Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Epigenetics Chromatin ; 13(1): 25, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32487212

ABSTRACT

BACKGROUND: DNA methylation is a highly studied epigenetic signature that is associated with regulation of gene expression, whereby genes with high levels of promoter methylation are generally repressed. Genomic imprinting occurs when one of the parental alleles is methylated, i.e., when there is inherited allele-specific methylation (ASM). A special case of imprinting occurs during X chromosome inactivation in females, where one of the two X chromosomes is silenced, to achieve dosage compensation between the sexes. Another more widespread form of ASM is sequence dependent (SD-ASM), where ASM is linked to a nearby heterozygous single nucleotide polymorphism (SNP). RESULTS: We developed a method to screen for genomic regions that exhibit loss or gain of ASM in samples from two conditions (treatments, diseases, etc.). The method relies on the availability of bisulfite sequencing data from multiple samples of the two conditions. We leverage other established computational methods to screen for these regions within a new R package called DAMEfinder. It calculates an ASM score for all CpG sites or pairs in the genome of each sample, and then quantifies the change in ASM between conditions. It then clusters nearby CpG sites with consistent change into regions. In the absence of SNP information, our method relies only on reads to quantify ASM. This novel ASM score compares favorably to current methods that also screen for ASM. Not only does it easily discern between imprinted and non-imprinted regions, but also females from males based on X chromosome inactivation. We also applied DAMEfinder to a colorectal cancer dataset and observed that colorectal cancer subtypes are distinguishable according to their ASM signature. We also re-discover known cases of loss of imprinting. CONCLUSION: We have designed DAMEfinder to detect regions of differential ASM (DAMEs), which is a more refined definition of differential methylation, and can therefore help in breaking down the complexity of DNA methylation and its influence in development and disease.


Subject(s)
Alleles , DNA Methylation , Epigenomics/methods , Sequence Analysis, DNA/methods , Colorectal Neoplasms/genetics , CpG Islands , Female , Genomic Imprinting , Humans , Male , Software
2.
BMC Cancer ; 20(1): 290, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32252665

ABSTRACT

BACKGROUND: Identifying molecular differences between primary and metastatic colorectal cancers-now possible with the aid of omics technologies-can improve our understanding of the biological mechanisms of cancer progression and facilitate the discovery of novel treatments for late-stage cancer. We compared the DNA methylomes of primary colorectal cancers (CRCs) and CRC metastases to the liver. Laser microdissection was used to obtain epithelial tissue (10 to 25 × 106 µm2) from sections of fresh-frozen samples of primary CRCs (n = 6), CRC liver metastases (n = 12), and normal colon mucosa (n = 3). DNA extracted from tissues was enriched for methylated sequences with a methylCpG binding domain (MBD) polypeptide-based protocol and subjected to deep sequencing. The performance of this protocol was compared with that of targeted enrichment for bisulfite sequencing used in a previous study of ours. RESULTS: MBD enrichment captured a total of 322,551 genomic regions (249.5 Mb or ~ 7.8% of the human genome), which included over seven million CpG sites. A few of these regions were differentially methylated at an expected false discovery rate (FDR) of 5% in neoplastic tissues (primaries: 0.67%, i.e., 2155 regions containing 279,441 CpG sites; liver metastases: 1%, i.e., 3223 regions containing 312,723 CpG sites) as compared with normal mucosa samples. Most of the differentially methylated regions (DMRs; 94% in primaries; 70% in metastases) were hypermethylated, and almost 80% of these (1882 of 2396) were present in both lesion types. At 5% FDR, no DMRs were detected in liver metastases vs. primary CRC. However, short regions of low-magnitude hypomethylation were frequent in metastases but rare in primaries. Hypermethylated DMRs were far more abundant in sequences classified as intragenic, gene-regulatory, or CpG shelves-shores-island segments, whereas hypomethylated DMRs were equally represented in extragenic (mainly, open-sea) and intragenic (mainly, gene bodies) sequences of the genome. Compared with targeted enrichment, MBD capture provided a better picture of the extension of CRC-associated DNA hypermethylation but was less powerful for identifying hypomethylation. CONCLUSIONS: Our findings demonstrate that the hypermethylation phenotype in CRC liver metastases remains similar to that of the primary tumor, whereas CRC-associated DNA hypomethylation probably undergoes further progression after the cancer cells have migrated to the liver.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epigenome , Liver Neoplasms/secondary , Colorectal Neoplasms/metabolism , Epigenesis, Genetic , High-Throughput Nucleotide Sequencing/methods , Humans , Laser Capture Microdissection/methods , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Phenotype , Promoter Regions, Genetic
3.
Chem Res Toxicol ; 30(3): 769-776, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28076683

ABSTRACT

Bioactivation as well as DNA repair affects the susceptibility of cancer cells to the action of DNA-alkylating chemotherapeutic drugs. However, information is limited with regard to the relative contributions of these processes to the biological outcome of metabolically activated DNA alkylating agents. We evaluated the influence of cellular bioactivation capacity and DNA repair on cytotoxicity of the DNA alkylating agent acylfulvene (AF). We compared the cytotoxicity and RNA synthesis inhibition by AF and its synthetic activated analogue iso-M0 in a panel of fibroblast cell lines with deficiencies in transcription-coupled (TC-NER) or global genome nucleotide excision repair (GG-NER). We related these data to the inherent bioactivation capacity of each cell type on the basis of mRNA levels. We demonstrated that specific inactivation of TC-NER by siRNA had the largest positive impact on AF activity in a cancer cell line. These findings establish that transcription-coupled DNA repair reduces cellular sensitivity to AF, independent of the requirement for bioactivation.


Subject(s)
DNA Repair , Sesquiterpenes/pharmacology , Spiro Compounds/pharmacology , Transcription, Genetic/drug effects , Activation, Metabolic , Cell Line , Humans , Sesquiterpenes/pharmacokinetics , Spiro Compounds/pharmacokinetics
4.
Genome Biol ; 15(2): R35, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24517713

ABSTRACT

Affinity capture of DNA methylation combined with high-throughput sequencing strikes a good balance between the high cost of whole genome bisulfite sequencing and the low coverage of methylation arrays. We present BayMeth, an empirical Bayes approach that uses a fully methylated control sample to transform observed read counts into regional methylation levels. In our model, inefficient capture can readily be distinguished from low methylation levels. BayMeth improves on existing methods, allows explicit modeling of copy number variation, and offers computationally efficient analytical mean and variance estimators. BayMeth is available in the Repitools Bioconductor package.


Subject(s)
Bayes Theorem , DNA Methylation/genetics , Evaluation Studies as Topic , High-Throughput Nucleotide Sequencing , CpG Islands/genetics , DNA Copy Number Variations , Genome, Human , Humans
5.
BMC Cancer ; 14: 46, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24472434

ABSTRACT

BACKGROUND: Biological processes are controlled by transcription networks. Expression changes of transcription factor (TF) genes in precancerous lesions are therefore crucial events in tumorigenesis. Our aim was to obtain a comprehensive picture of these changes in colorectal adenomas. METHODS: Using a 3-pronged selection procedure, we analyzed transcriptomic data on 34 human tissue samples (17 adenomas and paired samples of normal mucosa, all collected with ethics committee approval and written, informed patient consent) to identify TFs with highly significant tumor-associated gene expression changes whose potential roles in colorectal tumorigenesis have been under-researched. Microarray data were subjected to stringent statistical analysis of TF expression in tumor vs. normal tissues, MetaCore-mediated identification of TF networks displaying enrichment for genes that were differentially expressed in tumors, and a novel quantitative analysis of the publications examining the TF genes' roles in colorectal tumorigenesis. RESULTS: The 261 TF genes identified with this procedure included DACH1, which plays essential roles in the proper proliferation and differentiation of retinal and leg precursor cell populations in Drosophila melanogaster. Its possible roles in colorectal tumorigenesis are completely unknown, but it was found to be markedly overexpressed (mRNA and protein) in all colorectal adenomas and in most colorectal carcinomas. However, DACH1 expression was absent in some carcinomas, most of which were DNA mismatch-repair deficient. When networks were built using the set of TF genes identified by all three selection procedures, as well as the entire set of transcriptomic changes in adenomas, five hub genes (TGFB1, BIRC5, MYB, NR3C1, and TERT) where identified as putatively crucial components of the adenomatous transformation process. CONCLUSION: The transcription-regulating network of colorectal adenomas (compared with that of normal colorectal mucosa) is characterized by significantly altered expression of over 250 TF genes, many of which have never been investigated in relation to colorectal tumorigenesis.


Subject(s)
Adenoma/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Gene Expression Profiling , Transcription Factors/genetics , Adenoma/metabolism , Adenoma/pathology , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genes, myb , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Survivin , Telomerase/genetics , Telomerase/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
6.
PLoS One ; 8(7): e69473, 2013.
Article in English | MEDLINE | ID: mdl-23936024

ABSTRACT

We previously reported that the expression of KIAA1199 in human colorectal tumors (benign and malignant) is markedly higher than that in the normal colonic mucosa. In this study, we investigated the functions of the protein encoded by this gene, which are thus far unknown. Immunostaining studies were used to reveal its subcellular localization, and proteomic and gene expression experiments were conducted to identify proteins that might interact with KIAA1199 and molecular pathways in which it might play roles. Using colon cancer cell lines, we showed that both endogenous and ectopically expressed KIAA1199 is secreted into the extracellular environment. In the cells, it was found mainly in the perinuclear space (probably the ER) and cell membrane. Both cellular compartments were also over-represented in lists of proteins identified by mass spectrometry as putative KIAA1199 interactors and/or proteins encoded by genes whose transcription was significantly changed by KIAA1199 expression. These proteomic and transcriptomic datasets concordantly link KIAA1199 to several genes/proteins and molecular pathways, including ER processes like protein binding, transport, and folding; and Ca(2+), G-protein, ephrin, and Wnt signaling. Immunoprecipitation experiments confirmed KIAA1199's interaction with the cell-membrane receptor ephrin A2 and with the ER receptor ITPR3, a key player in Ca(2+) signaling. By modulating Ca(2+) signaling, KIAA1199 could affect different branches of the Wnt network. Our findings suggest it may negatively regulate the Wnt/CTNNB1 signaling, and its expression is associated with decreased cell proliferation and invasiveness.


Subject(s)
Colorectal Neoplasms/metabolism , Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cell Shape/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Hyaluronoglucosaminidase , Neoplasm Invasiveness , Protein Binding/genetics , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Wnt Signaling Pathway/genetics
7.
EMBO Mol Med ; 3(6): 334-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21538994

ABSTRACT

Improved colonoscopy is revealing precancerous lesions that were frequently missed in the past, and ∼30% of those detected today have nonpolypoid morphologies ranging from slightly raised to depressed. To characterize these lesions molecularly, we assessed transcription of 23,768 genes in 42 precancerous lesions (25 slightly elevated nonpolypoid and 17 pedunculated polypoid), each with corresponding samples of normal mucosa. Nonpolypoid versus polypoid morphology explained most gene expression variance among samples; histology, size, and degree of dysplasia were also linked to specific patterns. Expression changes in polypoid lesions frequently affected cell-cycling pathways, whereas cell-survival dysregulation predominated in nonpolypoid lesions. The latter also displayed fewer and less dramatic expression changes than polypoid lesions. Paradigmatic of this trend was progressive loss through the normal > nonpolypoid > polypoid > cancer sequence of TMIGD1 mRNA and protein. This finding, along with TMIGD1 protein expression patterns in tissues and cell lines, suggests that TMIGD1 might be associated with intestinal-cell differentiation. We conclude that molecular dysregulation in slightly elevated, nonpolypoid, precancerous colorectal lesions may be somewhat less severe than that observed in classic adenomatous polyps.


Subject(s)
Colorectal Neoplasms/pathology , Gene Expression Profiling , Membrane Glycoproteins/genetics , Precancerous Conditions/pathology , Adenomatous Polyps/metabolism , Adult , Aged , Aged, 80 and over , Colonoscopy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Male , Membrane Glycoproteins/metabolism , Middle Aged , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Principal Component Analysis
8.
Toxicol In Vitro ; 25(2): 446-53, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21095227

ABSTRACT

Cigarette smoke causes lung tumorigenesis; however, the mechanisms underlying transformation are unknown. We investigated if tobacco compounds induce DNA promoter hypermethylation in BEAS-2B cells treated with low doses of cigarette smoke condensate (CSC) for one month. Transcriptional profiles and anchorage-independent growth were explored using Affymetrix microarray and soft agar assay, respectively. To investigate if tobacco compounds induce hypermethylation, CSC/dimethyl sulfoxide (DMSO)-treated cells were further treated with 5-Aza-2'-deoxycytidine (5AzaC) and trychostatin A (TSA). This treatment was followed by transcriptional profiling. CSC-exposed cells acquired a fibroblast-like shape with enhanced anchorage-independent growth. Silencing of epithelial cadherin, the hallmark of epithelial to mesenchymal transition (EMT), was observed upon exposure to CSC. Changes in the expression of genes involved in epidermal development, intercellular junction formation, and cytoskeleton formation were identified. Gene expression profiles from 5AzaC- and TSA-treated cells revealed 130 genes possibly methylated due to chronic CSC exposure. Our results suggest that E-cadherin may also be silenced by hypermethylation in an in vitro model of chronic exposure to low doses of CSC. This study demonstrates evidence for a tobacco compound induced EMT-like process in vitro and provides insight into possible mechanisms of gene silencing occurring during this treatment.


Subject(s)
Bronchi/pathology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Nicotiana/toxicity , Smoke/adverse effects , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cells, Cultured , Cytoskeleton/physiology , DNA Methylation , Decitabine , Gene Expression Profiling , Glucosides/pharmacology , Humans , Hydroxamic Acids/pharmacology , Intercellular Junctions/physiology
9.
Cell Cycle ; 9(22): 4592-9, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21099343

ABSTRACT

We have previously shown that the DNA damage-induced G2 arrest is contributed by inhibition of Aurora A (AurA) and that transduction of active AurA into arrested cells allows bypassing the block through reactivation of CDK1. In this study, we investigated the mechanism of DNA damage-induced AurA inhibition. We provide evidence that ionizing radiation (IR) administered in mitosis, a time when AurA protein and enzymatic activity reach peak levels, impairs interaction with the partner TPX2, leading to inactivation of the kinase through dephosphorylation of AurA T-loop residue, T288. We find that decreased AurA-TPX2 complex formation in response to irradiation results from reduced cellular levels of TPX2, an effect that is both contributed by increased APC/CDH1-dependent protein degradation and decreased translation of TPX2 mRNA.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , Microtubule-Associated Proteins/metabolism , Mitosis/radiation effects , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Aurora Kinases , Cell Cycle Proteins/genetics , Gene Expression Regulation , HeLa Cells , Humans , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , RNA, Messenger/metabolism , Radiation, Ionizing
10.
Nucleic Acids Res ; 38(22): 8131-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20705653

ABSTRACT

It is known that transcription can induce DNA recombination, thus compromising genomic stability. RECQ5 DNA helicase promotes genomic stability by regulating homologous recombination. Recent studies have shown that RECQ5 forms a stable complex with RNA polymerase II (RNAPII) in human cells, but the cellular role of this association is not understood. Here, we provide evidence that RECQ5 specifically binds to the Ser2,5-phosphorylated C-terminal repeat domain (CTD) of the largest subunit of RNAPII, RPB1, by means of a Set2-Rpb1-interacting (SRI) motif located at the C-terminus of RECQ5. We also show that RECQ5 associates with RNAPII-transcribed genes in a manner dependent on the SRI motif. Notably, RECQ5 density on transcribed genes correlates with the density of Ser2-CTD phosphorylation, which is associated with the productive elongation phase of transcription. Furthermore, we show that RECQ5 negatively affects cell viability upon inhibition of spliceosome assembly, which can lead to the formation of mutagenic R-loop structures. These data indicate that RECQ5 binds to the elongating RNAPII complex and support the idea that RECQ5 plays a role in the maintenance of genomic stability during transcription.


Subject(s)
RNA Polymerase II/metabolism , RecQ Helicases/metabolism , Transcription, Genetic , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Cell Line , Cell Survival , Conserved Sequence , Humans , Molecular Sequence Data , Phosphorylation , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , RNA Polymerase II/chemistry , RecQ Helicases/chemistry , Repetitive Sequences, Amino Acid , Spliceosomes/metabolism
11.
Mol Cancer ; 8: 124, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20015382

ABSTRACT

BACKGROUND: Tumor development in the human colon is commonly accompanied by epigenetic changes, such as DNA methylation and chromatin modifications. These alterations result in significant, inheritable changes in gene expression that contribute to the selection of tumor cells with enhanced survival potential. RESULTS: A recent high-throughput gene expression analysis conducted by our group identified numerous genes whose transcription was markedly diminished in colorectal tumors. One of these, the protein-tyrosine phosphatase receptor type R (PTPRR) gene, was dramatically downregulated from the earliest stages of cellular transformation. Here, we show that levels of both major PTPRR transcript variants are markedly decreased (compared with normal mucosal levels) in precancerous and cancerous colorectal tumors, as well in colorectal cancer cell lines. The expression of the PTPRR-1 isoform was inactivated in colorectal cancer cells as a result of de novo CpG island methylation and enrichment of transcription-repressive histone-tail marks, mainly H3K27me3. De novo methylation of the PTPRR-1 transcription start site was demonstrated in 29/36 (80%) colorectal adenomas, 42/44 (95%) colorectal adenocarcinomas, and 8/8 (100%) liver metastases associated with the latter tumors. CONCLUSIONS: Epigenetic downregulation of PTPRR seems to be an early alteration in colorectal cell transformation, which is maintained during the clonal selection associated with tumor progression. It may represent a preliminary step in the constitutive activation of the RAS/RAF/MAPK/ERK signalling, an effect that will later be consolidated by mutations in genes encoding key components of this pathway.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Gene Silencing , Receptor-Like Protein Tyrosine Phosphatases, Class 7/genetics , Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , DNA Methylation , Humans , RNA, Messenger/genetics
12.
Cell ; 135(7): 1167-9, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109887

ABSTRACT

Whether 5-methylcytosine (meC) can be enzymatically removed from vertebrate DNA has been the subject of extensive study and also some controversy. Rai et al. (2008) now report that cytosine demethylation can be accomplished in a one-cell zebrafish embryo by the combined action of a cytidine deaminase and a thymine DNA glycosylase.


Subject(s)
Cytosine/metabolism , DNA Methylation , Animals , Cytidine Deaminase/metabolism , Thymine DNA Glycosylase/metabolism , Zebrafish
13.
Oncol Rep ; 17(6): 1421-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17487400

ABSTRACT

Epigenetic alterations have been reported in colorectal neoplasia which can either complement or in some cases be predisposed to genetic alterations such as K-ras mutations. We examined the promoter methylation status of the CDKN2A and O6-methylguanine-DNA methyltransferase (MGMT) genes, after sodium bisulfite conversion and DNA amplification with methylation specific PCR. Moreover, we searched for G to A transitions in codons 12 and 13 of the K-ras oncogene in normal colorectal mucosae, aberrant crypt foci (ACF, early premalignant lesions) and carcinomas. CDKN2A hypermethylation was an infrequent event in ACF (2 of 26, 7.7%). On the contrary, MGMT hypermethylation was found in the normal mucosae (3 of the 12 samples, 25%), in 14 of the 26 ACF (53.8%) and in 7 of the 9 (77.8%) carcinomas examined. K-ras mutations were evident in 6 ACF (23%) and in 3 carcinomas (33.3%), mostly associated with MGMT promoter hypermethylation. These findings strongly support the hypothesis that epigenetic mechanisms play an important role in the early steps of colorectal carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , O(6)-Methylguanine-DNA Methyltransferase/genetics , Aged , Aged, 80 and over , Carcinoma/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Female , Genes, ras/genetics , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , Mutation , Promoter Regions, Genetic , Sequence Analysis, DNA , Sulfites/chemistry
14.
Mol Cancer Res ; 5(12): 1263-75, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18171984

ABSTRACT

Colorectal cancers are believed to arise predominantly from adenomas. Although these precancerous lesions have been subjected to extensive clinical, pathologic, and molecular analyses, little is currently known about the global gene expression changes accompanying their formation. To characterize the molecular processes underlying the transformation of normal colonic epithelium, we compared the transcriptomes of 32 prospectively collected adenomas with those of normal mucosa from the same individuals. Important differences emerged not only between the expression profiles of normal and adenomatous tissues but also between those of small and large adenomas. A key feature of the transformation process was the remodeling of the Wnt pathway reflected in patent overexpression and underexpression of 78 known components of this signaling cascade. The expression of 19 Wnt targets was closely correlated with clear up-regulation of KIAA1199, whose function is currently unknown. In normal mucosa, KIAA1199 expression was confined to cells in the lower portion of intestinal crypts, where Wnt signaling is physiologically active, but it was markedly increased in all adenomas, where it was expressed in most of the epithelial cells, and in colon cancer cell lines, it was markedly reduced by inactivation of the beta-catenin/T-cell factor(s) transcription complex, the pivotal mediator of Wnt signaling. Our transcriptomic profiles of normal colonic mucosa and colorectal adenomas shed new light on the early stages of colorectal tumorigenesis and identified KIAA1199 as a novel target of the Wnt signaling pathway and a putative marker of colorectal adenomatous transformation.


Subject(s)
Adenoma/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Adenoma/pathology , Aged , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/pathology , Female , Genetic Markers , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Phylogeny , RNA, Messenger/metabolism , Transcription, Genetic
15.
Cancer Res ; 65(23): 10759-66, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322221

ABSTRACT

The human mismatch repair (MMR) proteins hMLH1 and hPMS2 function in MMR as a heterodimer. Cells lacking either protein have a strong mutator phenotype and display microsatellite instability, yet mutations in the hMLH1 gene account for approximately 50% of hereditary nonpolyposis colon cancer families, whereas hPMS2 mutations are substantially less frequent and less penetrant. Similarly, in the mouse model, Mlh1-/- animals are highly cancer prone and present with gastrointestinal tumors at an early age, whereas Pms2-/- mice succumb to cancer much later in life and do not present with gastrointestinal tumors. This evidence suggested that MLH1 might functionally interact with another MutL homologue, which compensates, at least in part, for a deficiency in PMS2. Sterility of Mlh1-/-, Pms2-/-, and Mlh3-/- mice implicated the Mlh1/Pms2 and Mlh1/Mlh3 heterodimers in meiotic recombination. We now show that the hMLH1/hMLH3 heterodimer, hMutLgamma, can also assist in the repair of base-base mismatches and single extrahelical nucleotides in vitro. Analysis of hMLH3 expression in colon cancer cell lines indicated that the protein levels vary substantially and independently of hMLH1. If hMLH3 participates in MMR in vivo, its partial redundancy with hPMS2, coupled with the fluctuating expression levels of hMLH3, may help explain the low penetrance of hPMS2 mutations in hereditary nonpolyposis colon cancer families.


Subject(s)
Base Pair Mismatch , Carrier Proteins/physiology , DNA Repair/physiology , Adaptor Proteins, Signal Transducing , Animals , Baculoviridae/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Humans , MutL Protein Homolog 1 , MutL Proteins , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Spodoptera/genetics , Spodoptera/metabolism
16.
Clin Cancer Res ; 11(23): 8332-40, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322293

ABSTRACT

PURPOSE: Many studies have evaluated the role of high levels of microsatellite instability (MSI) as a prognostic marker and predictor of the response to chemotherapy in colorectal cancer (CRC); however, the results are not conclusive. The aim of this study was to analyze the prognostic significance of high levels of MSI (MSI-H) in CRC patients in relation to fluorouracil-based chemotherapy. EXPERIMENTAL DESIGN: In three different institutions, 1,263 patients with CRC were tested for the presence of MSI, and CRC-specific survival was then analyzed in relation to MSI status, chemotherapy, and other clinical and pathologic variables. RESULTS: Two hundred and fifty-six tumors were MSI-H (20.3%): these were more frequently at a less advanced stage, right-sided, poorly differentiated, with mucinous phenotype, and expansive growth pattern than microsatellite stable carcinomas. Univariate and multivariate analyses of 5-year-specific survival revealed stage, tumor location, grade of differentiation, MSI, gender, and age as significant prognostic factors. The prognostic advantage of MSI tumors was particularly evident in stages II and III in which chemotherapy did not significantly affect the survival of MSI-H patients. Finally, we analyzed survival in MSI-H patients in relation to the presence of mismatch repair gene mutations. MSI-H patients with hereditary non-polyposis colorectal cancer showed a better prognosis as compared with sporadic MSI-H; however, in multivariate analysis, this difference disappeared. CONCLUSIONS: The type of genomic instability could influence the prognosis of CRC, in particular in stages II and III. Fluorouracil-based chemotherapy does not seem to improve survival among MSI-H patients. The survival benefit for patients with hereditary non-polyposis colorectal cancer is mainly determined by younger age and less advanced stage as compared with sporadic MSI-H counterpart.


Subject(s)
Colorectal Neoplasms/genetics , Genomic Instability , Microsatellite Repeats/genetics , Adaptor Proteins, Signal Transducing , Adenocarcinoma/diagnosis , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma, Mucinous/diagnosis , Adenocarcinoma, Mucinous/drug therapy , Adenocarcinoma, Mucinous/genetics , Antimetabolites, Antineoplastic/therapeutic use , Carrier Proteins/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/drug therapy , DNA-Binding Proteins/genetics , Female , Fluorouracil/therapeutic use , Humans , Male , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics , Neoplasm Staging , Nuclear Proteins/genetics , Prognosis , Prospective Studies , Survival Rate , Treatment Outcome
17.
Gastroenterology ; 129(3): 1047-59, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16143142

ABSTRACT

BACKGROUND & AIMS: Colon cancers with defective DNA mismatch repair (MMR) have peculiar molecular, pathologic, and clinical features, including high-level microsatellite instability, conspicuous lymphocytic infiltration, preferential location in the proximal colon, and better prognosis. Our aim was to characterize the transcriptional profile of this colon cancer subset. METHODS: An oligonucleotide microarray containing 12,625 probes was used to evaluate gene expression in 25 proximal colon cancers, 10 samples of normal colon mucosa, and 14 colon cancer cell lines. Transcriptional profiles of MMR-deficient cancers and cell lines were compared with those of their MMR-proficient counterparts. RESULTS: Unsupervised analysis of microarray data showed that MMR status exerts a predominant influence on the gene expression profile of proximal colon cancers. Hierarchical clustering divided the cancers into 2 groups corresponding almost perfectly with their MMR status. Supervised analysis identified numerous gene expression changes that represent a genetic signature of MMR-deficient colon cancers. Changes in genes involved in apoptosis and the immune response were consistent with the better prognosis of MMR-deficient cancers. In MMR-deficient cancers and cell lines, 4-1BBL, a crucial gene in the anti-tumor immune response, was, respectively, 2.4 and 6.0 times more expressed than in their MMR-proficient counterparts. This difference was confirmed by quantitative reverse-transcription polymerase chain reaction and flow cytometric assessment of 4-1BBL protein expression in colon cancer cell lines. Our analysis also showed novel possible gene targets of microsatellite instability. CONCLUSIONS: MMR inactivation produces distinct changes in the cellular messenger RNA pool, which is consistent with a unique tumorigenesis pathway.


Subject(s)
Base Pair Mismatch , Colonic Neoplasms/genetics , DNA Repair , Gene Expression Profiling , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Transcription, Genetic , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Carrier Proteins , Colon/pathology , Colon/physiology , DNA, Neoplasm/genetics , Female , Gene Expression Profiling/methods , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Male , Middle Aged , MutL Protein Homolog 1 , Sequence Deletion
18.
Am J Gastroenterol ; 100(10): 2280-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16181381

ABSTRACT

OBJECTIVES: Colorectal cancer (CRC) occurs rarely in young individuals (<45 yr) and represents one of the criteria for suspecting hereditary cancer families. In this study we evaluated clinical features and molecular pathways (chromosomal instability [CIN] and microsatellite instability [MSI]) in early-onset CRC of 71 patients. METHODS: Detailed family and personal history were obtained for each patient. Expression of APC, beta-catenin, p53, MLH1, MSH2, and MSH6 genes was evaluated by immunohistochemistry. MSI analysis was performed and constitutional main mutations of the mismatch repair (MMR) genes were searched by gene sequencing. RESULTS: Fourteen (19.7%) out of the 71 cases showed both MSI and altered expression of MMR proteins. In the 57 MSI-negative (MSI-) lesions altered expression of APC, beta-catenin, and p53 genes were found more frequently than in MSI-positive(MSI+) tumors. Seven (50%) out of the 14 patients with MSI+ tumors presented clinical features of Lynch syndrome (hereditary non-polyposis colorectal cancer [HNPCC]) and in all but one, constitutional mutations in MLH1 or MSH2 genes could be detected. The same mutations were also found in other family members. CONCLUSIONS: Our study demonstrates the involvement of CIN in a majority of early-onset colorectal tumors. Furthermore, we identified Lynch syndromes in seven cases (50%) of early-onset colorectal carcinomas with impairment of the MMR system. These results suggest that patients with early-onset CRC should be screened for hereditary cancer syndrome through clinical and molecular characterizations.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Carcinoma/metabolism , Colorectal Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adult , Age of Onset , Base Pair Mismatch/physiology , Carcinoma/genetics , Chromosomal Instability , Colorectal Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Female , Humans , Male , Microsatellite Repeats , Middle Aged , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , beta Catenin
19.
Gastroenterology ; 128(5): 1160-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15887099

ABSTRACT

BACKGROUND & AIMS: Germline mutations in the DNA mismatch repair (MMR) genes MSH2, MSH6, or MLH1 predispose to colorectal cancer (CRC) with an autosomal dominant inheritance pattern. The protein encoded by PMS2 is also essential for MMR; however, alterations in this gene have been documented only in extremely rare cases. We addressed this unexpected finding by analyzing a large series of CRCs. METHODS: Expression of MSH2, MSH6, MLH1, and PMS2 was studied by immunohistochemistry in 1048 unselected, consecutive CRCs. Where absence of MMR proteins was detected, microsatellite instability and cytosine methylation of the respective gene promoter were analyzed. The DNA of patients presenting with PMS2-deficient cancers was examined for germline and somatic alterations in the PMS2 gene. RESULTS: An aberrant pattern of MMR protein expression was detected in 13.2% of CRCs. Loss of expression of MSH2, MSH6, or MLH1 was found in 1.4%, 0.5%, and 9.8%, respectively. PMS2 deficiency accompanied by microsatellite instability was found in 16 cases (1.5%) with a weak family history of cancer. The PMS2 promoter was not hypermethylated in these cases. Despite interference of the PMS2 pseudogenes, we identified several heterozygous germline mutations in the PMS2 gene. CONCLUSIONS: PMS2 defects account for a small but significant proportion of CRCs and for a substantial fraction of tumors with microsatellite instability. However, the penetrance of heterozygous germline mutations in PMS2 is considerably lower than that of mutations in other MMR genes. The possible underlying causes of this unorthodox inheritance pattern are discussed.


Subject(s)
Adenoma/genetics , Adenoma/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Carrier Proteins , DNA Methylation , Female , Germ-Line Mutation , Heterozygote , Humans , Immunohistochemistry , Male , Microsatellite Repeats , Middle Aged , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
20.
Dis Colon Rectum ; 48(3): 567-71, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15719192

ABSTRACT

PURPOSE: Familial adenomatous polyposis is an autosomal dominant disease characterized by the presence of 100 or more colorectal adenomatous polyps. Mutations in the adenomatous polyposis coli gene are primarily responsible for the development of this disease. This study was designed to investigation of adenomatous polyposis coli (APC) gene mutations in members of familial adenomatous polyposis family to identify individuals at risk of the disease. METHODS: We examined one patient with familial adenomatous polyposis and 21 family members including one affected person from familial adenomatous polyposis and 20 nonsymptomatic persons. We studied E, D, F, and G segments of exon 15 of the adenomatous polyposis coli gene by heteroduplex analysis. RESULTS: We used silver staining method for staining. We found a mutation for five persons at segment F of exon 15 of the adenomatous polyposis coli gene. Two of them were affected by colorectal cancer, one of whom was the proband, and the other three were nonsymptomatic family members. The pathogenetic mutation was a T deletion at codon 1172, causing a frameshift in the adenomatous polyposis coli gene, as a result of the sequencing analysis of these cases. CONCLUSIONS: Investigation of adenomatous polyposis coli gene mutations is very important for the identification of genetic susceptibility to colorectal cancer and for the definition of tumor developing at an early stage. Furthermore, the identification of this mutation for the first time in a Turkish family will be useful to foster further studies on familial adenomatous polyposis in Turkey.


Subject(s)
Adenomatous Polyposis Coli/genetics , Genes, APC , Genetic Predisposition to Disease , Adult , Colorectal Neoplasms/genetics , DNA Mutational Analysis , Female , Heteroduplex Analysis , Humans , Male , Pedigree , Turkey
SELECTION OF CITATIONS
SEARCH DETAIL