Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Oncogene ; 38(33): 6109-6122, 2019 08.
Article in English | MEDLINE | ID: mdl-31308487

ABSTRACT

About 80% of colorectal cancers (CRCs) have chromosomal instability, which is an integral part of aggressive malignancy development, but the importance of specific copy number aberrations (CNAs) in modulating gene expression, particularly within the framework of clinically relevant molecular subtypes, remains mostly elusive. We performed DNA copy number profiling of 257 stage I-IV primary CRCs and integrative gene expression analysis in 151 microsatellite stable (MSS) tumors, focusing on high-level amplifications and the effect of CNAs on the characteristics of the gene expression-based consensus molecular subtypes (CMS). The results were validated in 323 MSS tumors from TCGA. Novel recurrent high-level amplifications (≥15 additional copies) with a major impact on gene expression were found for TOX3 (16q) at 1.5% frequency, as well as for CCND2 (12p) and ANXA11 (10q) at 1% frequency, in addition to the well-known targets ERBB2 (17q) and MYC (8q). Focal amplifications with ≥15 or ≥5 additional copies of at least one of these regions were associated with a poor overall survival among patients with stage I-III MSS CRCs (multivariable hazard ratio ≥3.2, p ≤ 0.01). All high-level amplifications were focal and had a more consistent relationship with gene expression than lower amplitude and/or broad-range amplifications, suggesting specific targeting during carcinogenesis. Genome-wide, copy number driven gene expression was enriched for pathways characteristic of the CMS2-epithelial/canonical subtype, including DNA repair and cell cycle progression. Furthermore, 50% of upregulated genes in CMS2-epithelial/canonical MSS CRCs were driven by CNAs, an enrichment compared with the other CMS groups, and associated with the stronger correspondence between CNAs and gene expression in malignant epithelial cells than in the cells of the tumor microenvironment (fibroblasts, endothelial cells, leukocytes). In conclusion, we identify novel recurrent amplifications with impact on gene expression in CRC and provide the first evidence that CMS2 may have a stronger copy-number related genetic basis than subtypes more heavily influenced by gene expression signals from the tumor microenvironment.


Subject(s)
Colorectal Neoplasms/classification , Colorectal Neoplasms/genetics , DNA Copy Number Variations/physiology , Gene Amplification/physiology , Transcriptome , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/mortality , DNA Mutational Analysis , Female , Gene Dosage/physiology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Microsatellite Instability , Microsatellite Repeats , Molecular Diagnostic Techniques/methods , Survival Analysis , Tumor Microenvironment/genetics
2.
PLoS One ; 10(4): e0122391, 2015.
Article in English | MEDLINE | ID: mdl-25879218

ABSTRACT

OBJECTIVE: We sought to investigate various molecular subtypes defined by genomic instability that may be related to early death and recurrence in colon cancer. METHODS: We sought to investigate various molecular subtypes defined by instability at microsatellites (MSI), changes in methylation patterns (CpG island methylator phenotype, CIMP) or copy number variation (CNV) in 8 genes. Stage II-III colon cancers (n = 64) were investigated by methylation-specific multiplex ligated probe amplification (MS-MLPA). Correlation of CNV, CIMP and MSI, with mutations in KRAS and BRAFV600E were assessed for overlap in molecular subtypes and early recurrence risk by uni- and multivariate regression. RESULTS: The CIMP phenotype occurred in 34% (22/64) and MSI in 27% (16/60) of the tumors, with noted CIMP/MSI overlap. Among the molecular subtypes, a high CNV phenotype had an associated odds ratio (OR) for recurrence of 3.2 (95% CI 1.1-9.3; P = 0.026). Losses of CACNA1G (OR of 2.9, 95% CI 1.4-6.0; P = 0.001), IGF2 (OR of 4.3, 95% CI 1.1-15.8; P = 0.007), CDKN2A (p16) (OR of 2.0, 95% CI 1.1-3.6; P = 0.024), and RUNX3 (OR of 3.4, 95% CI 1.3-8.7; P = 0.002) were associated with early recurrence, while MSI, CIMP, KRAS or BRAF V600E mutations were not. The CNV was significantly higher in deceased patients (CNV in 6 of 8) compared to survivors (CNV in 3 of 8). Only stage and loss of RUNX3 and CDKN2A were significant in the multivariable risk-model for early recurrence. CONCLUSIONS: A high copy number variation phenotype is a strong predictor of early recurrence and death, and may indicate a dose-dependent relationship between genetic instability and outcome. Loss of tumor suppressors RUNX3 and CDKN2A were related to recurrence-risk and warrants further investigation.


Subject(s)
Colonic Neoplasms/genetics , Core Binding Factor Alpha 3 Subunit/genetics , DNA Copy Number Variations , Genes, p16 , Genomic Instability , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Male , Middle Aged , Recurrence , Survival Analysis
3.
PLoS One ; 9(1): e86657, 2014.
Article in English | MEDLINE | ID: mdl-24466191

ABSTRACT

BACKGROUND: In colorectal cancer a distinct subgroup of tumours demonstrate the CpG island methylator phenotype (CIMP). However, a consensus of how to score CIMP is not reached, and variation in definition may influence the reported CIMP prevalence in tumours. Thus, we sought to compare currently suggested definitions and cut-offs for methylation markers and how they influence CIMP classification in colon cancer. METHODS: Methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA), with subsequent fragment analysis, was used to investigate methylation of tumour samples. In total, 31 CpG sites, located in 8 different genes (RUNX3, MLH1, NEUROG1, CDKN2A, IGF2, CRABP1, SOCS1 and CACNA1G) were investigated in 64 distinct colon cancers and 2 colon cancer cell lines. The Ogino gene panel includes all 8 genes, in addition to the Weisenberger panel of which only 5 of the 8 genes included were investigated. In total, 18 alternative combinations of scoring of CIMP positivity on probe-, gene-, and panel-level were analysed and compared. RESULTS: For 47 samples (71%), the CIMP status was constant and independent of criteria used for scoring; 34 samples were constantly scored as CIMP negative, and 13 (20%) consistently scored as CIMP positive. Only four of 31 probes (13%) investigated showed no difference in the numbers of positive samples using the different cut-offs. Within the panels a trend was observed that increasing the gene-level stringency resulted in a larger difference in CIMP positive samples than increasing the probe-level stringency. A significant difference between positive samples using 'the most stringent' as compared to 'the least stringent' criteria (20% vs 46%, respectively; p<0.005) was demonstrated. CONCLUSIONS: A statistical significant variation in the frequency of CIMP depending on the cut-offs and genes included in a panel was found, with twice as many positives samples by least compared to most stringent definition used.


Subject(s)
Colonic Neoplasms/genetics , CpG Islands/genetics , DNA Methylation/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Humans , Phenotype
5.
Mol Med ; 19: 286-93, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23979710

ABSTRACT

Lymph node (LN) harvest is influenced by several factors, including tumor genetics. Microsatellite instability (MSI) is associated with improved node harvest, but the association to other genetic factors is largely unknown. Research methods included a prospective series of stage I-III colon cancer patients undergoing ex vivo sentinel-node sampling. The presence of MSI, KRAS mutations in codons 12 and 13, and BRAF V600E mutations was analyzed. Uni- and multivariate regression models for node sampling were adjusted for clinical, pathological and molecular features. Of 204 patients, 67% had an adequate harvest (≥ 12 nodes). Adequate harvest was highest in patients whose tumors exhibited MSI (79%; odds ratio [OR] 2.5, 95% confidence interval [CI] 1.2-4.9; P = 0.007) or were located in the proximal colon (73%; 2.8, 1.5-5.3; P = 0.002). In multiple linear regression, MSI was a significant predictor of the total LN count (P = 0.02). Total node count was highest for cancers with MSI and no KRAS/BRAF mutations. The independent association between MSI and a high LN count persisted for stage I and II cancers (P = 0.04). Tumor location in the proximal colon was the only significant predictor of an adequate LN harvest (adjusted OR 2.4, 95% CI 1.2-4.9; P = 0.01). An increase in the total number of nodes harvested was not associated with an increase in nodal metastasis. In conclusion, number of nodes harvested is highest for cancers of the proximal colon and with MSI. The nodal harvest associated with MSI is influenced by BRAF and KRAS genotypes, even for cancers of proximal location. Mechanisms behind the molecular diversity and node yield should be further explored.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Lymph Nodes/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Microsatellite Instability , Middle Aged , Mutation , Neoplasm Staging , Prognosis , Prospective Studies , Proto-Oncogene Proteins p21(ras) , Young Adult
6.
Dig Surg ; 30(1): 12-25, 2013.
Article in English | MEDLINE | ID: mdl-23595116

ABSTRACT

Colorectal cancer (CRC) is, for sporadic forms, most strongly related to lifestyle factors. The epidemic of obesity and physical inactivity has great impact on disease patterns. Likewise, an altered metabolism has consequences at the cellular and molecular level with implications for cancer initiation and growth. Understanding the genetic hallmarks of cancers has improved over the years and now also includes cancer metabolic reprogramming. The initiation of cancer through genetic instability, including chromosomal instability, microsatellite instability and epigenetic silencing through the CpG island methylator phenotype follows pathways with distinct clinical, pathological, and genetic characteristics. These can potentially be used for molecular classification and comprehensive tumor profiling for improved diagnostics, prognosis and treatment in CRC. For one, epidermal growth factor receptor-directed treatment now considerably prolongs survival in metastatic disease, but defining the true responders from non-responders has emerged as complex. Further, the use of both non-steroidal anti-inflammatory drugs including cyclooxygenase-2 inhibitors is associated with a decreased incidence of adenoma and reduced mortality rate of CRC. This review gives a brief yet updated overview of the current understanding of CRC as a genetic and molecular disease with potential for clinical pathways of prevention, improved prediction and better prognosis in the future.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Adenoma/genetics , Adenoma/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Chromosomal Instability/genetics , CpG Islands/genetics , DNA Methylation , Epigenomics , Genotype , Humans , Mutation , Phenotype , Prognosis
7.
Discov Med ; 14(76): 207-14, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23021375

ABSTRACT

The promise of individualized treatment is gradually being fulfilled, and targeted therapy is becoming a powerful strategy to treat selected patients based on their molecular profile. For metastatic colorectal cancer (mCRC) patients anti-EGFR (epidermal growth factor receptor) targeted therapy has markedly improved disease control and survival. However, only a subgroup of patients with mCRC respond to anti-EGFR treatment, and selecting the patients with a positive effect from treatment is important for both the patient and the society. Patients with mutations in the KRAS gene are known as non-responders to anti-EGFR treatment and, consequently, KRAS testing has been employed in routine clinical practice for patient selection. However, a large number of the KRAS wildtype patients do not respond to this treatment. The molecular mechanism underlying response is not fully understood, and other members of the KRAS-BRAF pathway and PI3K-AKT pathway are investigated as predictive biomarkers. Furthermore, concordance of mutation status of primary tumors and their corresponding hepatic or pulmonary metastases, as well as treatment-induced mutations, possess another challenge for properly tailoring the appropriate therapy to this patient group. In this review, molecular biomarkers involved in prediction of response to anti-EGFR treatment are discussed.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , ras Proteins/metabolism , Biomarkers/metabolism , Cell Survival , Genes, ras , Humans , Models, Biological , Mutation , Neoplasm Metastasis
8.
Discov Med ; 12(66): 393-404, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22127110

ABSTRACT

Colorectal cancer (CRC) is a common malignancy and remains a formidable health burden in the world. Surgery is the cornerstone for curative treatment, and surgical technique has changed considerably for both colon and rectum cancer over the past decades. Laparoscopic approach has become standard for colon cancer in many institutions, and technical developments including robotic-assisted surgery is evolving for rectal cancer. Advances in (neo)adjuvant chemo-radiotherapy have increased survival and reduced recurrences, and the addition of targeted therapies has prolonged life in metastatic disease to considerable extent. The genetic understanding of the disease has led to introduction of molecular classification proposals, which exemplifies knowledge translated from basic science to clinical care. As CRC is a highly prevalent disease it lends itself to both prevention and screening, yet the best and most optimal approach has yet to be determined. Further, the advance of currently available diagnostic, staging, and treatment regimens for both local and metastatic disease calls for improved stratification and prognostication to better allocate resources and justify associated costs while reducing morbidity and even mortality from the disease. Clearly, the need to better stratify and correctly prescribe the best medication for the right patients to optimize outcome, reduce adverse events and costs, and rationalize resources is a mandatory task for current health systems. It is hoped that the increased understanding of colorectal cancer as a heterogeneous disease developing on similar yet diverse genetic and molecular mechanisms will further enhance the way these patients are managed in the future.


Subject(s)
Colorectal Neoplasms/therapy , Colorectal Neoplasms/economics , Colorectal Neoplasms/pathology , Genetic Predisposition to Disease , Genomic Instability , Health Care Rationing , Humans , Neoplasm Staging
9.
BMC Cancer ; 11: 211:1-11, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21624110

ABSTRACT

BACKGROUND: Frequent failure and severe side effects of current sarcoma therapy warrants new therapeutic approaches. The small-molecule MDM2 antagonist Nutlin-3a activates the p53 pathway and efficiently induces apoptosis in tumours with amplified MDM2 gene and overexpression of MDM2 protein. However, the majority of human sarcomas have normal level of MDM2 and the therapeutic potential of MDM2 antagonists in this group is still unclear. We have investigated if Nutlin-3a could be employed to augment the response to traditional therapy and/or reduce the genotoxic burden of chemotherapy. METHODS: A panel of sarcoma cell lines with different TP53 and MDM2 status were treated with Nutlin-3a combined with Doxorubicin, Methotrexate or Cisplatin, and their combination index determined. RESULTS: Clear synergism was observed when Doxorubicin and Nutlin-3a were combined in cell lines with wild-type TP53 and amplified MDM2, or with Methotrexate in both MDM2 normal and amplified sarcoma cell lines, allowing for up to tenfold reduction of cytotoxic drug dose. Interestingly, Nutlin-3a seemed to potentiate the effect of classical drugs as Doxorubicin and Cisplatin in cell lines with mutated TP53, but inhibited the effect of Methotrexate. CONCLUSION: The use of Nutlin in combination with classical sarcoma chemotherapy shows promising preclinical potential, but since clear biomarkers are still lacking, clinical trials should be followed up with detailed tumour profiling.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxins/pharmacology , Imidazoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
J Surg Oncol ; 104(7): 792-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21547915

ABSTRACT

BACKGROUND AND OBJECTIVES: The aim of the study was to evaluate the incidence of peritoneal carcinomatosis (PC) in a prospectively recorded series of colon cancer patients from a defined cohort and to compare clinicopathological characteristics, survival, and TP53 mutation status in primary tumors from patients with and without PC. METHODS: Clinical data from all colon cancer patients admitted in 1993-2006 were registered prospectively (n = 1,124). In a subset of PC patients, DNA was retrieved from tumor tissue and TP53 mutations analyzed and compared to the mutation status in a historical series. RESULTS: In the prospective series 10% of female and 7% of male patients had PC (P = 0.05). The PC patients were younger than those without PC (median 4 years, P = 0.002). The incidence of PC was 10.3% and 6.2% (P = 0.03) in patients with primary tumors in the right and left colon, respectively. TP53 was mutated in 57% of the PC patients as compared to 41% in the series of patients without PC (P = 0.05). CONCLUSIONS: The incidence of PC was higher in right-sided colon cancer and among women. PC patients were younger than non-PC patients, and PC was independently associated with TP53 mutation in the primary tumor.


Subject(s)
Carcinoma/epidemiology , Carcinoma/genetics , Colonic Neoplasms/pathology , Genes, p53/genetics , Mutation , Peritoneal Neoplasms/epidemiology , Peritoneal Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Carcinoma/secondary , Case-Control Studies , Colonic Neoplasms/epidemiology , DNA Mutational Analysis , DNA, Neoplasm/analysis , Female , Humans , Incidence , Male , Middle Aged , Multivariate Analysis , Norway/epidemiology , Peritoneal Neoplasms/secondary , Prospective Studies , Sex Distribution
11.
12.
Amino Acids ; 41(4): 875-84, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20191297

ABSTRACT

Nuclear localization of the metastasis-associated protein S100A4 has been shown to correlate with advanced disease stage in primary colorectal carcinomas (CRC), but nuclear function and its relevance for the metastatic capacity of tumor cells is still unclear. Among several nuclear interacting protein partners suggested for S100A4, the tumor suppressor protein p53 has attracted particular interest, and previous studies suggest direct and indirect modes of interaction between the two proteins. The present study was undertaken to assess coexpression and potential interaction in CRC. TP53 mutational status and S100A4 expression were investigated in a selected series of primary CRC specimens (n = 40) and cell lines (n = 17) using DNA sequencing, western blot, and double immunostaining. Additionally, S100A4 and p53 were experimentally up- and down-regulated in vitro to assess reciprocal effects. For the first time, S100A4 and p53 coexpression was demonstrated in individual CRC cells, with nuclear colocalization as a particularly interesting feature. In contrast to previous studies, no correlation was observed between TP53 mutational status and S100A4 expression, and no evidence was obtained to support reciprocal regulation between the two molecules in the HCT116 isogenic cell line model. In conclusion, S100A4 and p53 were shown to be colocalized in individual nuclei of CRC cells, and it might be speculated whether the proteins interact in this subcellular compartment.


Subject(s)
Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , S100 Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mutation , Rabbits , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics
13.
Int J Mol Sci ; 12(12): 9426-39, 2011.
Article in English | MEDLINE | ID: mdl-22272141

ABSTRACT

Colorectal cancer is a major health burden, and a leading cause of cancer-related deaths in industrialized countries. The steady improvements in surgery and chemotherapy have improved survival, but the ability to identify high- and low-risk patients is still somewhat poor. Molecular biology has, over the years, given insight into basic principles of colorectal cancer initiation and development. These findings include aberrations increasing risk of tumor development, genetic changes associated with the stepwise progression of the disease, and errors predicting response to a specific treatment. Potential biomarkers in colorectal cancer are extensively studied, and how the molecular aberrations relate to clinical features. Yet, little of this knowledge has been possible to transfer into clinical practice. In this review, an overview of colorectal cancer genetics will be given, as well as how aberrations found in this tumor type are proposed as biomarkers for risk prediction, as diagnostic tools, for prognosis or prediction of treatment outcome.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/diagnosis , Colorectal Neoplasms/diagnosis , Epigenesis, Genetic , Animals , Carcinoma/genetics , Colorectal Neoplasms/genetics , Humans , Microsatellite Repeats , Mutation
14.
PLoS One ; 5(11): e13978, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-21103049

ABSTRACT

The incidence of colorectal cancer (CRC) increases with age and early onset indicates an increased likelihood for genetic predisposition for this disease. The somatic genetics of tumor development in relation to patient age remains mostly unknown. We have examined the mutation status of five known cancer critical genes in relation to age at diagnosis, and compared the genomic complexity of tumors from young patients without known CRC syndromes with those from elderly patients. Among 181 CRC patients, stratified by microsatellite instability status, DNA sequence changes were identified in KRAS (32%), BRAF (16%), PIK3CA (4%), PTEN (14%) and TP53 (51%). In patients younger than 50 years (n = 45), PIK3CA mutations were not observed and TP53 mutations were more frequent than in the older age groups. The total gene mutation index was lowest in tumors from the youngest patients. In contrast, the genome complexity, assessed as copy number aberrations, was highest in tumors from the youngest patients. A comparable number of tumors from young (<50 years) and old patients (>70 years) was quadruple negative for the four predictive gene markers (KRAS-BRAF-PIK3CA-PTEN); however, 16% of young versus only 1% of the old patients had tumor mutations in PTEN/PIK3CA exclusively. This implies that mutation testing for prediction of EGFR treatment response may be restricted to KRAS and BRAF in elderly (>70 years) patients. Distinct genetic differences found in tumors from young and elderly patients, whom are comparable for known clinical and pathological variables, indicate that young patients have a different genetic risk profile for CRC development than older patients.


Subject(s)
Colorectal Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics , Age Factors , Age of Onset , Aged , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , Kaplan-Meier Estimate , Male , Microsatellite Instability , Middle Aged , Mutation , Neoplasm Staging
15.
Mol Cancer ; 9: 100, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20459617

ABSTRACT

BACKGROUND: Estimates suggest that up to 30% of colorectal cancers (CRC) may develop due to an increased genetic risk. The mean age at diagnosis for CRC is about 70 years. Time of disease onset 20 years younger than the mean age is assumed to be indicative of genetic susceptibility. We have compared high resolution tumor genome copy number variation (CNV) (Roche NimbleGen, 385 000 oligo CGH array) in microsatellite stable (MSS) tumors from two age groups, including 23 young at onset patients without known hereditary syndromes and with a median age of 44 years (range: 28-53) and 17 elderly patients with median age 79 years (range: 69-87). Our aim was to identify differences in the tumor genomes between these groups and pinpoint potential susceptibility loci. Integration analysis of CNV and genome wide mRNA expression data, available for the same tumors, was performed to identify a restricted candidate gene list. RESULTS: The total fraction of the genome with aberrant copy number, the overall genomic profile and the TP53 mutation spectrum were similar between the two age groups. However, both the number of chromosomal aberrations and the number of breakpoints differed significantly between the groups. Gains of 2q35, 10q21.3-22.1, 10q22.3 and 19q13.2-13.31 and losses from 1p31.3, 1q21.1, 2q21.2, 4p16.1-q28.3, 10p11.1 and 19p12, positions that in total contain more than 500 genes, were found significantly more often in the early onset group as compared to the late onset group. Integration analysis revealed a covariation of DNA copy number at these sites and mRNA expression for 107 of the genes. Seven of these genes, CLC, EIF4E, LTBP4, PLA2G12A, PPAT, RG9MTD2, and ZNF574, had significantly different mRNA expression comparing median expression levels across the transcriptome between the two groups. CONCLUSIONS: Ten genomic loci, containing more than 500 protein coding genes, are identified as more often altered in tumors from early onset versus late onset CRC. Integration of genome and transcriptome data identifies seven novel candidate genes with the potential to identify an increased risk for CRC.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Profiling , Genetic Predisposition to Disease , Adult , Age of Onset , Aged , Aged, 80 and over , Comparative Genomic Hybridization , DNA Mutational Analysis , Female , Genomic Instability , Humans , Male , Middle Aged , Tumor Suppressor Protein p53/genetics
16.
Pharmacogenet Genomics ; 19(10): 751-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19741569

ABSTRACT

PURPOSE: The pathogenesis of testicular germ cell tumor (TGCT) remains unknown. The aim of this study was to evaluate the pathogenic role of functional polymorphisms in detoxification enzymes among TGCT patients, through association studies of constitutive genotypes and medical parameters before and after chemotherapy. EXPERIMENTAL DESIGN: Germline deletion polymorphisms in the glutathione S-transferase mu 1 (GSTM1) and the GST theta 1 (GSTT1), and a functional single nucleotide polymorphism in GST pi 1 (GSTP1, Ile105Val), were analyzed in TGCT survivors (TCSs) (n = 675) and controls (n = 189). Statistical analyses were performed for the genotype distributions between the TCSs and control populations, and between the genotypes and clinicopathological parameters of the TCSs. RESULTS: The GST genotypes showed comparable distributions among the TCSs and the control population. However, the genotype combination GSTT1positive/GSTP1-GG or GSTP1-AG/GSTM1positive was more frequent among the TCSs [P = 0.050, odds ratio (OR): 1.47, 95% confidence interval (CI): 0.998-2.165]. The combined genotype GSTT1positive/GSTP1AA/GSTM1positive was associated with decreased risk of development of pure embryonal carcinoma (P = 0.009, OR: 0.309, 95% CI: 0.122-0.784) and the GSTP1-A-allele (i.e. genotypes GSTP-AA or GSTP-AG) was also associated with decreased risk for development of pure teratoma (P = 0.032, OR: 0.326, 95% CI: 0.122-0.873). Furthermore, the GSTP1-A-allele was overrepresented within the 'good prognosis group' (P = 0.032, OR: 2.407, 95% CI: 1.060-5.469), whereas the GSTM1nulltype was associated with the extent of TC qualifying as 'poor prognosis group' (P = 0.025, OR: 2.839, 95% CI: 1.104-7.301). The GSTP1-AG genotype was associated with necrosis in the tumor's post-chemotherapy histology (P = 0.001, OR: 16.087, 95% CI: 1.930-134.087). Failure, after platinum-based chemotherapy, was associated with the GSTT1positive/GSTP-AA or GSTP-GG/GSTM1-positive genotype (P = 0.019, OR: 2.168, 95% CI: 1.130-4.160). CONCLUSION: This study confirms an association between the GSTP1-G-allele and TGCT. Combinations of GST genotypes were associated with primary and post-chemotherapy tumor histology, and prognostic group presentation.


Subject(s)
Germinoma/genetics , Glutathione Transferase/genetics , Testicular Neoplasms/drug therapy , Testicular Neoplasms/genetics , Adolescent , Adult , Aged , Genetic Predisposition to Disease , Genotype , Germinoma/pathology , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Glutathione Transferase/metabolism , Humans , Male , Middle Aged , Neoplasm Staging , Polymorphism, Genetic , Testicular Neoplasms/pathology
17.
J Transl Med ; 6: 13, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18346269

ABSTRACT

BACKGROUND: Tumor-derived aberrantly methylated DNA might serve as diagnostic biomarkers for cancer, but so far, few such markers have been identified. The aim of the present study was to investigate the potential of the MAL (T-cell differentiation protein) gene as an early epigenetic diagnostic marker for colorectal tumors. METHODS: Using methylation-specific polymerase chain reaction (MSP) the promoter methylation status of MAL was analyzed in 218 samples, including normal mucosa (n = 44), colorectal adenomas (n = 63), carcinomas (n = 65), and various cancer cell lines (n = 46). Direct bisulphite sequencing was performed to confirm the MSP results. MAL gene expression was investigated with real time quantitative analyses before and after epigenetic drug treatment. Immunohistochemical analysis of MAL was done using normal colon mucosa samples (n = 5) and a tissue microarray with 292 colorectal tumors. RESULTS: Bisulphite sequencing revealed that the methylation was unequally distributed within the MAL promoter and by MSP analysis a region close to the transcription start point was shown to be hypermethylated in the majority of colorectal carcinomas (49/61, 80%) as well as in adenomas (45/63, 71%). In contrast, only a minority of the normal mucosa samples displayed hypermethylation (1/23, 4%). The hypermethylation of MAL was significantly associated with reduced or lost gene expression in in vitro models. Furthermore, removal of the methylation re-induced gene expression in colon cancer cell lines. Finally, MAL protein was expressed in epithelial cells of normal colon mucosa, but not in the malignant cells of the same type. CONCLUSION: Promoter hypermethylation of MAL was present in the vast majority of benign and malignant colorectal tumors, and only rarely in normal mucosa, which makes it suitable as a diagnostic marker for early colorectal tumorigenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , DNA Methylation , Membrane Transport Proteins/genetics , Myelin Proteins/genetics , Proteolipids/genetics , Colorectal Neoplasms/metabolism , Humans , Myelin and Lymphocyte-Associated Proteolipid Proteins , Promoter Regions, Genetic
18.
Pharmacogenet Genomics ; 17(2): 127-36, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17301692

ABSTRACT

BACKGROUND AND OBJECTIVE: Many environmental and genetic factors influence the development of chemoresistance. The goal of this study was to characterize the genetic variation in the ABCB1, GSTM1, GSTT1 and GSTP1 genes, as well as the haplotype structure in the ABCB1 gene. METHODS: Variants in these genes were studied in 109 healthy controls and 93 breast cancer cases, both of Caucasian origin. The cases were analyzed in relation to TP53 mutation status and response to doxorubicin. Both single and multiple single nucleotide polymorphism analyses were performed. RESULTS: Chi-square analyses revealed a significant association between TP53 mutation status and both the GA genotype of ABCB1 exon 11 (Ser400Asn) and the GG genotype of GSTP1 (Ile105Val; P<0.01 and P<0.05, respectively). Multifactor dimensionality reduction showed that carriers of the combined GG genotype for GSTP1 and the GG for ABCB1 exon 11 had the highest chance of acquiring a mutation in the TP53 gene (P<0.02). Haplotype analysis of ABCB1 revealed a significantly different distribution of haplotypes between the breast cancer cases and the controls (P<0.01). A specific haplotype association to TP53 mutation (P<0.01) distant metastases (P<0.05) and estrogen receptor status (P<0.05) was also observed in the case group. CONCLUSION: An association between polymorphisms in GSTP1 and ABCB1 and risk of acquiring intratumoral TP53 mutations suggests the existence of putative predisposing genotype backgrounds. The degree of linkage disequilibrium in the ABCB1 gene was higher in healthy individuals, whereas haplotypes in the cases seemed degenerated by a number of low frequency variants. This observation may either point to the existence of a protective haplotype in the controls or may underline the importance of the accumulation of low frequency variants as susceptibility factors.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Genetic Predisposition to Disease , Glutathione Transferase/genetics , Organic Anion Transporters/genetics , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Protein p53/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Case-Control Studies , Chi-Square Distribution , Doxorubicin/therapeutic use , Female , Genotype , Glutathione S-Transferase pi/genetics , Humans , Linkage Disequilibrium/genetics , Middle Aged , Mutation/genetics , Norway , Recombination, Genetic , Treatment Outcome
19.
Mol Cancer ; 5: 33, 2006 Sep 04.
Article in English | MEDLINE | ID: mdl-16952311

ABSTRACT

BACKGROUND: In order to gain new insights into the molecular mechanisms involved in prostate cancer, we performed array-based comparative genomic hybridization (aCGH) on a series of 46 primary prostate carcinomas using a 1 Mbp whole-genome coverage platform. As chromosomal comparative genomic hybridization (cCGH) data was available for these samples, we compared the sensitivity and overall concordance of the two methodologies, and used the combined information to infer the best of three different aCGH scoring approaches. RESULTS: Our data demonstrate that the reliability of aCGH in the analysis of primary prostate carcinomas depends to some extent on the scoring approach used, with the breakpoint estimation method being the most sensitive and reliable. The pattern of copy number changes detected by aCGH was concordant with that of cCGH, but the higher resolution technique detected 2.7 times more aberrations and 15.2% more carcinomas with genomic imbalances. We additionally show that several aberrations were consistently overlooked using cCGH, such as small deletions at 5q, 6q, 12p, and 17p. The latter were validated by fluorescence in situ hybridization targeting TP53, although only one carcinoma harbored a point mutation in this gene. Strikingly, homozygous deletions at 10q23.31, encompassing the PTEN locus, were seen in 58% of the cases with 10q loss. CONCLUSION: We conclude that aCGH can significantly improve the detection of genomic aberrations in cancer cells as compared to previously established whole-genome methodologies, although contamination with normal cells may influence the sensitivity and specificity of some scoring approaches. Our work delineated recurrent copy number changes and revealed novel amplified loci and frequent homozygous deletions in primary prostate carcinomas, which may guide future work aimed at identifying the relevant target genes. In particular, biallelic loss seems to be a frequent mechanism of inactivation of the PTEN gene in prostate carcinogenesis.


Subject(s)
Chromosome Aberrations , Genome, Human/genetics , Nucleic Acid Hybridization/methods , Prostatic Neoplasms/genetics , Gene Dosage/genetics , Humans , In Situ Hybridization, Fluorescence/methods , Male , Mutation/genetics , Tumor Suppressor Protein p53/genetics
20.
Neurosci Lett ; 322(2): 83-6, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11958849

ABSTRACT

We investigated the association of Parkinson's disease (PD) with tau gene H1 haplotypes in the Norwegian population. In a sample of 96 unrelated PD cases and 68 control subjects, we observed an increased risk of PD for persons with the tau H1 haplotype (odds ratio=5.52; 95% confidence interval: 2.64-11.10; P=2.17x10(-6)). Findings provide evidence that tau participates in the PD pathogenic process and demonstrate the value of isolated populations in mapping complex traits.


Subject(s)
Haplotypes/genetics , Parkinson Disease/genetics , tau Proteins/genetics , Adult , Aged , Aged, 80 and over , Confidence Intervals , Female , Genotype , Humans , Male , Middle Aged , Norway/epidemiology , Odds Ratio , Parkinson Disease/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...