Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Oncogene ; 26(2): 312-20, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-16819509

ABSTRACT

Serrated colorectal carcinomas (CRCs) are morphologically different from conventional CRCs and have been proposed to follow a distinct pathway of CRC formation. Despite studies of single molecular events in this tumor type, the diagnosis of serrated CRC relies on morphology and the putative unique biological character of these tumors has not been established. Here we show that the gene expression profiling of 37 CRCs separated serrated and conventional CRCs into two distinct branches in unsupervised hierarchical clustering (P-value 7.8 x 10(-7)), and revealed 201 differentially expressed genes representing potential biomarkers for serrated CRC. Immunohistochemistry was utilized to verify the key findings in the 37 CRCs examined by expression profiling, and a separate validation set of 37 serrated and 86 conventional CRCs was examined to evaluate the candidate biomarkers in an extended sample material. Ephrin receptor B2, hypoxia-inducible factor 1-alpha and patched appeared as proteins important for genesis of serrated CRC. This study establishes serrated CRCs as a biologically distinct subclass of CRC and represents a step forward in the molecular classification of these cancers. The study also provides a platform to understand the molecular basis of serrated CRC and in long term may contribute to the development of specific treatment options for this tumor type.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Adenoma/genetics , Adenoma/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Colonic Polyps/pathology , Colorectal Neoplasms/metabolism , DNA, Neoplasm , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Microsatellite Repeats , Middle Aged , Oligonucleotide Array Sequence Analysis
2.
J Med Genet ; 43(6): 523-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16155190

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a tumour predisposition syndrome caused by heterozygous germline mutations in the fumarate hydratase (FH) gene. The condition is characterised by predisposition to benign leiomyomas of the skin and the uterus, renal cell carcinoma (RCC), and uterine leiomyosarcoma (ULMS). To comprehensively examine the cancer risk and tumour spectrum in Finnish FH mutation positive families, genealogical and cancer data were obtained from 868 individuals. The cohort analysis of the standardised incidence ratios (SIR) was analysed from 256 individuals. FH mutation status was analysed from all available individuals (n = 98). To study tumour spectrum in FH mutation carriers, loss of the wild type allele was analysed from all available tumours (n = 22). The SIR was 6.5 for RCC and 71 for ULMS. The overall cancer risk was statistically significantly increased in the age group of 15-29 years, consistent with features of cancer predisposition families in general. FH germline mutation was found in 55% of studied individuals. Most RCC and ULMS tumours displayed biallelic inactivation of FH, as did breast and bladder cancers. In addition, several benign tumours including atypical uterine leiomyomas, kidney cysts, and adrenal gland adenomas were observed. The present study confirms with calculated risk ratios the association of early onset RCC and ULMS with FH germline mutations in Finns. Some evidence for association of breast and bladder carcinoma with HLRCC was obtained. The data enlighten the organ specific malignant potential of HLRCC.


Subject(s)
Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Neoplasms/genetics , Adolescent , Adult , Age Factors , Aged , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Cohort Studies , Female , Finland , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Leiomyomatosis/diagnosis , Leiomyomatosis/genetics , Male , Middle Aged , Neoplasms/diagnosis , Phenotype , Risk Factors
3.
Br J Cancer ; 92(12): 2240-8, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15956967

ABSTRACT

The majority of microsatellite instable (MSI) colorectal cancers are sporadic, but a subset belongs to the syndrome hereditary non-polyposis colorectal cancer (HNPCC). Microsatellite instability is caused by dysfunction of the mismatch repair (MMR) system that leads to a mutator phenotype, and MSI is correlated to prognosis and response to chemotherapy. Gene expression signatures as predictive markers are being developed for many cancers, and the identification of a signature for MMR deficiency would be of interest both clinically and biologically. To address this issue, we profiled the gene expression of 101 stage II and III colorectal cancers (34 MSI, 67 microsatellite stable (MSS)) using high-density oligonucleotide microarrays. From these data, we constructed a nine-gene signature capable of separating the mismatch repair proficient and deficient tumours. Subsequently, we demonstrated the robustness of the signature by transferring it to a real-time RT-PCR platform. Using this platform, the signature was validated on an independent test set consisting of 47 tumours (10 MSI, 37 MSS), of which 45 were correctly classified. In a second step, we constructed a signature capable of separating MMR-deficient tumours into sporadic MSI and HNPCC cases, and validated this by a mathematical cross-validation approach. The demonstration that this two-step classification approach can identify MSI as well as HNPCC cases merits further gene expression studies to identify prognostic signatures.


Subject(s)
Adenocarcinoma/genetics , Colonic Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Gene Expression/genetics , Adult , Aged , Aged, 80 and over , Base Pair Mismatch/genetics , Chromosomal Instability/genetics , DNA Repair/genetics , Gene Expression Profiling , Humans , Microsatellite Repeats/genetics , Middle Aged , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests
4.
Gut ; 51(1): 56-9, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12077092

ABSTRACT

BACKGROUND AND AIMS: Loss of DNA sequences from chromosome 18q21 is a major genetic change in colorectal tumorigenesis. Multiple genes have been identified in this area. One of these, DPC4 (deleted in pancreatic cancer 4, also known as SMAD4), is mutated in a minor subset of colorectal carcinomas as well as in germlines of humans predisposed to colon tumours. PATIENTS AND METHODS: The involvement of SMAD4 in sporadic colorectal neoplasia was evaluated by immunohistochemistry in 53 unselected cases and 27 cases displaying microsatellite instability. RESULTS: SMAD4 expression was absent in 20 of 53 (38%) unselected colorectal carcinomas, and reduced in another 15 (28%) cases. However, 26 of 27 cancers displaying microsatellite instability and TGF-betaIIR mutations were positive for SMAD4 immunostaining. CONCLUSIONS: Loss of SMAD4 expression may play a more prominent role in colon cancer than anticipated based on genetic evidence, but not in mutator phenotype tumours.


Subject(s)
Chromosomes, Human, Pair 18/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Trans-Activators/genetics , Colorectal Neoplasms/chemistry , DNA Mutational Analysis , DNA-Binding Proteins/analysis , Genetic Markers , Humans , Immunohistochemistry/methods , Loss of Heterozygosity , Microsatellite Repeats , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Smad4 Protein , Trans-Activators/analysis
5.
Int J Legal Med ; 116(1): 43-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11924708

ABSTRACT

Massive haemorrhage due to rupture of single pancreatic or peripancreatic vessels is a very rare but potentially lethal complication of acute and chronic pancreatitis. The splenic, gastroduodenal, and pancreatoduodenal arteries are the more commonly involved vessels, and rupture occurs mostly as a complication of large mature pseudocysts. We report a sudden death due to massive bleeding caused by rupture of the great pancreatic artery (arteria pancreatica magna), a complication of a small immature pseudocyst, in a 49-year-old male alcoholic with inactive chronic pancreatitis.


Subject(s)
Death, Sudden/etiology , Pancreas/blood supply , Pancreatic Pseudocyst/complications , Pancreatitis, Alcoholic/complications , Arteries , Death, Sudden/pathology , Humans , Male , Middle Aged , Pancreas/pathology , Pancreatic Pseudocyst/pathology , Pancreatitis, Alcoholic/pathology , Rupture, Spontaneous
6.
Cancer Res ; 61(19): 6991-5, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585722

ABSTRACT

Hypermethylation of the MLH1 promoter underlies most sporadic colorectal cancers with microsatellite instability (MSI). To investigate the role of hypermethylation in the normal colonic mucosa as a possible precursor lesion, we studied 700 bp upstream of MLH1 covering 51 CpG sites. We found partially methylated alleles in 15 of 34 (44%) patients <60 years of age and 20 of 24 (83%) patients > or =80 years of age (P = 0.0026). Fully methylated alleles were present in 18 of 33 (55%) patients with MSI+ tumors but in only 18 of 90 (20%) patients with MSI- tumors (P = 0.00019). By in situ analysis, methylation was patchy and located mainly in the cryptal regions close to the lumen. We conclude that the spread of methylation in the MLH1 promoter in the normal colonic mucosa is closely associated with age and the development of sporadic MSI+ colorectal cancers.


Subject(s)
Colon/physiology , Colorectal Neoplasms/genetics , DNA Methylation , Intestinal Mucosa/physiology , Microsatellite Repeats/genetics , Neoplasm Proteins/genetics , Precancerous Conditions/genetics , Adaptor Proteins, Signal Transducing , Adult , Age Factors , Aged , Aged, 80 and over , Carrier Proteins , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Humans , In Situ Hybridization/methods , Middle Aged , MutL Protein Homolog 1 , Nuclear Proteins , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , Sequence Analysis, DNA
7.
Am J Pathol ; 159(4): 1293-300, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11583957

ABSTRACT

Juvenile polyposis syndrome (JPS; OMIM 174900) is a rare disorder which is characterized by the presence of hamartomatous polyps throughout the gastrointestinal tract and an increased risk of gastrointestinal malignancy. Mutations of the SMAD4 gene on chromosome 18q21.1 have been shown to cause a subset of JPS cases, with estimates ranging from 20% to >50%. Characterization of the genes that cause the remainder of JPS cases relies on the certainty that SMAD4 is not the causative gene. We have undertaken a comprehensive analysis of germline SMAD4 mutations in a cohort of JPS patients to define the spectrum of mutations that cause JPS. We have analyzed a series of polyps from these patients for SMAD4 protein expression. We have also performed a blinded assessment of polyp material to look for morphological differences between polyps from patients with and without a germline SMAD4 mutation. The results indicate that almost all germline SMAD4 mutations are readily detectable by screening genomic DNA using polymerase chain reaction-based methods; SMAD4 can be excluded as the causative gene in the majority of our JPS cohort. Loss of SMAD4 expression occurs in most polyps from SMAD4 mutation carriers, even those with missense germline mutations. SMAD4 loss in polyps is, however, not a feature of cases that are not caused by SMAD4 mutations, indicating that these polyps develop along a SMAD4-independent pathway. The morphology of polyps from SMAD4 mutation carriers is subtly different from other JPS polyps, notably including a more prominent epithelial component in the former.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Mutation , Polyps/genetics , Polyps/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Amino Acid Sequence/genetics , Base Sequence/genetics , Child , Gastrointestinal Neoplasms/pathology , Heterozygote , Humans , Molecular Sequence Data , Mutation/genetics , Polyps/pathology , Smad4 Protein , Syndrome
8.
Am J Pathol ; 159(3): 825-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549574

ABSTRACT

Little has been known about the molecular background of familial multiple cutaneous leiomyomatosis (MCL). We report here a clinical, histopathological, and molecular study of a multiple cutaneous leiomyomatosis kindred with seven affected members. This detailed study revealed strong features of a recently described cancer predisposition syndrome, hereditary leiomyomatosis and renal cell cancer (HLRCC). The family was compatible with linkage to the HLRCC locus in 1q. Also, all seven cutaneous leiomyomas derived from the proband and analyzed for loss of heterozygosity displayed loss of the wild-type allele, confirming the association with a susceptibility gene in chromosome 1q. One individual had had renal cell cancer at the age of 35 years. This tumor displayed a rare papillary histopathology, which appears to be characteristic for HLRCC. The derived linkage, loss of heterozygosity, and clinical data suggest that MCL and HLRCC are a single disease with a variable phenotype. The possibility that members of leiomyomatosis families are predisposed to renal cell cancer should be taken into account.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , Neoplasms, Multiple Primary , Skin Neoplasms/genetics , Adult , Carcinoma, Renal Cell/pathology , Female , Genetic Linkage , Genetic Predisposition to Disease , Humans , Kidney Medulla/pathology , Kidney Neoplasms/pathology , Loss of Heterozygosity , Male , Pedigree
9.
Am J Hum Genet ; 69(4): 704-11, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11536076

ABSTRACT

Juvenile polyposis syndrome (JPS) is an inherited hamartomatous-polyposis syndrome with a risk for colon cancer. JPS is a clinical diagnosis by exclusion, and, before susceptibility genes were identified, JPS could easily be confused with other inherited hamartoma syndromes, such as Bannayan-Riley-Ruvalcaba syndrome (BRRS) and Cowden syndrome (CS). Germline mutations of MADH4 (SMAD4) have been described in a variable number of probands with JPS. A series of familial and isolated European probands without MADH4 mutations were analyzed for germline mutations in BMPR1A, a member of the transforming growth-factor beta-receptor superfamily, upstream from the SMAD pathway. Overall, 10 (38%) probands were found to have germline BMPR1A mutations, 8 of which resulted in truncated receptors and 2 of which resulted in missense alterations (C124R and C376Y). Almost all available component tumors from mutation-positive cases showed loss of heterozygosity (LOH) in the BMPR1A region, whereas those from mutation-negative cases did not. One proband with CS/CS-like phenotype was also found to have a germline BMPR1A missense mutation (A338D). Thus, germline BMPR1A mutations cause a significant proportion of cases of JPS and might define a small subset of cases of CS/BRRS with specific colonic phenotype.


Subject(s)
Abnormalities, Multiple/genetics , Germ-Line Mutation/genetics , Hamartoma Syndrome, Multiple/genetics , Intestinal Polyps/genetics , Protein Serine-Threonine Kinases , Receptors, Growth Factor , Receptors, Transforming Growth Factor beta/genetics , Abnormalities, Multiple/physiopathology , Bone Morphogenetic Protein Receptors, Type I , Colonic Neoplasms/complications , Colonic Neoplasms/genetics , DNA Mutational Analysis , Genotype , Hamartoma Syndrome, Multiple/complications , Hamartoma Syndrome, Multiple/physiopathology , Humans , Intestinal Polyps/complications , Intestinal Polyps/physiopathology , Loss of Heterozygosity/genetics , Microsatellite Repeats/genetics , Phenotype , Receptors, Transforming Growth Factor beta/chemistry , Syndrome
10.
Am J Pathol ; 159(1): 35-42, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438451

ABSTRACT

The distal half of chromosome arm 18q is frequently lost in ovarian carcinoma. To define the putative tumor suppressor locus/loci more precisely we performed allelic analysis with 27 polymorphic microsatellite markers located at 18q12.3-q23 in 64 serous and 9 mucinous ovarian carcinomas. Fifty-nine percent of the serous carcinomas, but only one (11%) of mucinous carcinomas, showed allelic loss at one or more loci (P = 0.018). In serous carcinomas, deletions were found to be associated with tumor grade and poor survival. The highest frequency of losses was detected at the distal part, 18q22-q23. Two minimal common regions of loss (MCRL) were identified at this region: MCRL1 between D18S465 and D18S61 at 18q22 (3.9 cM) and MCRL2 between D18S462 and D18S70 at 18q23 (5.8 cM). At 18q21.1, proximal to the MCRLs, there are three candidate tumor suppressor genes: SMAD4 (DPC4), SMAD2, and DCC. Their protein expression was studied by immunohistochemistry in normal ovarian tissue and serous carcinomas. Lost or very weak expression of SMAD4, SMAD2 and DCC was found in 28, 28, and 30% of serous carcinomas, respectively. Comparison of allelic loss and protein expression status indicated that none of these genes alone could be the target for the frequent allelic loss at 18q21.1. Together, these genes may account for a substantial proportion of the events, but not all of them. Thus, we propose that the frequent allelic loss at 18q is because of the effect of multiple genes, and there is at least one as yet unidentified tumor suppressor gene at 18q residing distal to SMAD4, SMAD2, and DCC involved in serous ovarian carcinoma.


Subject(s)
Alleles , Carcinoma/genetics , Chromosome Mapping , Chromosomes, Human, Pair 18/genetics , Genes, Tumor Suppressor/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Proteins , Adenocarcinoma, Mucinous/genetics , Carcinoma/metabolism , Cell Adhesion Molecules/metabolism , DCC Receptor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genes, DCC/genetics , Humans , Immunohistochemistry , Loss of Heterozygosity , Ovarian Neoplasms/metabolism , Receptors, Cell Surface , Smad2 Protein , Smad4 Protein , Trans-Activators/genetics , Trans-Activators/metabolism
11.
Cancer Res ; 61(11): 4545-9, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389088

ABSTRACT

Hereditary nonpolyposis colorectal cancer (HNPCC) is an autosomal dominant cancer predisposition syndrome caused by germ-line mutations in DNA mismatch repair genes. It is relevant to identify HNPCC patients because colonoscopic screening of individuals with HNPCC mutations reduces cancer morbidity and mortality. Microsatellite instability (MSI) is characteristic of HNPCC tumors. A panel of five markers (BAT25, BAT26, D2S123, D5S346, and D17S250, the so-called Bethesda markers) has been proposed for screening for MSI. To test a hypothesis that the use of BAT26 alone is feasible in screening for MLH1/MSH2 mutation-positive HNPCC patients, we compared the MSI results of 494 colorectal cancer patients obtained using BAT26 with results obtained using the Bethesda markers. BAT26 was able to identify all 27 mutation-positive individuals in this series. The marker failed to identify 2 high MSI tumors and 20 low MSI tumors, all of which expressed MLH1, MSH2, and MSH6 when scrutinized by immunohistochemistry.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA-Binding Proteins , Microsatellite Repeats/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Carrier Proteins , Genetic Testing/methods , Germ-Line Mutation , Humans , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/genetics , Nuclear Proteins , Proto-Oncogene Proteins/genetics
13.
Cancer Res ; 61(7): 2813-5, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306449

ABSTRACT

Hereditary nonpolyposis colorectal cancer syndrome is associated with an inherited predisposition to primarily colorectal cancer (CRC) and endometrial cancer (EC); however, the biological basis of the organ involvement remains unknown. As an attempt to explore whether the expression levels of MLH1, MSH2, and MSH6 may play a role, we used immunohistochemistry to study 42 ECs and 35 CRCs from patients carrying the same predisposing mutations. Among MSH2 mutation carriers, MLH1 was expressed in both tumor types, whereas MSH2 and, in many cases, also MSH6, were absent. Remarkably, among MLH1 mutation carriers, 54% of ECs (21 of 39), but none of the CRCs (0 of 32), lacked the MSH2 and/or MSH6 protein in addition to lacking MLH1 protein expression. These results demonstrate a marked difference between hereditary nonpolyposis colorectal cancer-related CRCs and ECs and suggest that the development of the latter tumors is selectively associated with the MSH2/MSH6 protein complex deficiency.


Subject(s)
Colonic Neoplasms/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , DNA-Binding Proteins/biosynthesis , Endometrial Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Adaptor Proteins, Signal Transducing , Carrier Proteins , Colonic Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA-Binding Proteins/deficiency , Dimerization , Endometrial Neoplasms/genetics , Female , Germ-Line Mutation , Humans , Immunohistochemistry , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Nuclear Proteins , Proto-Oncogene Proteins/deficiency
14.
Fam Cancer ; 1(2): 87-92, 2001.
Article in English | MEDLINE | ID: mdl-14574003

ABSTRACT

Hereditary nonpolyposis colorectal cancer syndrome (HNPCC) is an autosomal dominant condition accounting for 2-5% of all colorectal carcinomas as well as a small subset of endometrial, upper urinary tract and other gastrointestinal cancers. An assay to detect the underlying defect in HNPCC, inactivation of a DNA mismatch repair enzyme, would be useful in identifying HNPCC probands. Monoclonal antibodies against hMLH1 and hMSH2, two DNA mismatch repair proteins which account for most HNPCC cancers, are commercially available. This study sought to investigate the potential utility of these antibodies in determining the expression status of these proteins in paraffin-embedded formalin-fixed tissue and to identify key technical protocol components associated with successful staining. A set of 20 colorectal carcinoma cases of known hMLH1 and hMSH2 mutation and expression status underwent immunoperoxidase staining at multiple institutions, each of which used their own technical protocol. Staining for hMSH2 was successful in most laboratories while staining for hMLH1 proved problematic in multiple labs. However, a significant minority of laboratories demonstrated excellent results including high discriminatory power with both monoclonal antibodies. These laboratories appropriately identified hMLH1 or hMSH2 inactivation with high sensitivity and specificity. The key protocol point associated with successful staining was an antigen retrieval step involving heat treatment and either EDTA or citrate buffer. This study demonstrates the potential utility of immunohistochemistry in detecting HNPCC probands and identifies key technical components for successful staining.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Genetic Testing , Immunoenzyme Techniques/standards , Neoplasm Proteins/genetics , Adaptor Proteins, Signal Transducing , Antibodies, Monoclonal , Base Pair Mismatch , Carrier Proteins , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/immunology , DNA Repair , Diagnosis, Differential , Humans , Immunoenzyme Techniques/statistics & numerical data , International Cooperation , Laboratories/standards , MutL Protein Homolog 1 , Neoplasm Proteins/immunology , Nuclear Proteins , Observer Variation , Pedigree , Reproducibility of Results
15.
Br J Cancer ; 83(8): 1015-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10993648

ABSTRACT

The chromosome region 18q21 is frequently deleted in colorectal cancers. Three candidate tumour suppressor genes, DCC, SMAD4 and SMAD2, map to this region. The SMAD4(DPC4) gene was recently identified as a candidate pancreatic cancer suppressor gene. It is also a gene for juvenile polyposis tumour predisposition syndrome. Somatic SMAD4 mutations have been detected in some colorectal carcinomas. However, the frequency of these mutations is relatively low, and whether SMAD4 plays a key role in colorectal tumorigenesis is still unclear. In addition to loss of chromosomal material and intragenic mutations there is a third mechanism, DNA methylation, which may have an important role in gene inactivation. In the present study, we examined whether promoter hypermethylation could be a mechanism for SMAD4 inactivation. In total, 42 colorectal tumours were selected for the methylation analysis and no evidence of promoter hypermethylation was found. Our result suggests that hypermethylation of the SMAD4 promoter region is not a frequent event in colorectal tumorigenesis.


Subject(s)
Adenocarcinoma/genetics , Chromosome Deletion , Chromosomes, Human, Pair 18 , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Mutation , Promoter Regions, Genetic , Trans-Activators/genetics , Adenocarcinoma/pathology , Base Sequence , Chromosome Mapping , Colorectal Neoplasms/pathology , DNA Methylation , Humans , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Staging , Smad4 Protein
16.
J Clin Oncol ; 18(11): 2193-200, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10829038

ABSTRACT

PURPOSE: Cancer morbidity and mortality can be dramatically reduced by colonoscopic screening of individuals with the hereditary nonpolyposis colorectal cancer (HNPCC) syndrome, creating a need to identify HNPCC. We studied how HNPCC identification should be carried out on a large scale in a sensitive and efficient manner. PATIENTS AND METHODS: Colorectal cancer specimens from consecutive newly diagnosed patients were studied for microsatellite instability (MSI). Germline mutations in the MLH1 and MSH2 genes were searched for in MSI(+) individuals. RESULTS: Among 535 colorectal cancer patients, 66 (12%) were MSI(+). Among these, 18 (3.4% of the total) had disease-causing germline mutations in MLH1 or MSH2. Among these 18 patients, five were less than 50 years old, seven had a previous or synchronous colorectal or endometrial cancer, and 15 had at least one first-degree relative with colorectal or endometrial cancer. Notably, 17 (94%) of 18 patients had at least one of these three features, which were present in 22% of all 535 patients. Combining these data with a previous study of 509 patients, mutation-positive HNPCC accounts for 28 (2.7%) of 1,044 cases of colorectal cancer, predicting a greater than one in 740 incidence of mutation-positive individuals in this population. CONCLUSION: Large-scale molecular screening for HNPCC can be done by the described two-stage procedure of MSI determination followed by mutation analysis. Efficiency can be greatly improved by using three high-risk features to select 22% of all patients for MSI analysis, whereby only 6% need to have mutation analysis. Sensitivity is only slightly impaired by this procedure.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA, Neoplasm/analysis , Genetic Markers , Germ-Line Mutation , Adult , Aged , Aged, 80 and over , Base Pair Mismatch , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , DNA Mutational Analysis , DNA Repair , Female , Finland/epidemiology , Humans , Male , Microsatellite Repeats , Middle Aged , Mutation, Missense , Polymerase Chain Reaction , Registries
17.
Am J Pathol ; 156(5): 1773-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10793088

ABSTRACT

Microsatellite instability (MSI) is a hallmark of hereditary nonpolyposis colorectal cancer, and in these patients, results from inherited defects in DNA mismatch repair genes, mostly MSH2 and MLH1. MSI also occurs in 15% of sporadic colorectal cancers, but in these tumors, its basis is less well characterized. We investigated 46 sporadic MSI+ colorectal cancers for changes in MSH2 and MLH1 protein expression, followed by the analysis of somatic mutation, loss of heterozygosity (LOH), and promoter hypermethylation as possible underlying defects. Most cases (36/46, 78%) showed lost or reduced MLH1 expression. Among these, a majority (83%) was associated with MLH1 promoter hypermethylation, whereas the rates of LOH and somatic mutation of MLH1 were 24% and 13%, respectively. Hypermethylation and LOH were inversely correlated, suggesting that they had alternative functions in the inactivation of MLH1. MSH2 expression was lost in 7/46 (15%), and of these, 2 (29%) showed LOH and/or somatic mutation of MSH2. We conclude that most sporadic MSI+ colorectal cancers have an MLH1-associated etiology and that epigenetic modification is a major mechanism of MLH1 inactivation. Moreover, we found a significantly lower prevalence for MLH1 promoter hypermethylation in hereditary nonpolyposis colorectal cancer tumors with MLH1 germline mutations (12/26, 46%), which might explain some differences that are known to occur in the clinicopathological characteristics and tumorigenic pathways between sporadic and hereditary MSI+ colorectal cancers.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins , Microsatellite Repeats/genetics , Neoplasm Proteins/genetics , Adaptor Proteins, Signal Transducing , Carrier Proteins , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Methylation , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Immunohistochemistry , Loss of Heterozygosity , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Mutation , Neoplasm Proteins/analysis , Nuclear Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics
18.
Cancer Res ; 60(3): 546-8, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10676634

ABSTRACT

LKB1 serine/threonine kinase is a gene for Peutz-Jeghers cancer predisposition syndrome. Most studies have detected a low frequency of LKB1 defects in sporadic cancer. A notable exception is a recent report describing frequent, mostly missense type, LKB1 mutations in Korean distal colorectal tumors. To clarify the role of LKB1 in colon cancer, we scrutinized 50 left-sided Korean and Finnish specimens. No somatic mutations were found. The seven Korean somatic missense mutations reported previously were functionally analyzed, and five were found not to alter LKB1 kinase activity. One of these changes was found to be a germ-line polymorphism. LKB1 involvement in distal colorectal cancer is not common.


Subject(s)
Colorectal Neoplasms/etiology , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Colorectal Neoplasms/genetics , Humans , Mutation, Missense , Protein Kinases/metabolism
19.
Proc Natl Acad Sci U S A ; 97(3): 1236-41, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10655514

ABSTRACT

It is difficult to observe human tumor progression as precursor lesions are systematically removed. Alternatives to direct observations, commonly used to reveal the hidden past of species and populations, are sequence comparisons or molecular clocks. Noncoding microsatellite (MS) loci were employed as molecular tumor clocks in 13 human mutator phenotype (MSI(+)) colorectal tumors. Quantitative analysis revealed that specific patterns of somatic MS mutations accumulate with division after loss of mismatch repair (MMR). Tumors had unique patterns of MS mutation, and, therefore, based on this model, each tumor had its own unique history. Loss of MMR occurred very early relative to terminal clonal expansion, with an estimated average of 2,300 divisions since loss of MMR and 280 divisions since expansion. Contrary to the classical adenoma-cancer sequence, MSI(+) adenomas were nearly as old as cancers (2,000 versus 2,400 divisions since loss of MMR). Negative clinical examinations preceded six tumors, independently documenting an absence of visible precursors during early MSI(+) adenoma or cancer progression. These findings further extend a window beyond visible progression since loss of MMR appears to start a genetic phase involving clone sizes or phenotypes below a threshold of clinical detection. This previously occult prologue before visible neoplasia is longer and therefore likely more important than generally appreciated.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Colonic Neoplasms/genetics , Colonic Polyps/genetics , Colorectal Neoplasms/genetics , Rectal Neoplasms/genetics , Adenocarcinoma/pathology , Adenoma/pathology , Cell Division , Clone Cells/pathology , Colonic Neoplasms/pathology , Colonic Polyps/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mutational Analysis , DNA Repair/genetics , DNA, Neoplasm/genetics , Disease Progression , Humans , Male , Microsatellite Repeats , Precancerous Conditions/genetics , Rectal Neoplasms/pathology
20.
Am J Pathol ; 155(6): 1849-53, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10595914

ABSTRACT

Hereditary nonpolyposis colorectal cancer (HNPCC) is the most common of the well-defined colorectal cancer syndromes, accounting for at least 2% of the total colorectal cancer burden and carrying a greater than 80% lifetime risk of cancer. Significant reduction in cancer morbidity and mortality can be accomplished by appropriate clinical cancer screening of HNPCC patients with mutations in mismatch repair (MMR) genes. Thus, it is desirable to identify individuals who are mutation-positive. In individuals with cancer, mutation detection can be accomplished relatively efficiently by germline mutation analysis of individuals whose cancers show microsatellite instability (MSI). This study was designed to assess the feasibility of screening colorectal adenoma patients for HNPCC in the same manner. Among 378 adenoma patients, six (1.6%) had at least one MSI adenoma. Five out of the six patients (83%) had a germline MMR gene mutation. We conclude that MSI analysis is a useful method of prescreening colorectal adenoma patients for HNPCC.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms/genetics , DNA, Neoplasm/analysis , Genetic Markers , Germ-Line Mutation , Adenoma/pathology , Adult , Aged , Base Pair Mismatch , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mutational Analysis , DNA Repair , Humans , Microsatellite Repeats , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...