Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Oncogene ; 36(46): 6480-6489, 2017 11 16.
Article in English | MEDLINE | ID: mdl-28759045

ABSTRACT

Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) is frequently activated in cancers and can be counteracted with the clinical mTORC1 inhibitors everolimus and temsirolimus. Although mTORC1 and dual mTORC1/2 inhibitors are currently under development to treat various malignancies, the emergence of drug resistance has proven to be a major complication. Using the cis-Apc/Smad4 mouse model of locally invasive intestinal adenocarcinoma, we show that administration of everolimus or the dual mTORC1/2 inhibitor AZD8055 significantly reduces the growth of intestinal tumors. In contrast, although everolimus treatment at earlier phase of tumor progression delayed invasion of the tumors, both inhibitors exhibited little effect on blocking invasion of the tumors when administered later in their progression. Biochemical and immunohistochemical analyses revealed that treatment of cis-Apc/Smad4 mice with everolimus or AZD8055 induced marked increases in epidermal growth factor receptor (EGFR) and MEK/ERK signaling in tumor epithelial and stromal cells, respectively. Notably, co-administration of AZD8055 and the EGFR inhibitor erlotinib or the MEK inhibitor trametinib was sufficient to suppress tumor invasion in cis-Apc/Smad4 mice. These data indicate that mTOR inhibitor resistance in invasive intestinal tumors involves feedback signaling from both cancer epithelial and stromal cells, highlighting the role of tumor microenvironment in drug resistance, and support that simultaneous inhibition of mTOR and EGFR or MEK may be more effective in treating colon cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Intestinal Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Microenvironment/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Everolimus/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , Mice, 129 Strain , Mice, Inbred C57BL , Morpholines/pharmacology , Neoplasm Invasiveness , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
2.
Oncogene ; 36(42): 5885-5896, 2017 10 19.
Article in English | MEDLINE | ID: mdl-28628120

ABSTRACT

Tumor suppressor TP53 is frequently mutated in colorectal cancer (CRC), and most mutations are missense type. Although gain-of-functions by mutant p53 have been demonstrated experimentally, the precise mechanism for malignant progression in in vivo tumors remains unsolved. We generated ApcΔ716 Trp53LSL•R270H villin-CreER compound mice, in which mutant p53R270H was expressed in the intestinal epithelia upon tamoxifen treatment, and examined the intestinal tumor phenotypes and tumor-derived organoids. Mutant Trp53R270H, but not Trp53-null mutation accelerated submucosal invasion with generation of desmoplastic microenvironment. The nuclear accumulation of p53 was evident in ApcΔ716 Trp53R270H/R270H homozygous tumors like human CRC. Although p53 was distributed to the cytoplasm in ApcΔ716 Trp53+/R270H heterozygous tumors, it accumulated in the nuclei at the invasion front, suggesting a regulation mechanism for p53 localization by the microenvironment. Importantly, mutant p53 induced drastic morphological changes in the tumor organoids to complex glandular structures, which was associated with the acquisition of invasiveness. Consistently, the branching scores of human CRC that carry TP53 mutations at codon 273 significantly increased in comparison with those of TP53 wild-type tumors. Moreover, allografted ApcΔ716 Trp53R270H/R270H organoid tumors showed a malignant histology with an increased number of myofibroblasts in the stroma. These results indicate that nuclear-accumulated mutant p53R270H induces malignant progression of intestinal tumors through complex tumor gland formation and acquisition of invasiveness. Furthermore, RNA sequencing analyses revealed global gene upregulation by mutant p53R270H, which was associated with the activation of inflammatory and innate immune pathways. Accordingly, it is possible that mutant p53R270H induces CRC progression, not only by a cell intrinsic mechanism, but also by the generation or activation of the microenvironment, which may synergistically contribute to the acceleration of submucosal invasion. Therefore, the present study indicates that nuclear-accumulated mutant p53R270H is a potential therapeutic target for the treatment of advanced CRCs.


Subject(s)
Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/pathology , Liver Neoplasms/secondary , Mutation , Tumor Suppressor Protein p53/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Disease Progression , Gene Expression Profiling , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Invasiveness , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Microenvironment , Tumor Suppressor Protein p53/metabolism
3.
Cell Death Dis ; 7: e2136, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26962686

ABSTRACT

Recent studies have reported the role of Wnt/ß-catenin signaling in hair cell (HC) development, regeneration, and differentiation in the mouse cochlea; however, the role of Wnt/ß-catenin signaling in HC protection remains unknown. In this study, we took advantage of transgenic mice to specifically knockout or overactivate the canonical Wnt signaling mediator ß-catenin in HCs, which allowed us to investigate the role of Wnt/ß-catenin signaling in protecting HCs against neomycin-induced damage. We first showed that loss of ß-catenin in HCs made them more vulnerable to neomycin-induced injury, while constitutive activation of ß-catenin in HCs reduced HC loss both in vivo and in vitro. We then showed that loss of ß-catenin in HCs increased caspase-mediated apoptosis induced by neomycin injury, while ß-catenin overexpression inhibited caspase-mediated apoptosis. Finally, we demonstrated that loss of ß-catenin in HCs led to increased expression of forkhead box O3 transcription factor (Foxo3) and Bim along with decreased expression of antioxidant enzymes; thus, there were increased levels of reactive oxygen species (ROS) after neomycin treatment that might be responsible for the increased aminoglycoside sensitivity of HCs. In contrast, ß-catenin overexpression reduced Foxo3 and Bim expression and ROS levels, suggesting that ß-catenin is protective against neomycin-induced HC loss. Our findings demonstrate that Wnt/ß-catenin signaling has an important role in protecting HCs against neomycin-induced HC loss and thus might be a new therapeutic target for the prevention of HC death.


Subject(s)
Hair Cells, Auditory/metabolism , Hearing Loss/prevention & control , Neomycin/adverse effects , Wnt Signaling Pathway/drug effects , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Death , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Hair Cells, Auditory/pathology , Hearing Loss/chemically induced , Hearing Loss/genetics , Hearing Loss/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Neomycin/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
4.
J Periodontal Res ; 48(4): 405-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23050778

ABSTRACT

BACKGROUND AND OBJECTIVE: Wingless-type MMTV integration site family (Wnt)/ß-catenin signaling plays an essential role in cellular differentiation and matrix formation during skeletal development. However, little is known about its role in tooth-root formation. In a previous study, we found excessive formation of dentin and cementum in mice with constitutive ß-catenin stabilization in the dental mesenchyme. In the present study we analyzed the molar roots of these mice to investigate the role of Wnt/ß-catenin signaling in root formation in more detail. MATERIAL AND METHODS: We generated OC-Cre:Catnb(+/lox(ex3)) mice by intercrossing Catnb(+/lox(ex3)) and OC-Cre mice, and we analyzed their mandibular molars using radiography, histomorphometry and immunohistochemistry. RESULTS: OC-Cre:Catnb(+/lox(ex3)) mice showed impaired root formation. At the beginning of root formation in mutant molars, dental papilla cells did not show normal differentiation into odontoblasts; rather, they were prematurely differentiated and had a disorganized arrangement. Interestingly, SMAD family member 4 was upregulated in premature odontoblasts. In 4-wk-old mutant mice, molar roots were about half the length of those in their wild-type littermates. In contrast to excessively formed dentin in crown, root dentin was thin and hypomineralized in mutant mice. Biglycan and dentin sialophosphoprotein were downregulated in root dentin of mutant mice, whereas dentin matrix protein 1 and Dickkopf-related protein 1 were upregulated. Additionally, ectonucleotide pyrophosphatase/phosphodiesterase 1 was significantly downregulated in the cementoblasts of mutant molars. Finally, in the cementum of mutant mice, bone sialoprotein was downregulated but Dickkopf-related protein 2 was upregulated. CONCLUSION: These results suggest that temporospatial regulation of Wnt/ß-catenin signaling plays an important role in cell differentiation and matrix formation during root and cementum formation.


Subject(s)
Odontogenesis/physiology , Tooth Root/growth & development , Wnt Signaling Pathway/physiology , Animals , Biglycan/analysis , Cell Differentiation/physiology , Cell Polarity/physiology , Cementogenesis/physiology , Dental Cementum/pathology , Dental Papilla/pathology , Dentin/pathology , Dentinogenesis/physiology , Down-Regulation , Extracellular Matrix Proteins/analysis , Integrin-Binding Sialoprotein/analysis , Intercellular Signaling Peptides and Proteins/analysis , Mice , Mice, Mutant Strains , Molar/growth & development , Mutation/genetics , Odontoblasts/pathology , Phosphoproteins/analysis , Phosphoric Diester Hydrolases , Pyrophosphatases , Sialoglycoproteins/analysis , Signal Transduction/physiology , Smad4 Protein/analysis , Up-Regulation , Wnt Signaling Pathway/genetics
5.
Cell Death Dis ; 2: e124, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368895

ABSTRACT

Mutations in the Ras family of proteins (predominantly in H-Ras) occur in approximately 40% of urothelial cell carcinoma (UCC). However, relatively little is known about subsequent mutations/pathway alterations that allow tumour progression. Indeed, expressing mutant H-Ras within the mouse bladder does not lead to tumour formation, unless this is expressed at high levels. The Wnt signalling pathway is deregulated in approximately 25% of UCC, so we examined if this correlated with the activation of MAPK signalling in human UCC and found a significant correlation. To test the functional significance of this association we examined the impact of combining Ras mutation (H-Ras(Q61L) or K-Ras(G12D)) with an activating ß-catenin mutation within the mouse bladder using Cre-LoxP technology. Although alone, neither Ras mutation nor ß-catenin activation led to UCC (within 12 months), mice carrying both mutations rapidly developed UCC. Mechanistically this was associated with reduced levels of p21 with dependence on the MAPK signalling pathway. Moreover, tumours from these mice were sensitive to MEK inhibition. Importantly, in human UCC there was a negative correlation between levels of p-ERK and p21 suggesting that p21 accumulation may block tumour progression following Ras mutation. Taken together these data definitively show Ras pathway activation strongly cooperates with Wnt signalling to drive UCC in vivo.


Subject(s)
Mutation , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , beta Catenin/metabolism , ras Proteins/genetics , Animals , Female , Humans , Male , Mice , Mice, Transgenic , Neoplastic Processes , Signal Transduction , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , ras Proteins/metabolism
6.
Oncogene ; 30(2): 178-89, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20818428

ABSTRACT

Although deregulation of the Wnt signalling pathway has been implicated in urothelial cell carcinoma (UCC), the functional significance is unknown. To test its importance, we have targeted expression of an activated form of ß-catenin to the urothelium of transgenic mice using Cre-Lox technology (UroIICRE(+) ß-catenin(exon3/+)). Expression of this activated form of ß-catenin led to the formation of localized hyperproliferative lesions by 3 months, which did not progress to malignancy. These lesions were characterized by a marked increase of the phosphatase and tensin homologue (PTEN) tumour suppressor protein. This appears to be a direct consequence of activating Wnt signalling in the bladder as conditional deletion of the adenomatous polyposis coli (Apc) gene within the adult bladder led rapidly to coincident ß-catenin and PTEN expression. This PTEN expression blocked proliferation. Next, we combined PTEN deficiency with ß-catenin activation and found that this caused papillary UCC. These tumours had increased pAKT signalling and were dependent on mammalian target of rapamycin (mTOR). Importantly, in human UCC, there was a significant correlation between high levels of ß-catenin and pAKT (and low levels of PTEN). Taken together these data show that deregulated Wnt signalling has a critical role in promoting UCC, and suggests that human UCC that have high levels of Wnt and PI3 kinase signalling may be responsive to mTOR inhibition.


Subject(s)
Carcinoma, Papillary/metabolism , Cell Transformation, Neoplastic/metabolism , PTEN Phosphohydrolase/metabolism , Urinary Bladder Neoplasms/chemically induced , beta Catenin/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Proliferation , Cohort Studies , Female , Humans , Hyperplasia/chemically induced , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/metabolism , Sequence Deletion , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder/metabolism , Urothelium/metabolism , Wnt Proteins/metabolism
7.
J Dent Res ; 89(9): 909-14, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20530729

ABSTRACT

beta-Catenin signaling is required for embryonic tooth morphogenesis and promotes continuous tooth development when activated in embryos. To determine whether activation of this pathway in the adult oral cavity could promote tooth development, we induced mutation of epithelial beta-catenin to a stabilized form in adult mice. This caused increased proliferation of the incisor tooth cervical loop, outpouching of incisor epithelium, abnormal morphology of the epithelial-mesenchymal junction, and enhanced expression of genes associated with embryonic tooth development. Ectopic dental-like structures were formed from the incisor region following implantation into immunodeficient mice. Thus, forced activation of beta-catenin signaling can initiate an embryonic-like program of tooth development in adult rodent incisor teeth.


Subject(s)
Adult Stem Cells/physiology , Dental Papilla/cytology , Enamel Organ/cytology , Odontogenesis/genetics , beta Catenin/physiology , Animals , Epithelial Cells/cytology , Female , Fibroblast Growth Factor 8/biosynthesis , Fibroblast Growth Factor 8/genetics , Incisor/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Nude , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Signal Transduction , Tooth Calcification , Up-Regulation
9.
Oncogene ; 28(1): 31-40, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-18806829

ABSTRACT

Endometrioid adenocarcinoma is the most frequent form of endometrial cancer, usually developing in pre- and peri-menopausal women. beta-catenin abnormalities are common in endometrioid type endometrial carcinomas with squamous differentiation. To investigate the role of beta-catenin (Ctnnb1) in uterine development and tumorigenesis, mice were generated which expressed a dominant stabilized beta-catenin or had beta-catenin conditionally ablated in the uterus by crossing the PR(Cre) mouse with the Ctnnb1(f(ex3)/+) mouse or Ctnnb1(f/f) mouse, respectively. Both of the beta-catenin mutant mice have fertility defects and the ability of the uterus to undergo a hormonally induced decidual reaction was lost. Expression of the dominant stabilized beta-catenin, PR(cre/+)Ctnnb1(f(ex3)/+), resulted in endometrial glandular hyperplasia, whereas ablation of beta-catenin, PR(cre/+)Ctnnb1(f/f), induced squamous cell metaplasia in the murine uterus. Therefore, we have demonstrated that correct regulation of beta-catenin is important for uterine function as well as in the regulation of endometrial epithelial differentiation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Endometrial Hyperplasia/genetics , Endometrium/growth & development , beta Catenin/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Endometrial Hyperplasia/pathology , Endometrium/pathology , Female , Infertility, Female/genetics , Mice , Mice, Mutant Strains , beta Catenin/genetics
10.
Oncogene ; 26(32): 4679-88, 2007 Jul 12.
Article in English | MEDLINE | ID: mdl-17297455

ABSTRACT

Chemokines and their receptors are essential for leukocyte trafficking, and also implicated in cancer metastasis to specific organs. We have recently demonstrated that CXCR3 plays a critical role in metastasis of mouse melanoma cells to lymph nodes. Here, we show that some human colon cancer cell lines express CXCR3 constitutively. We constructed cells that expressed CXCR3 cDNA ('DLD-1-CXCR3'), and compared with nonexpressing controls by rectal transplantation in nude mice. Although both cell lines disseminated to lymph nodes at similar frequencies at 2 weeks, DLD-1-CXCR3 expanded more rapidly than the control in 4 weeks. In 6 weeks, 59% of mice inoculated with DLD1-CXCR3 showed macroscopic metastasis in para-aortic lymph nodes, whereas only 14% of those with the control (P<0.05). In contrast, metastasis to the liver or lung was rare, and unaffected by CXCR3 expression. In clinical colon cancer samples, we found expression of CXCR3 in 34% cases, most of which had lymph node metastasis. Importantly, patients with CXCR3-positive cancer showed significantly poorer prognosis than those without CXCR3, or those expressing CXCR4 or CCR7. These results indicate that activation of CXCR3 with its ligands stimulates colon cancer metastasis preferentially to the draining lymph nodes with poorer prognosis.


Subject(s)
Colonic Neoplasms/pathology , Lymph Nodes/pathology , Receptors, Chemokine/physiology , Animals , Cell Movement , DNA, Complementary/genetics , Humans , Ligands , Lymphatic Metastasis , Mice , Mice, Nude , Neoplasm Transplantation , Receptors, CXCR3 , Receptors, Chemokine/genetics
11.
Oncogene ; 26(24): 3511-20, 2007 May 24.
Article in English | MEDLINE | ID: mdl-17160019

ABSTRACT

Adenomatous polyposis coli (APC/Apc) gene encodes a key tumor suppressor whose mutations activate beta-catenin/T-cell factor (TCF)-mediated transcription (canonical Wnt signaling). Here, we show that Wnt signaling can cause chromosomal instability (CIN). As an indicator of CIN, we scored anaphase bridge index (ABI) in mouse polyps and ES cells where Wnt signaling was activated by Apc or beta-catenin mutations. We found three to nine times higher ABI than in wild-type controls. Furthermore, karyotype analysis confirmed that the Wnt signal-activated ES cells produced new chromosomal aberrations at higher rates; hence CIN. Consistently, expression of dominant-negative TCFs in these cells reduced their ABI. We also found that Wnt signal activation increased phosphorylation of Cdc2 (Cdk1) that inhibited its activity, and suppressed apoptosis upon exposure of the cells to nocodazole or colcemid. The data suggest that Wnt signaling stimulates the cells to escape from mitotic arrest and apoptosis, resulting in CIN. In human gastric cancer tissues with nuclear beta-catenin, ABI was significantly higher than in those without. These results collectively indicate that beta-catenin/TCF-mediated transcription itself increases CIN through dysregulation of G2/M progression.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Chromosomal Instability , Mutation , TCF Transcription Factors/genetics , beta Catenin/genetics , Adenoma/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cell Division/genetics , Cell Survival/genetics , Cells, Cultured , Chromosome Aberrations , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Embryonic Stem Cells , G2 Phase/genetics , Humans , Intestinal Neoplasms/genetics , Intestinal Polyps/genetics , Mice , Microtubules/metabolism , Signal Transduction , TCF Transcription Factors/metabolism , Transcription, Genetic , Wnt Proteins/metabolism , beta Catenin/metabolism
12.
Oncogene ; 25(57): 7522-30, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17143296

ABSTRACT

The canonical Wnt signaling plays important roles in embryonic development and tumorigenesis. For the latter, induced mutations in mice have greatly contributed to our understanding of the molecular mechanisms of cancer initiation and progression. Here, I will review recent reports on gastrointestinal cancer model mice, with an emphasis on the roles of the Wnt signal pathway. They include: mouse models for familial adenomatous polyposis; modifying factors that affect mouse intestinal polyposis, including the genes that help cancer progression; Wnt target genes that affect mouse intestinal polyposis; and a mouse model of gastric cancer that mimics Helicobacter pyroli infection.


Subject(s)
Gastrointestinal Neoplasms/metabolism , Wnt Proteins/physiology , Adenomatous Polyposis Coli/genetics , Animals , Disease Models, Animal , Disease Progression , Gastrointestinal Neoplasms/genetics , Humans , Mice , Mice, Transgenic , Models, Biological , Mutation , Signal Transduction , Wnt Proteins/genetics
13.
Oncogene ; 25(12): 1816-20, 2006 Mar 16.
Article in English | MEDLINE | ID: mdl-16278673

ABSTRACT

Germline mutations in the LKB1 gene are responsible for Peutz-Jeghers syndrome (PJS), which is characterized by gastrointestinal hamartomas and increasing risk of cancer. Mice with Lkb1(+/-) mutation develop gastric hamartomas after >20 weeks of age, and hepatocellular adenomas and carcinomas >30 weeks. It has been reported that, in PJS patients, carcinomas progressed from hamartomas contain p53 mutations, and that LKB1 regulates p53-dependent apoptosis. To investigate the roles of LKB1 and p53 mutations in tumorigenesis, we constructed compound mutant mice of Lkb1 and p53 genes. In the Lkb1(+/-)p53(-/-) mice, formation of gastric hamartomas and hepatic tumors was accelerated. However, histopathology of hamartomas was similar between Lkb1(+/-)p53(-/-) and Lkb1(+/-) mice, and Lkb1 genotype remained heterozygous, suggesting that the p53 mutation affected hamartoma initiation. Contrary to the heterozygous hamartomas in the stomach and duodenum, the hepatic adenomas in Lkb1(+/-)p53(-/-) mice showed loss of Lkb1 heterozygosity (LOH), suggesting that lack of p53 stimulated Lkb1 LOH and tumor initiation in the liver. Taken together, these results indicate that lack of p53 causes earlier onsets of gastric hamartomas and hepatic tumors in Lkb1(+/-)p53(-/-) mice.


Subject(s)
Adenoma/genetics , Hamartoma/genetics , Liver Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Stomach Diseases/genetics , Tumor Suppressor Protein p53/genetics , AMP-Activated Protein Kinases , Animals , Blotting, Western , Disease Models, Animal , Female , Loss of Heterozygosity , Male , Mice , Mice, Transgenic , Peutz-Jeghers Syndrome/genetics
14.
Gut ; 55(1): 1-5, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16344569

ABSTRACT

Germline mutations in LKB1 cause the rare cancer prone disorder Peutz-Jeghers syndrome (PJS). Gastrointestinal hamartomatous polyps constitute the major phenotypic trait in PJS. Hamartomatous polyps arising in PJS patients are generally considered to lack premalignant potential although rare neoplastic changes in these polyps and an increased gastrointestinal cancer risk in PJS are well documented. These conflicting observations are resolved in the current hypothesis by providing a unifying explanation for these contrasting features of PJS polyposis. We postulate that a genetic predisposition to epithelial prolapse underlies the formation of the polyps associated with PJS. Conventional sporadic adenomas arising in PJS patients will similarly show mucosal prolapse and carry the associated histological features.


Subject(s)
Intestinal Mucosa/pathology , Peutz-Jeghers Syndrome/genetics , AMP-Activated Protein Kinase Kinases , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Prolapse , Protein Serine-Threonine Kinases/genetics
15.
Br J Cancer ; 91(8): 1571-4, 2004 Oct 18.
Article in English | MEDLINE | ID: mdl-15467769

ABSTRACT

The fragile histidine triad (FHIT) gene is frequently inactivated in various types of tumours. However, the system-wide pathology caused by FHIT inactivation has not been examined in detail. Here we demonstrate that Fhit gene knockout mice develop tumours in the lymphoid tissue, liver, uterus, testis, forestomach and small intestine, together with structural abnormalities in the small intestinal mucosa. These results suggest that Fhit plays important roles in systemic tumour suppression and in the integrity of mucosal structure of the intestines.


Subject(s)
Acid Anhydride Hydrolases/genetics , Genes, Tumor Suppressor/physiology , Intestinal Neoplasms/pathology , Lymphoma/pathology , Neoplasm Proteins/genetics , Stomach Neoplasms/pathology , Testicular Neoplasms/pathology , Uterine Neoplasms/pathology , Animals , Female , Intestine, Small/abnormalities , Intestine, Small/pathology , Male , Mice , Mice, Knockout , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology
16.
Br J Cancer ; 90(3): 701-4, 2004 Feb 09.
Article in English | MEDLINE | ID: mdl-14760387

ABSTRACT

Cyclo-oxygenase (COX)-2 is induced in various types of cancer tissues. Here, we demonstrate stromal expression of both COX-2 and microsomal prostaglandin E(2) synthase (mPGES)-1 in gastrointestinal hamartomas developed in Lkb1(+/-), Smad4(+/-) and Cdx2(+/-)mice. These results suggest that PGE(2) produced by COX-2 and mPGES-1 plays an important role in hamartoma development regardless of the mutated genes causing hamartomas.


Subject(s)
Carrier Proteins , Hamartoma/genetics , Intramolecular Oxidoreductases/biosynthesis , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Stomach Diseases/genetics , AMP-Activated Protein Kinases , Adaptor Proteins, Signal Transducing , Animals , CDX2 Transcription Factor , Cyclooxygenase 2 , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Hamartoma/pathology , Hamartoma/veterinary , Homeodomain Proteins/genetics , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Intramolecular Oxidoreductases/genetics , Isoenzymes/genetics , Mice , Peroxidases/biosynthesis , Prostaglandin-E Synthases , Prostaglandin-Endoperoxide Synthases/genetics , Protein Serine-Threonine Kinases , Signal Transduction , Smad4 Protein , Stomach Diseases/pathology , Stomach Diseases/veterinary , Trans-Activators/genetics , Transforming Growth Factor beta
17.
Int J Oncol ; 21(4): 769-74, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12239615

ABSTRACT

CDX2, a transcriptional factor expressed in the intestine, is implicated in the development and maintenance of the intestinal mucosa. Recent studies have demonstrated that CDX2 is expressed in the intestinal metaplasia of the stomach and intestinal-type gastric cancer, while it is not expressed in the normal gastric mucosa. To investigate the role of CDX2 in gastric cancer, we determined CDX2 expression and cell proliferation rate in various types of gastric cancer tissues by immunostaining. Surgically dissected gastric cancer tissues were collected from 40 patients. Consistent with previous reports, CDX2 was expressed in most gastric mucosa samples with intestinal metaplasia (89%, 16/18), although it was not found in the adjacent normal mucosa. CDX2 expression was also detected in 64% (18/28) of intestinal-type gastric cancer cases, whereas it was not observed in the diffuse-type gastric cancer (0/12). Moreover, the CDX2-positive gastric cancer samples showed significantly lower index for Ki-67 immunostaining, indicating reduced cell proliferation rates than in the CDX2-negative samples. Importantly, multivariate analysis for the overall survival rate revealed that the CDX2-positive gastric cancer patients survived significantly longer than the CDX2-negative patients. Even among the intestinal-type gastric cancer cases, the CDX2-positive group showed a lower Ki-67 index and longer postoperative survival than the CDX2-negative group. These results collectively indicate that CDX2 expression in gastric cancer tissues can be a novel prognostic marker for patient survival.


Subject(s)
Homeodomain Proteins/biosynthesis , Intestines/pathology , Metaplasia/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Aged , Aged, 80 and over , CDX2 Transcription Factor , Cell Division , Female , Humans , Immunohistochemistry , Ki-67 Antigen/biosynthesis , Male , Metaplasia/mortality , Middle Aged , Multivariate Analysis , Prognosis , Stomach Neoplasms/mortality , Time Factors , Trans-Activators
18.
Nat Med ; 7(9): 1048-51, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533709

ABSTRACT

Arachidonic acid is metabolized to prostaglandin H(2) (PGH(2)) by cyclooxygenase (COX). COX-2, the inducible COX isozyme, has a key role in intestinal polyposis. Among the metabolites of PGH(2), PGE(2) is implicated in tumorigenesis because its level is markedly elevated in tissues of intestinal adenoma and colon cancer. Here we show that homozygous deletion of the gene encoding a cell-surface receptor of PGE(2), EP2, causes decreases in number and size of intestinal polyps in Apc(Delta 716) mice (a mouse model for human familial adenomatous polyposis). This effect is similar to that of COX-2 gene disruption. We also show that COX-2 expression is boosted by PGE(2) through the EP2 receptor via a positive feedback loop. Homozygous gene knockout for other PGE(2) receptors, EP1 or EP3, did not affect intestinal polyp formation in Apc(Delta 716) mice. We conclude that EP2 is the major receptor mediating the PGE2 signal generated by COX-2 upregulation in intestinal polyposis, and that increased cellular cAMP stimulates expression of more COX-2 and vascular endothelial growth factor in the polyp stroma.


Subject(s)
Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/physiopathology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Prostaglandin E/genetics , Adenoma/genetics , Adenoma/pathology , Adenomatous Polyposis Coli/pathology , Animals , Cyclooxygenase 2 , Dinoprostone/metabolism , Endothelial Growth Factors/genetics , Homozygote , Isoenzymes/genetics , Lymphokines/genetics , Mice , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Nat Immunol ; 2(9): 863-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11526403

ABSTRACT

Mutation or ablation of T cell factor 1 and lymphocyte enhancer factor 1 indicated involvement of the Wnt pathway in thymocyte development. The central effector of the Wnt pathway is beta-catenin, which undergoes stabilization upon binding of Wnt ligands to frizzled receptors. We report here that conditional stabilization of beta-catenin in immature thymocytes resulted in the generation of single positive T cells that lacked the alpha beta TCR and developed in the absence of pre-TCR signaling and TCR selection. Although active beta-catenin induced differentiation in the absence of TCRs, its action was associated with reduced proliferation and survival when compared to developmental changes induced by the pre-TCR or the alpha beta TCR.


Subject(s)
Cytoskeletal Proteins/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction , T-Lymphocytes/immunology , Thymus Gland/immunology , Trans-Activators , Animals , Antigens, Differentiation, T-Lymphocyte/analysis , Apoptosis , Cell Cycle , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Exons , Flow Cytometry , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell, alpha-beta/analysis , Sequence Deletion , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , beta Catenin
20.
Cancer Res ; 61(4): 1733-40, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245490

ABSTRACT

Mutations in the human adenomatous polyposis (APC) gene are causative for familial adenomatous polyposis (FAP), a rare condition in which numerous colonic polyps arise during puberty and, if left untreated, lead to colon cancer. The APC gene is a tumor suppressor that has been termed the "gatekeeper gene" for colon cancer. In addition to the 100% mutation rate in FAP patients, the APC gene is mutated in >80% of sporadic colon and intestinal cancers. The Apc gene in mice has been mutated either by chemical carcinogenesis, resulting in the Min mouse Apcdelta850, or by heterologous recombination, resulting in the Apcdelta716 or Apedelta1368 mice (M. Oshima et al., Proc. Natl. Acad. Sci. USA, 92: 4482-4486, 1995). Although homozygote Apc-/- mice are embryonically lethal, the heterozygotes are viable but develop numerous intestinal polyps with loss of Apc heterozygosity within the polyps (M. Oshima et al., Proc. Natl. Acad. Sci. USA, 92: 4482-4486, 1995). The proinflammatory, prooncogenic protein cyclooxygenase (COX)-2 has been shown to be markedly induced in the Apcdelta716 polyps at an early stage of polyp development (M. Oshima et al., Cell, 87: 803-809, 1996). We demonstrate here that treatment with the specific COX-2 inhibitor rofecoxib results in a dose-dependent reduction in the number and size of intestinal and colonic polyps in the Apcdelta716 mouse. The plasma concentration of rofecoxib that resulted in a 55% inhibition of polyp number and an 80% inhibition of polyps > 1 mm in size is comparable with the human clinical steady-state concentration of 25 mg rofecoxib (Vioxx) taken once daily (A. Porras et al., Clin. Pharm. Ther., 67: 137, 2000). Polyps from both untreated and rofecoxib- or sulindac-treated Apcdelta716 mice expressed COX-1 and -2, whereas normal epithelium from all mice expressed COX-1 but minimal amounts of COX-2. Polyps from either rofecoxib- or sulindac-treated mice had lower rates of DNA replication, expressed less proangiogenic vascular endothelial-derived growth factor and more membrane-bound beta-catenin, but showed unchanged nuclear localization of this transcription factor. This study showing the inhibition of polyposis in the Apcdelta716 mouse suggests that the specific COX-2 inhibitor rofecoxib (Vioxx) has potential as a chemopreventive agent in human intestinal and colon cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Genes, APC/genetics , Intestinal Neoplasms/prevention & control , Intestinal Polyps/prevention & control , Isoenzymes/antagonists & inhibitors , Lactones/pharmacology , Trans-Activators , Animals , Anticarcinogenic Agents/pharmacokinetics , Cell Nucleus/metabolism , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacokinetics , Cytoskeletal Proteins/metabolism , DNA Replication/drug effects , Dose-Response Relationship, Drug , Female , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Intestinal Polyps/enzymology , Intestinal Polyps/genetics , Isoenzymes/biosynthesis , Lactones/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/biosynthesis , Sulfones , Sulindac/analogs & derivatives , Sulindac/pharmacokinetics , Sulindac/pharmacology , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...