Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Nucleic Acids Res ; 52(D1): D1210-D1217, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38183204

ABSTRACT

The Catalogue Of Somatic Mutations In Cancer (COSMIC), https://cancer.sanger.ac.uk/cosmic, is an expert-curated knowledgebase providing data on somatic variants in cancer, supported by a comprehensive suite of tools for interpreting genomic data, discerning the impact of somatic alterations on disease, and facilitating translational research. The catalogue is accessed and used by thousands of cancer researchers and clinicians daily, allowing them to quickly access information from an immense pool of data curated from over 29 thousand scientific publications and large studies. Within the last 4 years, COSMIC has substantially expanded its utility by adding new resources: the Mutational Signatures catalogue, the Cancer Mutation Census, and Actionability. To improve data accessibility and interoperability, somatic variants have received stable genomic identifiers that are associated with their genomic coordinates in GRCh37 and GRCh38, and new export files with reduced data redundancy have been made available for download.


Subject(s)
Databases, Genetic , Genomics , Neoplasms , Humans , Databases, Factual , Knowledge Bases , Mutation , Neoplasms/genetics , Databases, Genetic/trends , Internet
2.
BMC Bioinformatics ; 23(1): 457, 2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36324085

ABSTRACT

BACKGROUND: Cancer evolution consists of a stepwise acquisition of genetic and epigenetic changes, which alter the gene expression profiles of cells in a particular tissue and result in phenotypic alterations acted upon by natural selection. The recurrent appearance of specific genetic lesions across individual cancers and cancer types suggests the existence of certain "driver mutations," which likely make up the major contribution to tumors' selective advantages over surrounding normal tissue and as such are responsible for the most consequential aspects of the cancer cells' gene expression patterns and phenotypes. We hypothesize that such mutations are likely to cluster with specific dichotomous shifts in the expression of the genes they most closely control, and propose GMMchi, a Python package that leverages Gaussian Mixture Modeling to detect and characterize bimodal gene expression patterns across cancer samples, as a tool to analyze such correlations using 2 × 2 contingency table statistics. RESULTS: Using well-defined simulated data, we were able to confirm the robust performance of GMMchi, reaching 85% accuracy with a sample size of n = 90. We were also able to demonstrate a few examples of the application of GMMchi with respect to its capacity to characterize background florescent signals in microarray data, filter out uninformative background probe sets, as well as uncover novel genetic interrelationships and tumor characteristics. Our approach to analysing gene expression analysis in cancers provides an additional lens to supplement traditional continuous-valued statistical analysis by maximizing the information that can be gathered from bulk gene expression data. CONCLUSIONS: We confirm that GMMchi robustly and reliably extracts bimodal patterns from both colorectal cancer (CRC) cell line-derived microarray and tumor-derived RNA-Seq data and verify previously reported gene expression correlates of some well-characterized CRC phenotypes. AVAILABILITY: The Python package GMMchi and our cell line microarray data used in this paper is available for downloading on GitHub at https://github.com/jeffliu6068/GMMchi .


Subject(s)
Algorithms , Gene Expression Profiling , Gene Expression Profiling/methods , Cluster Analysis , Normal Distribution , Transcriptome
3.
Cells ; 8(3)2019 03 22.
Article in English | MEDLINE | ID: mdl-30909444

ABSTRACT

The caudal-related homeobox protein 1 (CDX1) is a transcription factor, which is important in the development, differentiation, and homeostasis of the gut. Although the involvement of CDX genes in the regulation of the expression levels of a few glycosyltransferases has been shown, associations between glycosylation phenotypes and CDX1 mRNA expression have hitherto not been well studied. Triggered by our previous study, we here characterized the N-glycomic phenotype of 16 colon cancer cell lines, selected for their differential CDX1 mRNA expression levels. We found that high CDX1 mRNA expression associated with a higher degree of multi-fucosylation on N-glycans, which is in line with our previous results and was supported by up-regulated gene expression of fucosyltransferases involved in antenna fucosylation. Interestingly, hepatocyte nuclear factors (HNF)4A and HNF1A were, among others, positively associated with high CDX1 mRNA expression and have been previously proven to regulate antenna fucosylation. Besides fucosylation, we found that high CDX1 mRNA expression in cancer cell lines also associated with low levels of sialylation and galactosylation and high levels of bisection on N-glycans. Altogether, our data highlight a possible role of CDX1 in altering the N-glycosylation of colorectal cancer cells, which is a hallmark of tumor development.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glycomics , Homeodomain Proteins/genetics , Transcriptome/genetics , Cell Line, Tumor , Fucose/metabolism , Glycosylation , Hexosamines/metabolism , Homeodomain Proteins/metabolism , Humans , Lewis Blood Group Antigens/chemistry , Lewis Blood Group Antigens/metabolism , N-Acetylneuraminic Acid/metabolism , Phenotype , Polysaccharides/chemistry , Polysaccharides/metabolism , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Lab Chip ; 18(13): 1891-1902, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29873383

ABSTRACT

Sequencing the genomes of individual cells enables the direct determination of genetic heterogeneity amongst cells within a population. We have developed an injection-moulded valveless microfluidic device in which single cells from colorectal cancer derived cell lines (LS174T, LS180 and RKO) and fresh colorectal tumors have been individually trapped, their genomes extracted and prepared for sequencing using multiple displacement amplification (MDA). Ninety nine percent of the DNA sequences obtained mapped to a reference human genome, indicating that there was effectively no contamination of these samples from non-human sources. In addition, most of the reads are correctly paired, with a low percentage of singletons (0.17 ± 0.06%) and we obtain genome coverages approaching 90%. To achieve this high quality, our device design and process shows that amplification can be conducted in microliter volumes as long as the lysis is in sub-nanoliter volumes. Our data thus demonstrates that high quality whole genome sequencing of single cells can be achieved using a relatively simple, inexpensive and scalable device. Detection of genetic heterogeneity at the single cell level, as we have demonstrated for freshly obtained single cancer cells, could soon become available as a clinical tool to precisely match treatment with the properties of a patient's own tumor.


Subject(s)
DNA, Neoplasm/genetics , Genome, Human/genetics , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Sequence Analysis, DNA/instrumentation , Single-Cell Analysis/instrumentation , Cell Line, Tumor , Humans , Single-Cell Analysis/methods
5.
Proc Natl Acad Sci U S A ; 113(36): E5344-53, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27543333

ABSTRACT

Proliferation and invasion of cancer cells require favorable pH, yet potentially toxic quantities of acid are produced metabolically. Membrane-bound transporters extrude acid from cancer cells, but little is known about the mechanisms that handle acid once it is released into the poorly perfused extracellular space. Here, we studied acid handling by myofibroblasts (colon cancer-derived Hs675.T, intestinal InMyoFib, embryonic colon-derived CCD-112-CoN), skin fibroblasts (NHDF-Ad), and colorectal cancer (CRC) cells (HCT116, HT29) grown in monoculture or coculture. Expression of the acid-loading transporter anion exchanger 2 (AE2) (SLC4A2 product) was detected in myofibroblasts and fibroblasts, but not in CRC cells. Compared with CRC cells, Hs675.T and InMyoFib myofibroblasts had very high capacity to absorb extracellular acid. Acid uptake into CCD-112-CoN and NHDF-Ad cells was slower and comparable to levels in CRC cells, but increased alongside SLC4A2 expression under stimulation with transforming growth factor ß1 (TGFß1), a cytokine involved in cancer-stroma interplay. Myofibroblasts and fibroblasts are connected by gap junctions formed by proteins such as connexin-43, which allows the absorbed acid load to be transmitted across the stromal syncytium. To match the stimulatory effect on acid uptake, cell-to-cell coupling in NHDF-Ad and CCD-112-CoN cells was strengthened with TGFß1. In contrast, acid transmission was absent between CRC cells, even after treatment with TGFß1. Thus, stromal cells have the necessary molecular apparatus for assembling an acid-venting route that can improve the flow of metabolic acid through tumors. Importantly, the activities of stromal AE2 and connexin-43 do not place an energetic burden on cancer cells, allowing resources to be diverted for other activities.


Subject(s)
Colorectal Neoplasms/metabolism , Connexin 43/genetics , Transforming Growth Factor beta1/metabolism , Acids/toxicity , Cell Proliferation/drug effects , Chloride-Bicarbonate Antiporters/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Connexin 43/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Hydrogen-Ion Concentration , Myofibroblasts/metabolism , Myofibroblasts/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Transforming Growth Factor beta1/pharmacology
6.
Proc Natl Acad Sci U S A ; 113(15): E2162-71, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27036009

ABSTRACT

Pericryptal myofibroblasts in the colon and rectum play an important role in regulating the normal colorectal stem cell niche and facilitating tumor progression. Myofibroblasts previously have been distinguished from normal fibroblasts mostly by the expression of α smooth muscle actin (αSMA). We now have identified AOC3 (amine oxidase, copper containing 3), a surface monoamine oxidase, as a new marker of myofibroblasts by showing that it is the target protein of the myofibroblast-reacting mAb PR2D3. The normal and tumor tissue distribution and the cell line reactivity of AOC3 match that expected for myofibroblasts. We have shown that the surface expression of AOC3 is sensitive to digestion by trypsin and collagenase and that anti-AOC3 antibodies can be used for FACS sorting of myofibroblasts obtained by nonenzymatic procedures. Whole-genome microarray mRNA-expression profiles of myofibroblasts and skin fibroblasts revealed four additional genes that are significantly differentially expressed in these two cell types: NKX2-3 and LRRC17 in myofibroblasts and SHOX2 and TBX5 in skin fibroblasts. TGFß substantially down-regulated AOC3 expression in myofibroblasts but in skin fibroblasts it dramatically increased the expression of αSMA. A knockdown of NKX2-3 in myofibroblasts caused a decrease of myofibroblast-related gene expression and increased expression of the fibroblast-associated gene SHOX2, suggesting that NKX2-3 is a key mediator for maintaining myofibroblast characteristics. Our results show that colorectal myofibroblasts, as defined by the expression of AOC3, NKX2-3, and other markers, are a distinctly different cell type from TGFß-activated fibroblasts.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Amine Oxidase (Copper-Containing)/genetics , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Colon/cytology , Colon/metabolism , Colorectal Neoplasms/metabolism , Fibroblasts/cytology , Gene Expression , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Oligonucleotide Array Sequence Analysis , Proteins/genetics , RNA, Messenger/metabolism , Skin/cytology , Skin/metabolism , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
7.
Cancer Res ; 74(20): 5866-77, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25168481

ABSTRACT

Drugs that inhibit insulin-like growth factor 1 (IGFI) receptor IGFIR were encouraging in early trials, but predictive biomarkers were lacking and the drugs provided insufficient benefit in unselected patients. In this study, we used genetic screening and downstream validation to identify the WNT pathway element DVL3 as a mediator of resistance to IGFIR inhibition. Sensitivity to IGFIR inhibition was enhanced specifically in vitro and in vivo by genetic or pharmacologic blockade of DVL3. In breast and prostate cancer cells, sensitization tracked with enhanced MEK-ERK activation and relied upon MEK activity and DVL3 expression. Mechanistic investigations showed that DVL3 is present in an adaptor complex that links IGFIR to RAS, which includes Shc, growth factor receptor-bound-2 (Grb2), son-of-sevenless (SOS), and the tumor suppressor DAB2. Dual DVL and DAB2 blockade synergized in activating ERKs and sensitizing cells to IGFIR inhibition, suggesting a nonredundant role for DVL3 in the Shc-Grb2-SOS complex. Clinically, tumors that responded to IGFIR inhibition contained relatively lower levels of DVL3 protein than resistant tumors, and DVL3 levels in tumors correlated inversely with progression-free survival in patients treated with IGFIR antibodies. Because IGFIR does not contain activating mutations analogous to EGFR variants associated with response to EGFR inhibitors, we suggest that IGF signaling achieves an equivalent integration at the postreceptor level through adaptor protein complexes, influencing cellular dependence on the IGF axis and identifying a patient population with potential to benefit from IGFIR inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drug Resistance, Neoplasm , Insulin-Like Growth Factor I/physiology , Phosphoproteins/physiology , Receptor, IGF Type 1/antagonists & inhibitors , ras Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Dishevelled Proteins , Gene Expression , Head and Neck Neoplasms/metabolism , Humans , Inhibitory Concentration 50 , Isoxazoles/pharmacology , MAP Kinase Signaling System , Male , Mice , Pyrimidines/pharmacology , Receptor, IGF Type 1/metabolism , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays
8.
Cancer Res ; 74(12): 3238-47, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24755471

ABSTRACT

Human colorectal cancer cell lines are used widely to investigate tumor biology, experimental therapy, and biomarkers. However, to what extent these established cell lines represent and maintain the genetic diversity of primary cancers is uncertain. In this study, we profiled 70 colorectal cancer cell lines for mutations and DNA copy number by whole-exome sequencing and SNP microarray analyses, respectively. Gene expression was defined using RNA-Seq. Cell line data were compared with those published for primary colorectal cancers in The Cancer Genome Atlas. Notably, we found that exome mutation and DNA copy-number spectra in colorectal cancer cell lines closely resembled those seen in primary colorectal tumors. Similarities included the presence of two hypermutation phenotypes, as defined by signatures for defective DNA mismatch repair and DNA polymerase ε proofreading deficiency, along with concordant mutation profiles in the broadly altered WNT, MAPK, PI3K, TGFß, and p53 pathways. Furthermore, we documented mutations enriched in genes involved in chromatin remodeling (ARID1A, CHD6, and SRCAP) and histone methylation or acetylation (ASH1L, EP300, EP400, MLL2, MLL3, PRDM2, and TRRAP). Chromosomal instability was prevalent in nonhypermutated cases, with similar patterns of chromosomal gains and losses. Although paired cell lines derived from the same tumor exhibited considerable mutation and DNA copy-number differences, in silico simulations suggest that these differences mainly reflected a preexisting heterogeneity in the tumor cells. In conclusion, our results establish that human colorectal cancer lines are representative of the main subtypes of primary tumors at the genomic level, further validating their utility as tools to investigate colorectal cancer biology and drug responses.


Subject(s)
Colorectal Neoplasms/genetics , Cell Line, Tumor , Chromosome Aberrations , Colorectal Neoplasms/metabolism , DNA Copy Number Variations , DNA Mutational Analysis , Exome , Gene Dosage , Gene Frequency , Genes, Neoplasm , Humans , Microsatellite Instability , Transcriptome
9.
Cancer Res ; 74(9): 2377-84, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24717177

ABSTRACT

Despite the millions of dollars spent on target validation and drug optimization in preclinical models, most therapies still fail in phase III clinical trials. Our current model systems, or the way we interpret data from them, clearly do not have sufficient clinical predictive power. Current opinion suggests that this is because the cell lines and xenografts that are commonly used are inadequate models that do not effectively mimic and predict human responses. This has become such a widespread belief that it approaches dogma in the field of drug discovery and optimization and has spurred a surge in studies devoted to the development of more sophisticated animal models such as orthotopic patient-derived xenografts in an attempt to obtain more accurate estimates of whether particular cancers will respond to given treatments. Here, we explore the evidence that has led to the move away from the use of in vitro cell lines and toward various forms of xenograft models for drug screening and development. We review some of the pros and cons of each model and give an overview of ways in which the use of cell lines could be modified to improve the predictive capacity of this well-defined model.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Humans
10.
Proc Natl Acad Sci U S A ; 109(51): 21046-51, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23213241

ABSTRACT

A significant proportion of colorectal cancer (CRC) patients are resistant to anti-ERBB1 [avian erythroblastic leukemia viral (v-erb-b) oncogene homolog, receptor for EGF] monoclonal antibodies (Mabs). We evaluated both immune and nonimmune effects of cetuximab (anti-ERBB1 Mab), trastuzumab (anti-ERBB2 Mab), pertuzumab (anti-ERBB2 Mab), and lapatinib (dual ERBB1 and ERBB2 tyrosine kinase inhibitor) in a large well-characterized panel of 64 CRC cell lines to find response predictive tumor characteristics. There was a significant correlation between the direct effects of cetuximab and lapatinib. Both agents were associated (P = 0.0004) with "triple' wild-type status in KRAS, BRAF, and PIK3CA exon 20. Most cell lines were resistant to the direct effects of anti-ERBB2 Mabs, suggesting that the effects of lapatinib might mainly be through ERBB1. Microarray mRNA expression profiles of sensitive and resistant cell lines showed that although ERBB1 receptor or ligand levels did not associate with cetuximab sensitivity, high levels of ERBB2 (P = 0.036) and amphiregulin (P = 0.026) predicted sensitivity to lapatinib. However, higher ERBB1 expression predicted susceptibility to cetuximab-induced antibody-dependent cellular cytotoxicity and occurred independently of KRAS/BRAF/PIK3CA mutations (P = 0.69). Lapatinib may be an effective alternative therapy to cetuximab in triple wild-type tumors. Microarray analysis provides suggestive biomarkers for resistance. ERBB1 levels, independent of mutation status, predict immune killing. Therefore, anti-ERBB1 antibodies may be considered in CRC tumors with higher ERBB1 expression and favorable FcγR polymorphisms.


Subject(s)
Colorectal Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cetuximab , Genes, ras , Humans , Immune System , Lapatinib , Models, Genetic , Oligonucleotide Array Sequence Analysis , Polymorphism, Genetic , Quinazolines/pharmacology , Trastuzumab
11.
Proc Natl Acad Sci U S A ; 107(49): 21058-63, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21097699

ABSTRACT

Replication error deficient (RER+) colorectal cancers are a distinct subset of colorectal cancers, characterized by inactivation of the DNA mismatch repair system. These cancers are typically pseudodiploid, accumulate mutations in repetitive sequences as a result of their mismatch repair deficiency, and have distinct pathologies. Regulatory sequences controlling all aspects of mRNA processing, especially including message stability, are found in the 3'UTR sequence of most genes. The relevant sequences are typically A/U-rich elements or U repeats. Microarray analysis of 14 RER+ (deficient) and 16 RER- (proficient) colorectal cancer cell lines confirms a striking difference in expression profiles. Analysis of the incidence of mononucleotide repeat sequences in the 3'UTRs, 5'UTRs, and coding sequences of those genes most differentially expressed in RER+ versus RER- cell lines has shown that much of this differential expression can be explained by the occurrence of a massive enrichment of genes with 3'UTR T repeats longer than 11 base pairs in the most differentially expressed genes. This enrichment was confirmed by analysis of two published consensus sets of RER differentially expressed probesets for a large number of primary colorectal cancers. Sequence analysis of the 3'UTRs of a selection of the most differentially expressed genes shows that they all contain deletions in these repeats in all RER+ cell lines studied. These data strongly imply that deregulation of mRNA stability through accumulation of mutations in repetitive regulatory 3'UTR sequences underlies the striking difference in expression profiles between RER+ and RER- colorectal cancers.


Subject(s)
3' Untranslated Regions/genetics , Colorectal Neoplasms/genetics , DNA Replication , Gene Expression Regulation, Neoplastic , Poly T/genetics , Sequence Deletion , Base Pair Mismatch , Cell Line, Tumor , Humans , Regulatory Sequences, Nucleic Acid , Repetitive Sequences, Nucleic Acid
12.
Proc Natl Acad Sci U S A ; 107(8): 3722-7, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133591

ABSTRACT

Cancer stem cells (CSCs) are the subpopulation of cells within a tumor that can self-renew, differentiate into multiple lineages, and drive tumor growth. Here we describe a two-pronged approach for the identification and characterization of CSCs from colorectal cancer cell lines, using a Matrigel-based differentiation assay, and cell surface markers CD44 and CD24. About 20 to 30% of cells from the SW1222 cell line form megacolonies in Matrigel that have complex 3D structures resembling colonic crypts. The megacolonies' capacity to self-renew in vitro is direct evidence that they contain the CSCs. Furthermore, just 200 cells from SW1222 megacolonies initiate tumors in NOD/SCID mice. We also showed that CD44(+)CD24(+) cells enriched for colorectal CSCs in the HT29 and SW1222 cell lines, which can self-renew and reform all four CD44/CD24 subpopulations, are the most clonogenic in vitro and can initiate tumors in vivo. A single SW1222 CD44(+)CD24(+) CSC, when grown in Matrigel, can form large megacolonies that differentiate into enterocyte, enteroendocrine, and goblet cell lineages. The HCT116 line does not differentiate or express CDX1, nor does it contain subpopulations of cells with greater tumor-forming capacity, suggesting that HCT116 contains mainly CSCs. However, forced expression of CDX1 in HCT116 leads to reduced clonogenicity and production of differentiating crypt-containing colonies, which can explain the selection for reduced CDX1 expression in many colorectal cancers. In summary, colorectal cancer cell lines contain subpopulations of CSCs, characterized by their cell surface markers and colony morphology, which can self-renew and differentiate into multiple lineages.


Subject(s)
Colorectal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , CD24 Antigen/biosynthesis , Cell Differentiation , Cell Line, Tumor , Cell Separation , Collagen , Drug Combinations , Homeodomain Proteins/biosynthesis , Humans , Hyaluronan Receptors/biosynthesis , Laminin , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , Proteoglycans
13.
Proc Natl Acad Sci U S A ; 106(6): 1936-41, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19188603

ABSTRACT

CDX1 is a transcription factor that plays a key role in intestinal development and differentiation. However, the downstream targets of CDX1 are less well defined than those of its close homologue, CDX2. We report here the identification of downstream targets of CDX1 using microarray gene-expression analysis and other approaches. Keratin 20 (KRT20), a member of the intermediate filament and a well-known marker of intestinal differentiation, was initially identified as one of the genes likely to be directly regulated by CDX1. CDX1 and KRT20 mRNA expression were significantly correlated in a panel of 38 colorectal cancer cell lines. Deletion and mutation analysis of the KRT20 promoter showed that the minimum regulatory region for the control of KRT20 expression by CDX1 is within 246 bp upstream of the KRT20 transcription start site. ChIP analysis confirmed that CDX1 binds to the predicted CDX elements in this region of the KRT20 promoter in vivo. In addition, immunohistochemistry showed expression of CDX1 parallels that of KRT20 in the normal crypt, which further supports their close relationship. In summary, our observations strongly imply that KRT20 is directly regulated by CDX1, and therefore suggest a role for CDX1 in maintaining differentiation in intestinal epithelial cells. Because a key feature of the development of a cancer is an unbalanced program of proliferation and differentiation, dysregulation of CDX1 may be an advantage for the development of a colorectal carcinoma. This could, therefore, explain the relatively frequent down regulation of CDX1 in colorectal carcinomas by hypermethylation.


Subject(s)
Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/physiology , Keratin-20/genetics , Binding Sites , Cell Line, Tumor , Colorectal Neoplasms/etiology , Colorectal Neoplasms/genetics , Gastrointestinal Tract , Gene Expression Profiling , Genes, Homeobox/physiology , Homeodomain Proteins/genetics , Humans , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic
14.
Eur Urol ; 56(3): 486-94, 2009 Sep.
Article in English | MEDLINE | ID: mdl-18514389

ABSTRACT

BACKGROUND: The Wnt signalling pathway directs aspects of embryogenesis and is thought to contribute to maintenance of certain stem cell populations. Disruption of the pathway has been observed in many different tumour types. In bowel, stomach, and endometrial cancer, this is usually due to mutation of genes encoding Wnt pathway components APC or beta-catenin. Such mutations are rare in hepatocellular carcinomas and medulloblastomas with Wnt pathway dysfunction, and there, mutation in genes for other Wnt molecules, such as Axin, is more frequently found. OBJECTIVE: Although evidence of abnormal activation of the Wnt pathway in prostate cancer has been demonstrated by several groups, APC and beta-catenin mutations are infrequent. We sought mutations in genes encoding Wnt pathway participants in a panel of prostate cancer clinical specimens and cell lines. DESIGN, SETTING, AND PARTICIPANTS: DNA was obtained from 49 advanced prostate cancer specimens using laser microdissection followed by whole genome amplification and 8 prostate cancer cell lines. MEASUREMENTS: The DNA samples were screened for mutations in the genes encoding APC, beta-catenin, and Axin. The subcellular distribution of beta-catenin expression was assessed in the clinical specimens using immunohistochemistry. RESULTS AND LIMITATIONS: Abnormal patterns of beta-catenin expression, suggesting Wnt pathway dysregulation, were observed in 71% of specimens. One APC mutation, two beta-catenin gene mutations, and 7 DNA sequence variations in the Axin gene were detected. Four different Axin polymorphisms were also found in the cell lines. The study does not provide definite evidence that the observed sequence changes alter protein function, promoting neoplasia, but the potential functional relevance of these variants is discussed. CONCLUSIONS: These data contribute to our understanding of the role of Wnt dysregulation in prostatic tumourigenesis and support the current interest in the pathway as a therapeutic target. Of particular interest, we identified three new potentially functionally relevant AXIN1 mutations.


Subject(s)
Mutation , Prostatic Neoplasms/genetics , Repressor Proteins/genetics , Aged , Aged, 80 and over , Axin Protein , Genes, APC , Humans , Male , Middle Aged , Prostatic Neoplasms/pathology , Signal Transduction/genetics , Wnt Proteins/genetics , beta Catenin/genetics
15.
Proc Natl Acad Sci U S A ; 102(8): 2868-73, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15710876

ABSTRACT

Apc(Min) mice have provided an example of a locus (Modifier of Min1; Mom1) modifying adenoma numbers in the intestines of inbred strains. Linkage analysis located Mom1 on chromosome 4, and further investigation identified secretory phospholipase A2 (Pla2g2a) as a candidate gene. Because of unknown variation introduced by a single founding male mouse, our Min stock, although Pla2g2a(Mom1-s), was not on a pure C57BL/6J background and exhibited several polymorphic loci, including a region on chromosome 18 distal to Apc. Through selective breeding for homozygosity for distal chromosome 18 markers, six recombinant lines that presented with limited intraline variation in adenoma numbers were established. One line (V) showed a particularly severe phenotype (mean adenoma number +/- SEM, 370 +/- 21) compared with the other lines that recorded significantly lower means (3- to 5-fold; P < 10(-3), t test). Intercrosses between lines I and V showed suppression of the severe phenotype in the N1 generation. In N2 (and subsequent) backcrosses, tumor multiplicity depended on the origins of the WT and Min Apc alleles. Mice carrying both alleles from line V had a severe phenotype; others had mild disease very similar to line I (likelihood ratio statistic > 49.0; likelihood of odds > 10; P < 10(-5)). Frequency of allele loss at Apc was increased significantly in adenomas of mice with more severe disease. We propose that a modifier gene close to Apc or structural variation on chromosome 18 modifies polyp numbers in our mice, possibly by altering the frequency of WT Apc allele loss.


Subject(s)
Adenoma/genetics , Genes, APC , Intestinal Neoplasms/genetics , Alleles , Animals , Chromosome Mapping , Female , Genetic Linkage , Genetic Predisposition to Disease , Intestinal Polyps/genetics , Male , Mice , Mice, Inbred C57BL , Phenotype
16.
Proc Natl Acad Sci U S A ; 101(45): 15992-7, 2004 Nov 09.
Article in English | MEDLINE | ID: mdl-15520370

ABSTRACT

Clear-cut inherited Mendelian traits, such as familial adenomatous polyposis or hereditary nonpolyposis colorectal cancer, account for <4% of colorectal cancers. Another 20% of all colorectal cancers are thought to occur in individuals with a significant inherited multifactorial susceptibility to colorectal cancer that is not obviously familial. Incompletely penetrant, comparatively rare missense variants in the adenomatous polyposis coli gene, which is responsible for familial adenomatous polyposis, have been described in patients with multiple colorectal adenomas. These variants represent a category of variation that has been suggested, quite generally, to account for a substantial fraction of such multifactorial inherited susceptibility. The aim of this study was to explore this rare variant hypothesis for multifactorial inheritance by using multiple colorectal adenomas as the model. Patients with multiple adenomas were screened for germ-line variants in a panel of candidate genes. Germ-line DNA was obtained from 124 patients with between 3 and 100 histologically proven synchronous or metachronous adenomatous polyps. All patients were tested for the adenomatous polyposis coli variants I1307K and E1317Q, and variants were also sought in AXIN1 (axin), CTNNB1 (beta-catenin), and the mismatch repair genes hMLH1 and hMSH2. The control group consisted of 483 random controls. Thirty of 124 (24.9%) patients carried potentially pathogenic germ-line variants as compared with 55 ( approximately 12%) of the controls. This overall difference is highly significant, suggesting that many rare variants collectively contribute to the inherited susceptibility to colorectal adenomas.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Alleles , Axin Protein , Base Pair Mismatch , Base Sequence , Case-Control Studies , DNA Mutational Analysis , DNA Repair/genetics , DNA, Neoplasm/genetics , Gene Frequency , Genes, APC , Genetic Variation , Germ-Line Mutation , Humans , Microsatellite Repeats , Models, Genetic , Repressor Proteins/genetics , Signal Transduction/genetics
17.
Br Med Bull ; 64: 27-43, 2002.
Article in English | MEDLINE | ID: mdl-12421723

ABSTRACT

Familial adenomatous polyposis and hereditary non-polyposis colorectal cancer are dominantly inherited conditions with 100% and 80% life-time risk of developing colorectal cancer, respectively. The genetic mutations responsible for these two conditions lie in the adenomatous polyposis coli (APC) and mismatch repair genes. These same genes also play a key role in the formation of sporadic colorectal cancers, which arise on a background of a similar spectrum of mutations to the hereditary cancers. This article examines the genetic mechanisms underlying the hereditary colorectal cancers, as well as genetic predisposition to colorectal cancer in the general population in the absence of a clear-cut genetic syndrome. Colorectal cancer arises as the cumulative effect of multiple mutations within the cell, allowing it to escape growth and regulatory control mechanisms. This step-wise progression of mutations facilitates the histological transition from normal mucosa to adenoma to carcinoma. The latter part of this paper focuses on the key genetic events underlying this process and provides an overview of the genetic mechanisms responsible for colorectal tumorigenesis.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Diseases, Inborn/etiology , Adenoma/genetics , Adenoma/pathology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Colorectal Neoplasms/pathology , Disease Progression , Genes, APC , Genetic Diseases, Inborn/pathology , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Mutation , Risk Factors
18.
Genes Chromosomes Cancer ; 34(4): 390-7, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12112528

ABSTRACT

The long arm of human chromosome 7 between 7q22 and 7q36 has been identified as a region harboring one or more tumor-suppressor genes (TSGs) inactivated in acute myeloid leukemia (AML). Additional TSGs mapping to other chromosomes may well be involved in the etiology of this disease. For example, experiments using a mouse model system have indicated the possible presence of an AML TSG at 11p11-12. Microcell-mediated chromosome transfer (MMCT) has been used to introduce human chromosomes 7 and 11 into a murine myeloid leukemia cell line. A proportion of MMCT hybrid clones containing either whole chromosome 7 or fragments of chromosome 11 showed a significant delay in leukemogenic onset when injected into syngeneic mice. Screening of hybrid clones did not associate any human microsatellite markers with decreased leukemogenic potential in vivo. However, preliminary evidence was obtained of allelic loss at chromosomal regions homologous with human 7q22 in murine F1 hybrid AMLs. Our data provide functional evidence of AML-associated TSGs localized to human chromosomes 7 and 11 in support of previously published studies on cytogenetic and allelic losses associated with AML development.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 7/genetics , Gene Transfer Techniques , Genes, Tumor Suppressor , Leukemia, Myeloid/genetics , Neoplasm Transplantation , Tumor Suppressor Proteins , Acute Disease , Animals , Chimera/genetics , Chromosome Banding , Chromosome Mapping/methods , Clone Cells , Cytogenetic Analysis/methods , Cytoskeletal Proteins , Flow Cytometry/methods , Genetic Markers/genetics , Homeodomain Proteins/genetics , Humans , LIM Domain Proteins , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/pathology , Loss of Heterozygosity/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred CBA , Neoplasm Transplantation/methods , Nucleic Acid Amplification Techniques , Peroxidase/metabolism , Polymerase Chain Reaction , RNA-Binding Proteins , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL