkers of human CRC.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Animals , Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Gene Deletion , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Mice , Mice, Transgenic , Prohibitins , Proteomics
10.
Gut ; 61(10): 1435-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22138533

ABSTRACT

OBJECTIVE: Expression of the Wnt target gene ASCL2 is elevated in 78% of intestinal neoplasia datasets (Oncomine), suggesting a role for deregulated ASCL2 in the aetiology of intestinal tumourigenesis. Furthermore, ectopic expression of Ascl2 has previously been shown to lead to hyperplasia in the mouse. However, elevated levels of ASCL2 does not have an impact on the overall survival or recurrence-free survival rates in colorectal cancer patients. Here the authors use a novel mouse model to analyse the role of Ascl2 in intestinal tumourigenesis and address the controversy surrounding the relevance of this gene to the aetiology of colorectal cancer. DESIGN: The authors have generated a mouse possessing a transgene carrying the Ascl2 gene together with its endogenous promoter and regulatory regions, thereby elevating Ascl2 expression in an authentic manner. The authors have further intercrossed these Ascl2 overexpressers to the classic Apc(Min) model, to study the consequence of elevated Ascl2 expression in intestinal tumourigenesis. RESULTS: Here the authors genetically demonstrate that elevated expression of Ascl2 in a Wnt signalling dependent manner specifically in the stem cell compartment of the intestine neither increases tumour formation nor diminishes survival in a well established intestinal tumour model, the Apc(min) mouse. CONCLUSION: The authors conclude that ectopic expression of Ascl2 is more important in the aetiology of neoplasia than overexpression of Ascl2.


Subject(s)
Adenoma/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Adenoma/mortality , Animals , Colorectal Neoplasms/mortality , Intestinal Mucosa/pathology , Mice , Stem Cells/pathology , Up-Regulation , Wnt Signaling Pathway
11.
Gut ; 60(7): 893-901, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21303912

ABSTRACT

BACKGROUND: Rectal epithelial cell mitosis and crypt size, as well as expression of proinflammatory genes including macrophage migration inhibitory factor (MIF), are increased 6 months after Roux-en-Y gastric bypass (RYGB) in morbidly obese patients. Tests were carried out to determine whether these putative colorectal cancer risk biomarkers remained elevated long term after RYGB, and the mechanistic basis, as well as the functional consequences, of Mif upregulation in intestinal epithelial cells was investigated. METHODS: Rectal mucosa and blood were obtained a median of 3 years after RYGB from the original cohort of patients with RYGB (n = 19) for crypt microdissection, real-time PCR, immunohistochemistry for MIF and immunoassay of proinflammatory markers. Immunohistochemistry for Mif and bromodeoxyuridine labelling were performed on AhCre⁺ mouse and Apc(Min/⁺) mouse (with and without functional Mif alleles) intestine, respectively. RESULTS: Rectal epithelial cell mitosis and crypt size remained elevated 3 years after RYGB compared with preoperative values (1.7- and 1.5-fold, respectively; p < 0.05). There was a 40-fold (95% CI 13 to 125) increase in mucosal MIF transcript levels at 3 years associated with increased epithelial cell MIF protein levels. Conditional Apc loss in AhCre⁺ mice led to increased epithelial cell Mif content. Mif deficiency in Apc(Min/⁺) mice was associated with a combined defect in intestinal epithelial cell proliferation and migration, which was reflected by the longitudinal clinical data. CONCLUSIONS: Mucosal abnormalities persist 3 years after RYGB and include elevation of the protumorigenic cytokine MIF, which is upregulated following Apc loss and which contributes to intestinal epithelial cell homeostasis. These observations should prompt clinical studies of colorectal neoplastic risk after RYGB.


Subject(s)
Gastric Bypass/adverse effects , Macrophage Migration-Inhibitory Factors/metabolism , Obesity, Morbid/surgery , Rectum/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Movement/physiology , Cell Proliferation , Colorectal Neoplasms/etiology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Follow-Up Studies , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophage Migration-Inhibitory Factors/blood , Macrophage Migration-Inhibitory Factors/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitosis , Postoperative Period , Rectum/metabolism , Treatment Outcome , Up-Regulation
12.
Cancer Res ; 70(20): 8149-58, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20736363

ABSTRACT

Inactivation of the Apc gene is recognized as the key early event in the development of sporadic colorectal cancer (CRC), where its loss leads to constitutive activation of ß-catenin/T-cell factor 4 signaling and hence transcription of Wnt target genes such as c-Myc. Our and other previous studies have shown that although cyclin D1 is required for adenoma formation, it is not immediately upregulated following Apc loss within the intestine, suggesting that proliferation following acute Apc loss may be dependent on another D-type cyclin. In this study, we investigated the expression and functional relevance of cyclin D2 following Apc loss in the intestinal epithelium. Cyclin D2 is upregulated immediately following Apc loss, which corresponded with a significant increase in cyclin-dependent kinase 4 (CDK4) and hyperphosphorylated Rb levels. Deficiency of cyclin D2 resulted in a reduction in enterocyte proliferation and crypt size within Apc-deficient intestinal epithelium. Moreover, cyclin D2 dramatically reduced tumor growth and development in Apc(Min/+) mice. Importantly, cyclin D2 knockout did not affect proliferation of normal enterocytes, and furthermore, CDK4/6 inhibition also suppressed the proliferation of adenomatous cells and not normal cells from Apc(Min/+) mice. Taken together, these results indicate that cyclin D-CDK4/6 complexes are required for the efficient proliferation of cells with deregulated Wnt signaling, and inhibiting this complex may be an effective chemopreventative strategy in CRC.


Subject(s)
Adenoma/pathology , Cyclin D2/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Genes, APC , Adenoma/genetics , Animals , Apoptosis , Cell Division , Cyclin D2/deficiency , Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , DNA Primers , Gene Deletion , Genes, myc , Immunohistochemistry , Intestinal Polyposis/genetics , Intestinal Polyposis/pathology , Mice , Mitotic Index , Polymerase Chain Reaction , Up-Regulation
13.
Gastroenterology ; 136(7): 2316-2324.e1-3, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19268669

ABSTRACT

BACKGROUND AND AIMS: The Wnt pathway has previously been shown to play a role in hepatic zonation. Herein, we have explored the role of 3 key components (Apc, beta-catenin, and c-Myc) of the Wnt pathway in the zonation of ammonia metabolizing enzymes. METHODS: Conditional deletion of Apc, beta-catenin, and c-Myc was induced in the livers of mice and the expression of periportal and perivenous hepatocyte markers was determined by polymerase chain reaction, Western blotting, and immunohistochemical techniques. RESULTS: Under normal circumstances, the urea cycle enzyme carbamoylphosphate synthetase I (CPS I) is present in the periportal, intermediate, and the first few layers of the perivenous zone. In contrast, glutamine synthetase (GS)--and nuclear beta-catenin--is expressed in a complementary fashion in the last 1-2 cell layers of the perivenous zone. Conditional loss of Apc resulted in the expression of nuclear beta-catenin and GS in most hepatocytes irrespective of zone. Induction of GS in hepatocytes outside the normal perivenous zone was accompanied by a reduction in the expression of CPS I. Deletion of beta-catenin induces a loss of GS and a complementary increase in expression of CPS I irrespective of whether Apc is present. Remarkably, deletion of c-Myc did not perturb the pattern of zonation. CONCLUSIONS: It has been shown that the Wnt pathway is key to imposing the pattern of zonation within the liver. Herein we have addressed the relevance of 3 major Wnt pathway components and show critically that the zonation is c-Myc independent but beta-catenin dependent.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Gene Deletion , Gene Expression Regulation , Hepatocytes/enzymology , Hepatocytes/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Models, Animal , Proto-Oncogene Proteins c-myc/genetics , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Signal Transduction/physiology , Wnt Proteins/genetics , beta Catenin/genetics
14.
Proc Natl Acad Sci U S A ; 105(48): 18919-23, 2008 Dec 02.
Article in English | MEDLINE | ID: mdl-19033191

ABSTRACT

Dysregulated Wnt signaling is seen in approximately 30% of hepatocellular carcinomas; thus, finding pathways downstream of the activation of Wnt signaling is key. Here, using cre-lox technology, we deleted the Apc gene in the adult mouse liver and observed a rapid increase in nuclear beta-catenin and c-Myc, which is associated with an induction of proliferation that led to hepatomegaly within 4 days of gene deletion. To investigate the downstream pathways responsible for these phenotypes, we analyzed the impact of inactivating APC in the context of deficiency of the potentially key effectors beta-catenin and c-Myc. beta-catenin loss rescues both the proliferation and hepatomegaly phenotypes after APC loss. However, c-Myc deletion, which rescues the phenotypes of APC loss in the intestine, had no effect on the phenotypes of APC loss in the liver. The consequences of the deregulation of the Wnt pathway within the liver are therefore strikingly different from those observed within the intestine, with the vast majority of Wnt targets being beta-catenin-dependent but c-Myc-independent in the liver.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Liver/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/deficiency , Adenomatous Polyposis Coli Protein/genetics , Animals , Gene Deletion , Liver/cytology , Liver/pathology , Male , Mice , Mice, Transgenic , Microarray Analysis , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Wnt Proteins/genetics , beta Catenin/genetics
15.
Mol Cell Biol ; 28(19): 6094-103, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18644872

ABSTRACT

We have previously shown that deficiency of the methyl binding domain protein Mbd2 dramatically reduces adenoma burden on an Apc(Min/+) background. To investigate the mechanism underlying this phenomenon, we have determined the effect of Mbd2 deficiency upon the phenotypes imposed by the conditional deletion of Apc in the small intestine. Microarray analysis demonstrated a partial suppression of the Wnt pathway in the absence of Mbd2. Mbd2 deficiency also influenced one immediate cellular consequence of Apc loss, with normalization of Paneth cell positioning. From a mechanistic perspective, we show that deficiency of Mbd2 elevates levels of the known Wnt target Lect2, and we confirm here that Mbd2 binds the Lect2 promoter in association with NuRD. Furthermore, we show that Lect2 is capable of functioning as a Wnt pathway repressor. These results therefore provide a mechanistic basis for the epigenetic control of adenoma formation mediated through Mbd2.


Subject(s)
Signal Transduction , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, APC , Intercellular Signaling Peptides and Proteins/metabolism , Intestine, Small/pathology , Paneth Cells/metabolism , Promoter Regions, Genetic , Wnt Proteins/metabolism
16.
BMC Cancer ; 8: 162, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18533991

ABSTRACT

BACKGROUND: p53 is an important tumour suppressor with a known role in the later stages of colorectal cancer, but its relevance to the early stages of neoplastic initiation remains somewhat unclear. Although p53-dependent regulation of Wnt signalling activity is known to occur, the importance of these regulatory mechanisms during the early stages of intestinal neoplasia has not been demonstrated. METHODS: We have conditionally deleted the Adenomatous Polyposis coli gene (Apc) from the adult murine intestine in wild type and p53 deficient environments and subsequently compared the phenotype and transcriptome profiles in both genotypes. RESULTS: Expression of p53 was shown to be elevated following the conditional deletion of Apc in the adult small intestine. Furthermore, p53 status was shown to impact on the transcription profile observed following Apc loss. A number of key Wnt pathway components and targets were altered in the p53 deficient environment. However, the aberrant phenotype observed following loss of Apc (rapid nuclear localisation of beta-catenin, increased levels of DNA damage, nuclear atypia, perturbed cell death, proliferation, differentiation and migration) was not significantly altered by the absence of p53. CONCLUSION: p53 related feedback mechanisms regulating Wnt signalling activity are present in the intestine, and become activated following loss of Apc. However, the physiological Wnt pathway regulation by p53 appears to be overwhelmed by Apc loss and consequently the activity of these regulatory mechanisms is not sufficient to modulate the immediate phenotypes seen following Apc loss. Thus we are able to provide an explanation to the apparent contradiction that, despite having a Wnt regulatory capacity, p53 loss is not associated with early lesion development.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, APC , Intestinal Neoplasms/genetics , Intestine, Small/physiology , Tumor Suppressor Protein p53/genetics , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Deletion , Gene Expression Regulation , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Mice , Signal Transduction , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/deficiency , Wnt Proteins/metabolism
17.
Nature ; 446(7136): 676-9, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17377531

ABSTRACT

The APC gene encodes the adenomatous polyposis coli tumour suppressor protein, germline mutation of which characterizes familial adenomatous polyposis (FAP), an autosomal intestinal cancer syndrome. Inactivation of APC is also recognized as the key early event in the development of sporadic colorectal cancers, and its loss results in constitutive activity of the beta-catenin-Tcf4 transcription complex. The proto-oncogene c-MYC has been identified as a target of the Wnt pathway in colorectal cancer cells in vitro, in normal crypts in vivo and in intestinal epithelial cells acutely transformed on in vivo deletion of the APC gene; however, the significance of this is unclear. Therefore, to elucidate the role Myc has in the intestine after Apc loss, we have simultaneously deleted both Apc and Myc in the adult murine small intestine. Here we show that loss of Myc rescued the phenotypes of perturbed differentiation, migration, proliferation and apoptosis, which occur on deletion of Apc. Remarkably, this rescue occurred in the presence of high levels of nuclear beta-catenin. Array analysis revealed that Myc is required for the majority of Wnt target gene activation following Apc loss. These data establish Myc as the critical mediator of the early stages of neoplasia following Apc loss.


Subject(s)
Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Gene Deletion , Genes, APC , Genes, myc/genetics , Intestine, Small/metabolism , Proto-Oncogene Proteins c-myc/deficiency , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Enterocytes/metabolism , Intestine, Small/cytology , Male , Mice , Phenotype , Proto-Oncogene Proteins c-myc/genetics
18.
BMC Cancer ; 6: 113, 2006 May 03.
Article in English | MEDLINE | ID: mdl-16672050

ABSTRACT

BACKGROUND: Therapeutic regulation of PPARdelta activity using selective agonists has been proposed for various disorders. However, the consequences of altered peroxisome proliferator-activated receptor delta (PPARdelta) activity in the context of intestinal tumourigenesis remain somewhat unclear. Contradictory evidence suggesting PPARdelta either attenuates or potentiates intestinal neoplasia. To further investigate the PPARdelta dependency of intestinal tumourigenesis, we have analysed the consequences of PPARdelta deficiency upon intestinal neoplasia occurring in mice with impaired mismatch DNA repair. METHODS: Mice deficient for both PPARdelta and the mismatch repair gene Mlh1 were produced and the incidence and severity of intestinal neoplasia recorded. RESULTS: No significant differences between the control genotypes and the double mutant genotypes were recorded indicating that deficiency of PPARdelta does not modify impaired mismatch repair induced neoplasia. CONCLUSION: In contrast with the previously observed acceleration of intestinal neoplasia in the context of the ApcMin/+ mouse, PPARdelta deficiency does not alter the phenotype of mismatch repair deficiency. This data supports the notion that PPARdelta is not required for adenoma formation and indicate that any pro-tumourigenic effect of PPARdelta inactivation may be highly context dependent.


Subject(s)
Adenoma/etiology , DNA Repair , Intestinal Neoplasms/etiology , PPAR delta/physiology , Adaptor Proteins, Signal Transducing , Adenoma/genetics , Animals , Base Pair Mismatch , Carrier Proteins/genetics , Intestinal Neoplasms/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , MutL Protein Homolog 1 , Nuclear Proteins/genetics , PPAR delta/genetics , Survival Analysis
19.
Oncogene ; 24(55): 8205-10, 2005 Dec 08.
Article in English | MEDLINE | ID: mdl-16116480

ABSTRACT

Deregulation of Wnt signalling has recently been implicated in human renal cancer. Here, we directly test this association by using a Cre-LoxP strategy to inactivate the Adenomatous Polyposis Coli (Apc) gene in the murine renal epithelium. Mice homozygous for a conditional Apc allele were intercrossed with mice transgenic for Cre recombinase under control of the Cyp1A promoter, which delivers constitutive recombination within a proportion of cells in the renal epithelium. Inactivation of Apc leads to the accumulation of nuclear beta-catenin and the rapid development of multiple dysplastic foci. Renal carcinoma was observed with an earliest onset of 4 months. This predisposition was accelerated by p53 deficiency, reducing the earliest onset to 2 months. Compared to other murine models of kidney neoplasia, this represents particularly rapid onset of disease, and so implicates an important role for Apc in suppressing renal carcinoma.


Subject(s)
Gene Deletion , Genes, APC , Kidney Neoplasms/genetics , Animals , Homozygote , Integrases/genetics , Integrases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Urothelium/pathology , Urothelium/physiology
20.
J Biol Chem ; 280(31): 28463-7, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-15946945

ABSTRACT

Cyclin D1 is postulated to be a target of the canonical Wnt pathway and critical for intestinal adenoma development. We show here that, unlike cyclin D1 reporter assays, endogenous cyclin D1 levels are not affected following antagonism of the Wnt pathway in vitro, nor is cyclin D1 immediately up-regulated following conditional loss of Apc in vivo. Cyclin D1 levels do, however, increase in a delayed manner in a small subset of cells, suggesting such up-regulation occurs as a secondary event. We also analyzed the immediate consequences of Apc loss in a cyclin D1(-/-) background and failed to find any cyclin D1-dependent phenotypes. However, we did observe elevated cyclin D1 expression in lesions developing 20 days after Apc loss. In these circumstances, all adenomas (but not smaller lesions) showed cyclin D1 up-regulation. Finally in a smaller study, we analyzed whether cyclin D1 deficiency affected adenoma formation 20 days following induced loss of Apc. Unlike AhCre(+) Apc(fl/fl) mice (which all developed adenomas), doubly mutant AhCre(+) Apc(fl/fl) cyclin D1(-/-) mice only developed small lesions. Taken together, this argues that cyclin D1 up-regulation in intestinal neoplasia is important for tumor progression rather than initiation.


Subject(s)
Cyclin D1/physiology , Cytoskeletal Proteins/metabolism , Genes, APC , Trans-Activators/metabolism , Adenoma , Animals , Cell Line, Tumor , Colorectal Neoplasms , Cyclin D1/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Integrases/metabolism , Intestinal Neoplasms , Mice , Mice, Inbred C57BL , Phenotype , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...