Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 19(1): 38-51, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27292189

ABSTRACT

The intestinal stem cell niche provides cues that actively maintain gut homeostasis. Dysregulation of these cues may compromise intestinal regeneration upon tissue insult and/or promote tumor growth. Here, we identify secreted phospholipases A2 (sPLA2s) as stem cell niche factors with context-dependent functions in the digestive tract. We show that group IIA sPLA2, a known genetic modifier of mouse intestinal tumorigenesis, is expressed by Paneth cells in the small intestine, while group X sPLA2 is expressed by Paneth/goblet-like cells in the colon. During homeostasis, group IIA/X sPLA2s inhibit Wnt signaling through intracellular activation of Yap1. However, upon inflammation they are secreted into the intestinal lumen, where they promote prostaglandin synthesis and Wnt signaling. Genetic ablation of both sPLA2s improves recovery from inflammation but increases colon cancer susceptibility due to release of their homeostatic Wnt-inhibitory role. This "trade-off" effect suggests sPLA2s have important functions as genetic modifiers of inflammation and colon cancer.


Subject(s)
Group II Phospholipases A2/metabolism , Group X Phospholipases A2/metabolism , Homeostasis , Inflammation/pathology , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/pathology , Intestines/pathology , Stem Cell Niche , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Differentiation , Cell Lineage , Dinoprostone/biosynthesis , Inflammation/enzymology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Neoplasms/genetics , Intracellular Space/metabolism , Mice, Inbred C57BL , Organoids/metabolism , Paneth Cells/enzymology , Paneth Cells/pathology , Phosphoproteins/metabolism , Phosphorylation , Stem Cells/pathology , Wnt Signaling Pathway , YAP-Signaling Proteins
2.
Carcinogenesis ; 35(1): 2-13, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23955540

ABSTRACT

Wnt signaling plays a central role in mammary stem cell (MaSC) homeostasis and in breast cancer. In particular, epigenetic alterations at different members of the Wnt pathway have been identified among triple-negative, basal-like breast cancers. Previously, we developed a mouse model for metaplastic breast adenocarcinoma, a subtype of triple-negative breast cancer, by targeting a hypomorphic mutations in the endogenous Apc gene (Apc (1572T/+)). Here, by employing the CD24 and CD29 cell surface antigens, we have identified a subpopulation of mammary cancer stem cells (MaCSCs) from Apc (1572T/+) capable of self-renewal and differentiation both in vivo and in vitro. Moreover, immunohistochemical analysis of micro- and macrolung metastases and preliminary intravenous transplantation assays suggest that the MaCSCs underlie metastasis at distant organ sites. Expression profiling of the normal and tumor cell subpopulations encompassing MaSCs and CSCs revealed that the normal stem cell compartment is more similar to tumor cells than to their own differentiated progenies. Accordingly, Wnt signaling appears to be active in both the normal and cancer stem cell compartments, although at different levels. By comparing normal with cancer mouse mammary compartments, we identified a MaCSC gene signature able to predict outcome in breast cancer in man. Overall, our data indicate that constitutive Wnt signaling activation affects self-renewal and differentiation of MaSCs leading to metaplasia and basal-like adenocarcinomas.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Wnt Signaling Pathway/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , CD24 Antigen/metabolism , Cell Differentiation , Female , Humans , Integrin beta1/metabolism , Mammary Glands, Animal/cytology , Mice , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Predictive Value of Tests , Reference Values , Transcriptome , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
3.
Carcinogenesis ; 34(11): 2629-38, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23764752

ABSTRACT

Whereas aberrant activation of canonical Wnt/ß-catenin signaling underlies the majority of colorectal cancer cases, the contribution of non-canonical Wnt signaling is unclear. As enhanced expression of the most extensively studied non-canonical Wnt ligand WNT5A is observed in various diseases including colon cancer, WNT5A is gaining attention nowadays. Numerous in vitro studies suggest modulating capacities of WNT5A on proliferation, differentiation, migration and invasion, affecting tumor and non-mutant cells. However, a possible contribution of WNT5A to colorectal cancer remains to be elucidated. We have analyzed WNT5A expression in colorectal cancer profiling data sets, altered WNT5A expression in colon cancer cells and used our inducible Wnt5a transgenic mouse model to gain more insight into the role of WNT5A in intestinal cancer. We observed that increased WNT5A expression is associated with poor prognosis of colorectal cancer patients. WNT5A knockdown in human colon cancer cells caused reduced directional migration, deregulated focal adhesion site formation and reduced invasion, whereas Wnt5a administration promoted the directional migration of colon cancer cells. Despite these observed protumorigenic activities of WNT5A, the induction of Wnt5a expression in intestinal tumors of Apc1638N mice was not sufficient to augment malignancy or metastasis by itself. In conclusion, WNT5A promotes adhesion sites to form in a focal fashion and promotes the directional migration and invasion of colon cancer cells. Although these activities appear insufficient by themselves to augment malignancy or metastasis in Apc1638N mice, they might explain the poor colon cancer prognosis associated with enhanced WNT5A expression.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/pathology , Intestines/pathology , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Focal Adhesions , Humans , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Invasiveness , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Wnt Proteins/genetics , Wnt-5a Protein
4.
Dev Biol ; 369(1): 91-100, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22691362

ABSTRACT

Wnt5a is essential during embryonic development, as indicated by mouse Wnt5a knockout embryos displaying outgrowth defects of multiple structures including the gut. The dynamics of Wnt5a involvement in these processes is unclear, and perinatal lethality of Wnt5a knockout embryos has hampered investigation of Wnt5a during postnatal stages in vivo. Although in vitro studies have suggested a relevant role for Wnt5a postnatally, solid evidence for a significant impact of Wnt5a within the complexity of an adult organism is lacking. We generated a tightly-regulated inducible Wnt5a transgenic mouse model and investigated the effects of Wnt5a induction during different time-frames of embryonic development and in adult mice, focusing on the gastrointestinal tract. When induced in embryos from 10.5 dpc onwards, Wnt5a expression led to severe outgrowth defects affecting the gastrointestinal tracts, limbs, facial structures and tails, closely resembling the defects observed in Wnt5a knockout mice. However, Wnt5a induction from 13.5 dpc onwards did not cause this phenotype, indicating that the most critical period for Wnt5a in embryonic development is prior to 13.5 dpc. In adult mice, induced Wnt5a expression did not reveal abnormalities, providing the first in vivo evidence that Wnt5a has no major impact on mouse intestinal homeostasis postnatally. Protein expression of Wnt5a receptor Ror2 was strongly reduced in adult intestine compared to embryonic stages. Moreover, we uncovered a regulatory process where induction of Wnt5a causes downregulation of its receptor Ror2. Taken together, our results indicate a role for Wnt5a during a restricted time-frame of embryonic development, but suggest no impact during homeostatic postnatal stages.


Subject(s)
Aging/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Intestines/embryology , Wnt Proteins/metabolism , Aging/drug effects , Animals , Cell Lineage/drug effects , Cell Lineage/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Gene Expression Regulation, Developmental/drug effects , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/drug effects , Mice , Mice, Transgenic , Models, Animal , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tetracycline/pharmacology , Wnt Proteins/genetics , Wnt-5a Protein
5.
Gut ; 60(9): 1204-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21307168

ABSTRACT

Objective Deregulation of the Wnt signalling pathway by mutations in the Apc or ß-catenin genes underlies colorectal carcinogenesis. As a result, ß-catenin stabilises, translocates to the nucleus, and activates gene transcription. Intestinal tumours show a heterogeneous pattern of nuclear ß-catenin, with the highest levels observed at the invasion front. Activation of receptor tyrosine kinases in these tumour areas by growth factors expressed by surrounding stromal cells phosphorylate ß-catenin at tyrosine residues, which is thought to increase ß-catenin nuclear translocation and tumour invasiveness. This study investigates the relevance of ß-catenin tyrosine phosphorylation for Wnt signalling and intestinal tumorigenesis in vivo. Design A conditional knock-in mouse model was generated into which the phospho-mimicking Y654E modification in the endogenous ß-catenin gene was introduced. Results This study provided in vivo evidence that ß-catenin(E654) is characterised by reduced affinity for cadherins, increased signalling and strongly increased phosphorylation at serine 675 by protein kinase A (PKA). In addition, homozygosity for the ß-catenin(E654) targeted allele caused embryonic lethality, whereas heterozygosity predisposed to intestinal tumour development, and strongly enhanced Apc-driven intestinal tumour initiation associated with increased nuclear accumulation of ßcatenin. Surprisingly, the expression of ß-catenin(E654) did not affect histological grade or induce tumour invasiveness. Conclusions A thus far unknown mechanism was uncovered in which Y654 phosphorylation of ß-catenin facilitates additional phosphorylation at serine 675 by PKA. In addition, in contrast to the current belief that ß-catenin Y654 phosphorylation increases tumour progression to a more invasive phenotype, these results show that it rather increases tumour initiation by enhancing Wnt signalling.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Wnt Proteins/physiology , beta Catenin/metabolism , Adenoma/genetics , Adenoma/metabolism , Animals , COS Cells , Cadherins/metabolism , Cell Membrane/metabolism , Cell Transformation, Neoplastic/genetics , Chlorocebus aethiops , Colorectal Neoplasms/genetics , Cyclic AMP-Dependent Protein Kinases/pharmacology , Embryo Loss/genetics , Gene Knock-In Techniques , Genes, APC , Genotype , Heterozygote , Homozygote , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/physiology
6.
Hum Mutat ; 31(11): 1205-15, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20725929

ABSTRACT

The MUTYH gene encodes a DNA glycosylase involved in base excision repair (BER). Biallelic pathogenic MUTYH variants have been associated with colorectal polyposis and cancer. The pathogenicity of a few variants is beyond doubt, including c.536A4G/p.Tyr179Cys and c.1187G4A/p.Gly396Asp (previously c.494A4G/p.Tyr165Cys and c.1145G4A/p.Gly382Asp).However, for a substantial fraction of the detected variants, the clinical significance remains uncertain,compromising molecular diagnostics and thereby genetic counseling. We have established an interactive MUTYH gene sequence variant database (www.lovd.nl/MUTYH) with the aim of collecting and sharing MUTYH genotype and phenotype data worldwide. To support standard variant description, we chose NM_001128425.1 as the reference sequence. The database includes records with variants per individual, linked to available phenotype and geographic origin data as well as records with in vitro functional and in silico test data. As of April 2010, the database contains 1968 published and 423 unpublished submitted entries, and 230 and 61 unique variants,respectively. This open-access repository allows all involved to quickly share all variants encountered and communicate potential consequences, which will be especially useful to classify variants of uncertain significance.


Subject(s)
DNA Glycosylases/genetics , Databases, Genetic , Genetic Variation , Adenomatous Polyposis Coli/genetics , Alternative Splicing , Amino Acid Sequence , Base Sequence , DNA/genetics , DNA Glycosylases/chemistry , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Molecular Sequence Data , Mutation , Netherlands , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary
7.
J Pathol ; 221(4): 379-89, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20549647

ABSTRACT

Accumulating evidence has suggested that tumours have a hierarchical organization in which only the cancer stem cells (CSCs) have tumour-initiating properties. Several surface antigens have been employed to isolate CSCs from various malignancies, although not from oesophageal adenocarcinoma (EA). We tested whether Barrett's oesophagus (BE) and EA might serve as a model for the CSC concept. In vivo assays were performed by transplantation of serially diluted bulk EA cells into NOD-SCID mice to establish the presence and frequency of tumour-initiating cells. These were found to be present as ca. 1 in 64 000 cells. The transplanted tumours fully recapitulated the primary lesions. Subsequently, a panel of previously established CSC markers was employed for immunohistochemistry. CD24, CD29 and CD44 showed heterogeneous staining in EA. Nuclear beta-catenin accumulation increased during progression from metaplasia to dysplasia and was often observed in the basal compartment with CD24 and CD29 staining. However, the overall staining patterns were not such to clearly point out specific candidate markers. Accordingly, all markers were employed to sort the corresponding subpopulations of cancer cells and transplant them at low multiplicities in NOD-SCID mice. No increased tumour-initiating capacity of sorted EA cells was observed upon transplantation. These results indicate that tumour-initiating cells are present in EA, thus reflecting a hierarchical organization. However, antibodies directed against novel surface antigens are needed to detect subpopulations enriched for CSCs in EA by transplantation assays.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Esophageal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Adenocarcinoma/metabolism , Animals , Barrett Esophagus/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Progression , Esophageal Neoplasms/metabolism , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Precancerous Conditions/metabolism , Precancerous Conditions/pathology
8.
Acta Histochem ; 109(4): 266-72, 2007.
Article in English | MEDLINE | ID: mdl-17445872

ABSTRACT

Long-standing ulcerative colitis (UC) has been associated with a high risk of developing colonic adenocarcinoma. Importantly, both low- and high-grade dysplasia are strongly related to the presence or development of malignancy. The canonical Wnt/beta-catenin signaling pathway is of crucial importance in cancer development and progression, but its role in UC-related carcinogenesis remains to be determined. We evaluated the immunolabeling patterns of beta-catenin, as well as the products of Wnt-associated cancer genes E-cadherin, cyclin D1 and c-myc, along the dysplasia-carcinoma pathway in UC. For this purpose, immunohistochemistry (IHC) was performed on 18 adenocarcinomas and 17 dysplasias, derived from 21 patients. We found that intracellular beta-catenin accumulation, the hallmark of Wnt signaling activation, is observed in dysplasia, together with enhanced labeling of nuclear protein cyclin D1 and reduction of membranous labeling of E-cadherin. c-myc displayed moderate immunolabeling in the (pre)malignant lesions. Thus, the Wnt pathway is activated in early stages of malignant progression in UC. Furthermore, upregulation of the oncogene cyclin D1 and downregulation of tumor suppressor E-cadherin also occurs in the (pre)neoplastic state. This may contribute to the high potential for malignant degeneration of dysplasia in UC-related colitis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Wnt Proteins/metabolism , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , Colitis, Ulcerative/genetics , Colonic Neoplasms/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Middle Aged , Signal Transduction , Wnt Proteins/genetics
9.
Virchows Arch ; 449(6): 716-21, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17091253

ABSTRACT

Long-standing ulcerative colitis is associated with an elevated risk of developing colonic adenocarcinoma. A very limited group of patients present with multiple synchronous cancers. This could be due to either a multifocal presentation of the same neoplastic clone or different tumors arising in a large area of polyclonal dysplastic colonic mucosa ("field cancerization"). Here, we describe a patient with long-standing colitis and three different tumors in the rectosigmoid part of the large bowel. Clonal evaluation of the lesions was performed by array-based comparative genomic hybridization. These three neoplasms showed a comparable pattern of genomic alterations characterized by gains of chromosomes 12, 13, and 20. Noteworthy, dysplastic mucosa distal to the three cancers displayed a completely different pattern of genomic changes indicating that different cell lineages were present. In addition, all three carcinomas were microsatellite stable and revealed identical immunoprofiles for several cancer-associated genes. We conclude that these three multifocal tumors must have originated from the same preneoplastic lineage.


Subject(s)
Adenocarcinoma/genetics , Colitis, Ulcerative/complications , Colonic Neoplasms/genetics , Adenocarcinoma/etiology , Aged , Aged, 80 and over , Chromosome Aberrations , Colonic Neoplasms/etiology , Female , Humans , Microsatellite Instability , Nucleic Acid Hybridization
10.
Am J Hum Genet ; 72(5): 1088-100, 2003 May.
Article in English | MEDLINE | ID: mdl-12658575

ABSTRACT

The identification of germline mutations in families with HNPCC is hampered by genetic heterogeneity and clinical variability. In previous studies, MSH2 and MLH1 mutations were found in approximately two-thirds of the Amsterdam-criteria-positive families and in much lower percentages of the Amsterdam-criteria-negative families. Therefore, a considerable proportion of HNPCC seems not to be accounted for by the major mismatch repair (MMR) genes. Does the latter result from a lack of sensitivity of mutation detection techniques, or do additional genes underlie the remaining cases? In this study we address these questions by thoroughly investigating a cohort of clinically selected North American families with HNPCC. We analyzed 59 clinically well-defined U.S. families with HNPCC for MSH2, MLH1, and MSH6 mutations. To maximize mutation detection, different techniques were employed, including denaturing gradient gel electrophoresis, Southern analysis, microsatellite instability, immunohistochemistry, and monoallelic expression analysis. In 45 (92%) of the 49 Amsterdam-criteria-positive families and in 7 (70%) of the 10 Amsterdam-criteria-negative families, a mutation was detected in one of the three analyzed MMR genes. Forty-nine mutations were in MSH2 or MLH1, and only three were in MSH6. A considerable proportion (27%) of the mutations were genomic rearrangements (12 in MSH2 and 2 in MLH1). Notably, a deletion encompassing exons 1-6 of MSH2 was detected in seven apparently unrelated families (12% of the total cohort) and was subsequently proven to be a founder. Screening of a second U.S. cohort with HNPCC from Ohio allowed the identification of two additional kindreds with the identical founder deletion. In the present study, we show that optimal mutation detection in HNPCC is achieved by combining accurate and expert clinical selection with an extensive mutation detection strategy. Notably, we identified a common North American deletion in MSH2, accounting for approximately 10% of our cohort. Genealogical, molecular, and haplotype studies showed that this deletion represents a North American founder mutation that could be traced back to the 19th century.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Founder Effect , Gene Deletion , Point Mutation , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Base Sequence , Carrier Proteins , Cohort Studies , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Exons/genetics , Female , Genetic Testing , Haplotypes , Humans , Male , Microsatellite Repeats/genetics , Middle Aged , Molecular Sequence Data , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/genetics , Nuclear Proteins , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...