Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Lab Invest ; 99(10): 1454-1469, 2019 10.
Article in English | MEDLINE | ID: mdl-31148594

ABSTRACT

Somatic APC (adenomatous polyposis coli), TP53, KRAS mutations are present in roughly 80%, 60%, and 40%, respectively, of human colorectal cancers (CRCs). Most TP53 mutant alleles in CRCs encode missense mutant proteins with loss-of-function (LOF) of p53's transcriptional activity and dominant negative (DN) effects on wild-type p53 function. Missense mutant p53 proteins have been reported to exert gain-of-function (GOF) effects in cancer. We compared the phenotypic effects of the common human cancer-associated TP53 R273H missense mutation to p53 null status in a genetically engineered mouse CRC model. Inactivation of one allele of Apc together with activation of a Kras mutant allele in mouse colon epithelium instigated development of serrated and hyperplastic epithelium and adenomas (AK mice). Addition of a Trp53R270H or Trp53null mutant allele to the model (AKP mice) led to markedly shortened survival and increased tumor burden relative to that of AK mice, including adenocarcinomas in AKP mice. Comparable life span and tumor burden were seen in AKP mice carrying Trp53R270H or Trp53null alleles, along with similar frequencies of spontaneous metastasis to lymph nodes, lung, and liver. The fraction of adenocarcinomas with submucosa or deeper invasion was higher in AKP270/fl mice than in AKPfl/fl mice, but the incidence of adenocarcinomas per mouse did not differ significantly between AKPfl/fl and AKP270/fl mice. In line with their comparable biological behaviors, mouse primary tumors and tumor-derived organoids with the Trp53R270H or Trp53null alleles had highly similar gene expression profiles. Human CRCs with TP53 R273 missense mutant or null alleles also had essentially homogeneous gene expression patterns. Our findings indicate the R270H/R273H p53 mutant protein does not manifest definite GOF biological effects in mouse and human CRCs, suggesting possible GOF effects of mutant p53 in cancer phenotypes are likely allele-specific and/or context-dependent.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis , Disease Progression , Epithelial-Mesenchymal Transition , Gene Expression , Humans , Mice, Transgenic , Mutation, Missense , Neoplasm Invasiveness , Neoplasm Metastasis
2.
Elife ; 62017 01 10.
Article in English | MEDLINE | ID: mdl-28072391

ABSTRACT

While 20-30% of colorectal cancers (CRCs) may arise from precursors with serrated glands, only 8-10% of CRCs manifest serrated morphology at diagnosis. Markers for distinguishing CRCs arising from 'serrated' versus 'conventional adenoma' precursors are lacking. We studied 36 human serrated CRCs and found CDX2 loss or BRAF mutations in ~60% of cases and often together (p=0.04). CDX2Null/BRAFV600E expression in adult mouse intestinal epithelium led to serrated morphology tumors (including carcinomas) and BRAFV600E potently interacted with CDX2 silencing to alter gene expression. Like human serrated lesions, CDX2Null/BRAFV600E-mutant epithelium expressed gastric markers. Organoids from CDX2Null/BRAFV600E-mutant colon epithelium showed serrated features, and partially recapitulated the gene expression pattern in mouse colon tissues. We present a novel mouse tumor model based on signature defects seen in many human serrated CRCs - CDX2 loss and BRAFV600E. The mouse intestinal tumors show significant phenotypic similarities to human serrated CRCs and inform about serrated CRC pathogenesis.


Subject(s)
CDX2 Transcription Factor/metabolism , Carcinogenesis , Colorectal Neoplasms/physiopathology , Proto-Oncogene Proteins B-raf/metabolism , Animals , CDX2 Transcription Factor/genetics , Colon/pathology , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Profiling , Intestinal Mucosa/pathology , Mice , Organoids , Proto-Oncogene Proteins B-raf/genetics
3.
PLoS Genet ; 11(11): e1005638, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26528816

ABSTRACT

Adenomatous polyposis coli (APC) inactivating mutations are present in most human colorectal cancers and some other cancers. The APC protein regulates the ß-catenin protein pool that functions as a co-activator of T cell factor (TCF)-regulated transcription in Wnt pathway signaling. We studied effects of reduced dosage of the Ctnnb1 gene encoding ß-catenin in Apc-mutation-induced colon and ovarian mouse tumorigenesis and cell culture models. Concurrent somatic inactivation of one Ctnnb1 allele, dramatically inhibited Apc mutation-induced colon polyposis and greatly extended Apc-mutant mouse survival. Ctnnb1 hemizygous dose markedly inhibited increases in ß-catenin levels in the cytoplasm and nucleus following Apc inactivation in colon epithelium, with attenuated expression of key ß-catenin/TCF-regulated target genes, including those encoding the EphB2/B3 receptors, the stem cell marker Lgr5, and Myc, leading to maintenance of crypt compartmentalization and restriction of stem and proliferating cells to the crypt base. A critical threshold for ß-catenin levels in TCF-regulated transcription was uncovered for Apc mutation-induced effects in colon epithelium, along with evidence of a feed-forward role for ß-catenin in Ctnnb1 gene expression and CTNNB1 transcription. The active ß-catenin protein pool was highly sensitive to CTNNB1 transcript levels in colon cancer cells. In mouse ovarian endometrioid adenocarcinomas (OEAs) arising from Apc- and Pten-inactivation, while Ctnnb1 hemizygous dose affected ß-catenin levels and some ß-catenin/TCF target genes, Myc induction was retained and OEAs arose in a fashion akin to that seen with intact Ctnnb1 gene dose. Our findings indicate Ctnnb1 gene dose exerts tissue-specific differences in Apc mutation-instigated tumorigenesis. Differential expression of selected ß-catenin/TCF-regulated genes, such as Myc, likely underlies context-dependent effects of Ctnnb1 gene dosage in tumorigenesis.


Subject(s)
Colonic Neoplasms/genetics , Genes, APC , Mutation , Ovarian Neoplasms/genetics , beta Catenin/metabolism , Animals , Female , Mice
4.
Am J Pathol ; 183(2): 493-503, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23769888

ABSTRACT

We generated transgenic mice in which human CDX2 gene elements control expression of a tamoxifen-regulated Cre protein (CDX2P-CreER(T2)) to allow for inducible gene targeting in intestinal epithelium. After tamoxifen dosing of CDX2P-CreER(T2) mice, Cre activity was detected in the distal ileal, cecal, colonic, and rectal epithelium, with selected crypt base, transit amplifying, and surface cells all capable of activating Cre function. Four weeks after tamoxifen dosing of CDX2P-CreER(T2) mice carrying a Cre-activated fluorescent reporter, single crypts were uniformly fluorescence positive or negative, reflecting Cre activation in crypt stem cells. Biallelic inactivation of the Apc tumor suppressor gene via the CDX2P-CreER(T2) transgene in colon epithelium led to acute alterations in cell proliferation, apoptosis, and morphology, along with mitotic spindle misorientation, ß-catenin nuclear localization, and induction of the intestinal stem cell markers Lgr5 and Musashi-1 and the Sox9 transcription factor. Normal mouse colon epithelium lacks Paneth cells, a key small intestine niche cell type, and Paneth cell differentiation is dependent on Sox9 function. In Apc-deficient colon epithelium, ectopic Paneth-like cells were seen outside the crypt base, such as new crypt budding sites. Our data indicate Apc inactivation via CDX2P-CreER(T2) targeting in mouse colon epithelium is sufficient to induce adenomatous changes and the generation of Paneth-like cells from neoplastic progenitors, with potentially significant roles in colon adenoma development and progression.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Gene Silencing/physiology , Genes, APC/physiology , Homeodomain Proteins/genetics , Paneth Cells/metabolism , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Adenomatous Polyposis Coli/genetics , Animals , CDX2 Transcription Factor , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Spindle Apparatus/physiology , Tamoxifen/pharmacology , Transgenes/genetics
5.
Gastroenterology ; 141(3): 1003-1013.e1-10, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21699772

ABSTRACT

BACKGROUND & AIMS: Adenomatous polyps are precursors to colorectal cancer (CRC), whereas hyperplastic polyps (HPPs) have low risk of progression to CRC. Mutations in KRAS are found in ∼40% of CRCs and large adenomas and a subset of HPPs. We investigated the reasons why HPPs with KRAS mutations lack malignant potential and compared the effects of Kras/KRAS activation with those of Apc/APC inactivation, which promotes adenoma formation. METHODS: We activated a KrasG12D mutant allele or inactivated Apc alleles in mouse colon epithelium and analyzed phenotypes and expression of selected genes and proteins. The mouse data were validated using samples of human HPPs and adenomas. Signaling pathways and factors contributing to Kras/KRAS-induced phenotypes were studied in intestinal epithelial cells. RESULTS: Activation of Kras led to hyperplasia and serrated crypt architecture akin to that observed in human HPPs. We also observed loss of Paneth cells and increases in goblet cell numbers. Abnormalities in Kras-mediated differentiation and proliferation required mitogen-activated protein kinase signaling and were linked to activation of the Hes1 transcription factor. Human HPPs also had activation of HES1. In contrast to Apc/APC inactivation, Kras/KRAS activation did not increase expression of crypt stem cell markers in colon epithelium or colony formation in vitro. Kras/KRAS activation was not associated with substantial induction of p16(INK4a) protein expression in mouse colon epithelium or human HPPs. CONCLUSIONS: Although Kras/KRAS mutation promotes serrated and hyperplastic morphologic features in colon epithelium, it is not able to initiate adenoma development, perhaps in part because activated Kras/KRAS signaling does not increase the number of presumptive stem cells in affected crypts.


Subject(s)
Cell Differentiation/physiology , Colon/pathology , Intestinal Mucosa/pathology , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/physiology , Stem Cells/pathology , Adenoma/pathology , Adenoma/physiopathology , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Line , Cell Proliferation , Colon/physiology , Colonic Polyps/pathology , Colonic Polyps/physiopathology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Disease Models, Animal , Disease Progression , Homeodomain Proteins/physiology , Humans , Hyperplasia , Intestinal Mucosa/physiology , Mice , Mice, Transgenic , Signal Transduction/physiology , Transcription Factor HES-1
6.
J Immunol ; 174(9): 5644-9, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843564

ABSTRACT

Although PGE(2) is a potent inhibitor of fibroblast function, PGE(2) levels are paradoxically elevated in murine lungs undergoing fibrotic responses. Pulmonary fibroblasts from untreated mice expressed all four E prostanoid (EP) receptors for PGE(2). However, following challenge with the fibrogenic agent, bleomycin, fibroblasts showed loss of EP2 expression. Lack of EP2 expression correlated with an inability of fibroblasts from bleomycin-treated mice to be inhibited by PGE(2) in assays of proliferation or collagen synthesis and blunted cAMP elevations in response to PGE(2). PGE(2) was similarly unable to suppress proliferation or collagen synthesis in fibroblasts from EP2(-/-) mice despite expression of the other EP receptors. EP2(-/-), but not EP1(-/-) or EP3(-/-) mice, showed exaggerated fibrotic responses to bleomycin administration in vivo as compared with wild-type controls. EP2 loss on fibroblasts was verified in a second model of pulmonary fibrosis using FITC. Our results for the first time link EP2 receptor loss on fibroblasts following fibrotic lung injury to altered suppression by PGE(2) and thus identify a novel fibrogenic mechanism.


Subject(s)
Bleomycin/toxicity , Dinoprostone/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Growth Inhibitors/pharmacology , Pulmonary Fibrosis/pathology , Receptors, Prostaglandin E/metabolism , Animals , Cell Separation , Cyclic AMP/biosynthesis , Cyclic AMP/metabolism , Dinoprostone/metabolism , Down-Regulation/drug effects , Fibroblasts/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Receptors, Prostaglandin E/deficiency , Receptors, Prostaglandin E/genetics , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...