Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 87(10): 1128-35, 2002 Nov 04.
Article in English | MEDLINE | ID: mdl-12402152

ABSTRACT

Elevated expression and/or activity of c-Src, the prototype of the Src family of protein tyrosine kinases, is associated with the development of human colon cancer. However, despite the known pleiotropic effects of these kinases in promoting (a) cell growth downstream of growth factor receptors, and (b) the dynamic regulation of integrin adhesions in fibroblast model systems, their precise role in epithelial cancer cells is unknown. Here we addressed whether elevated expression and activity of cellular Src alters cell proliferation and/or cell-matrix adhesion in cancer cells from the Fidler model of colorectal metastasis. Although elevated Src correlates with ability to metastasise to the liver after intrasplenic injection, we found that this was not linked to enhanced growth, either in vitro or in vivo as sub-cutaneous tumours. However, elevated Src was associated with enhanced attachment to extracellular matrix. In addition, adhesion to fibronectin, was suppressed by agents that inhibited Src activity, while enforced elevation of Src in non-metastatic cells was sufficient to stimulate adhesion to fibronectin and enhanced assembly of adhesion complexes, without influencing cell growth. Thus, we conclude that one role of elevated Src in human colon cancer cells is to modulate integrin-dependent cell-matrix attachment and formation of adhesion structures, which may, in turn, influence cell motility and integrin-dependent cellular responses.


Subject(s)
Cell-Matrix Junctions/physiology , Colorectal Neoplasms/pathology , Oncogene Protein pp60(v-src)/metabolism , Protein-Tyrosine Kinases/physiology , Cell Adhesion , Colorectal Neoplasms/metabolism , Humans , Neoplasm Metastasis , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured
2.
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
4.
Methods Mol Med ; 50: 175-83, 2001.
Article in English | MEDLINE | ID: mdl-21318826

ABSTRACT

While Peutz-Jeghers syndrome (PJS) has been acknowledged as a clinical entity for decades (1,2), the molecular background for the disease has been unraveled only very recently. PJS has two cardinal features: First, many but not all patients display mucocutaneous melanin pigmentation that is most prominently seen around the mouth, but can also be present for example in the buccal mucosa, lips, palms, feet, and in the anal region. Second, the patients have a predisposition to hamartomatous intestinal polyps. These lesions can occur anywhere in the gastrointestinal tract, but are most commonly seen in the small intestine (3). Tumor predisposition is not limited to intestinal hamartomas. The patients have a relatively unfocused increased risk of cancer, which has been reported to be 10- to 18-fold of that of the general population. Especially the relative risk for breast and gynecologic cancers is high (4,5). Other sites possibly involved include at least small and large intestine, and pancreas (3). Benign testicular tumors also occur commonly in the syndrome (6). Some of the malignant tumors may arise from the benign hamartomatous lesions, which appear to have some malignant potential at least in the context of PJS (7-10).

5.
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
6.
Oncogene ; 19(1): 164-8, 2000 Jan 06.
Article in English | MEDLINE | ID: mdl-10644993

ABSTRACT

Germ-line mutations of the LKB1 gene cause Peutz-Jeghers syndrome (PJS) characterized by mucocutaneous pigmentation, predisposition to benign hamartomas of the gastrointestinal tract and also to several types of tumors. However, somatic mutations of this gene are very rare. To examine inactivation of LKB1 by epigenetic mechanisms, we investigated a series of primary tumors and cancer cell lines, for hypermethylation affecting the CpG island located in the 5' region of the LKB1 gene using Methylation-specific PCR (MSP). First, we screened 51 cancer cell lines. Only three colorectal and one cervical carcinoma cell lines were methylated at LKB1, and loss of the LKB1 transcript was demonstrated. Treatment with the demethylating agent 5-aza-2'-deoxycytidine restored LKB1 expression. To address the incidence of LKB1 epigenetic inactivation in primary tumors, we analysed colorectal, breast, gastric, pancreatic, thyroid, bladder and testicular carcinomas (n=195). Normal tissues from the mentioned organs were unmethylated in this region. Among the described tumors, only one colorectal carcinoma and three testicular tumors displayed LKB1 promoter hypermethylation. Further study of those histological types more commonly associated with PJS, demonstrated that LKB1 promoter hypermethylation was present in five of 11 (45%) papillary breast carcinomas. Finally, in three patients with a strong family story suggestive of PJS disease, abnormal LKB1 methylation was found in four of 22 (18%) hamartomatous polyps lesions. Our findings provide an alternative pathway for inactivation of the LKB1 tumor suppressor gene involving promoter hypermethylation.


Subject(s)
DNA Methylation , Neoplasms/genetics , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Genes, Tumor Suppressor , Humans , Promoter Regions, Genetic , Tumor Cells, Cultured
7.
Genes Chromosomes Cancer ; 26(1): 54-61, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10441006

ABSTRACT

Juvenile polyposis (JP) is a dominantly inherited condition characterized by the development of multiple hamartomatous tumors, juvenile polyps, in the gastrointestinal tract. The aim of this study was to clarify the role of SMAD4 in JP. DNA from four unrelated JP kindreds and three sporadic JP cases was available for mutation screening. Two truncating defects (one in a familial and one in a sporadic case) and one missense change (in a familial case) that was absent in 55 control samples were detected. To study the possibility that germline mutations in other genes encoding different components of the TGF-beta signaling pathway may be present in these JP patients, mutation analyses of the SMAD2, SMAD3, and SMAD7 genes were also performed. No mutations of these genes were detected in any of the patients. Our results confirm that SMAD4 is a gene predisposing to JP and suggest the existence of further JP loci other than the SMAD2, SMAD3, or SMAD7 genes. Genes Chromosomes Cancer 26:54-61, 1999.


Subject(s)
Adenomatous Polyposis Coli/genetics , Adolescent , Adult , Base Sequence , Child , Chromosomes, Human, Pair 18/genetics , DNA Mutational Analysis , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Family Health , Female , Genetic Markers , Humans , Male , Middle Aged , Mutation , Pedigree , Smad2 Protein , Smad3 Protein , Smad4 Protein , Smad7 Protein , Trans-Activators/genetics
8.
J Med Genet ; 36(5): 365-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10353780

ABSTRACT

Germline mutations of the LKB1 (STK11) serine/threonine kinase gene (chromosome 19p13.3) cause Peutz-Jeghers syndrome, which is characterised by hamartomas of the gastrointestinal tract and typical pigmentation. Peutz-Jeghers syndrome carries an overall risk of cancer that may be up to 20 times that of the general population. Here, we report the results of a screen for germline LKB1 mutations by DNA sequencing in 12 Peutz-Jeghers patients (three sporadic and nine familial cases). Mutations were found in seven (58%) cases, in exons 1, 2, 4, 6, and 9. Five of these mutations, two of which are identical, are predicted to lead to a truncated protein (three frameshifts, two nonsense changes). A further mutation is an in frame deletion of 6 bp, resulting in a deletion of lysine and asparagine; the second of these amino acids is conserved between species. The seventh mutation is a missense change in exon 2, converting lysine to arginine, affecting non-conserved amino acids and of uncertain functional significance. Despite the fact that Peutz-Jeghers syndrome is usually an early onset disease with characteristic clinical features, predictive and diagnostic testing for LKB1 mutations will be useful for selected patients in both familial and non-familial contexts.


Subject(s)
Germ-Line Mutation , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Adult , Base Sequence , DNA Mutational Analysis , Exons/genetics , Humans
9.
Am J Pathol ; 154(3): 677-81, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079245

ABSTRACT

Germline mutations of LKB1/Peutz-Jeghers syndrome gene predispose carriers to hamartomatous polyposis of the gastrointestinal tract as well as to cancer of different organ systems. Although Peutz-Jeghers syndrome patients frequently present with neoplasms of the colon, stomach, small intestine, pancreas, breast, ovaries, and cervix, somatic mutations appear to be rare in the sporadic tumor types thus far studied (colorectal, gastric, testicular, and breast cancers). To evaluate whether somatic mutations of LKB1 contribute to the tumorigenesis of yet unstudied tumor types, we screened 14 cell lines and 129 tumor specimens from different cancers for a genetic defect in LKB1. Six melanoma and eight myeloma cell lines were scrutinized for LKB1 somatic mutations by genomic sequencing. No changes were found in the coding LKB1 sequence and exon/intron boundaries. Next, we analyzed 12 pancreatic, 8 gastric, 12 ovarian granulosa cell, 26 cervical, 28 lung, 24 soft tissue, and 19 renal tumors by single-strand conformational polymorphism analysis. Three changes in LKB1 coding nucleotide sequence were identified. One base pair deletion at A957 and G958 substitution by T occurred in a cervical adenocarcinoma sample, resulting in a frameshift and premature stop codon at position 335. Substitution of A581 by T occurred in a lung adenocarcinoma sample, resulting in the change of aspartic acid at position 194 to valine. A loss of another allele was detected in this sample. One silent change, C1257T, was found in a pancreatic carcinoma sample. The changes were not present in the matched normal tissue DNA samples. Our results suggest that mutational inactivation of LKB1 is a rare event in most sporadic tumor types.


Subject(s)
Mutation/genetics , Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Amino Acid Substitution/genetics , Base Sequence/genetics , DNA, Neoplasm/genetics , Humans , Molecular Sequence Data , Polymorphism, Genetic/genetics , Polymorphism, Single-Stranded Conformational , Tumor Cells, Cultured
10.
Hum Mol Genet ; 8(1): 45-51, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9887330

ABSTRACT

Germline mutations in LKB1 have been reported to underlie familial Peutz-Jeghers syndrome (PJS) with intestinal hamartomatous polyps and an elevated risk of various neoplasms. To investigate the prevalence of LKB1 germline mutations in PJS more generally, we studied samples from 33 unrelated PJS patients including eight non-familial sporadic patients, 20 familial patients and five patients with unknown family history. Nineteen germline mutations were identified, 12 (60%) in familial and four (50%) in sporadic cases. LKB1 mutations were not detected in 14 (42%) patients, indicating that the existence of additional minor PJS loci cannot be excluded. LKB1 is predicted to encode a serine/threonine kinase. To demonstrate the putative Lkb1 kinase function and to study the consequences of LKB1 mutations in PJS and sporadic tumors, we have analyzed the kinase activity of wild-type and mutant Lkb1 proteins. Interestingly, while most of the small deletions or missense mutations resulted in loss-of-function alleles, one missense mutation (G163D) previously identified in a sporadic testicular tumor demonstrated severely impaired but detectable kinase activity.


Subject(s)
Germ-Line Mutation , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , Testicular Neoplasms/genetics , AMP-Activated Protein Kinase Kinases , Alleles , Base Sequence , DNA/genetics , DNA Primers/genetics , Female , Humans , Male , Peutz-Jeghers Syndrome/enzymology , Point Mutation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Testicular Neoplasms/enzymology
12.
Cancer Res ; 58(10): 2087-90, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9605748

ABSTRACT

Germ-line mutations in a serine/threonine kinase gene, LKB1, were recently shown to underlie Peutz-Jeghers syndrome (PJS), a hereditary disorder that predisposes to benign and malignant tumors of multiple organ systems. Most mutations that have been described thus far dramatically change the predicted protein and are likely to be of an inactivating nature. This observation and a previous observation that the LKB1 locus is often deleted in PJS polyps suggest that the gene may function as a tumor suppressor. We examined whether somatic mutations in this gene are present in sporadic carcinomas of the colon and testis, tumors that are characteristic of PJS. First, 20 randomly selected colorectal and 28 testicular tumors were analyzed by single-strand conformation polymorphism analysis. No mutations in LKB1 were found in colorectal tumors. One testicular tumor displayed a heterozygous missense type variant, in which glycine 163 was changed to aspartic acid. This change was absent in the DNA of normal tissue. To better focus our efforts, we tested 75 additional colon carcinomas for loss of heterozygosity at 19p, where LKB1 is localized. Of 75 samples analyzed, 50 were informative with a closely linked marker, D19S886, and 13 (26%) of these displayed loss of heterozygosity. The 13 tumors were scrutinized for LKB1 mutations by genomic sequencing. This analysis revealed no changes. Together, these findings suggest that somatic mutations of LKB1 are not frequent in colorectal and testicular cancer.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , DNA, Neoplasm/analysis , Mutation/genetics , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Testicular Neoplasms/genetics , AMP-Activated Protein Kinase Kinases , Base Sequence , Exons/genetics , Humans , Introns/genetics , Loss of Heterozygosity/genetics , Male , Molecular Sequence Data , Sequence Alignment
13.
J Med Genet ; 35(1): 42-4, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9475093

ABSTRACT

A locus for Peutz-Jeghers syndrome (PJS) was recently mapped to chromosome 19p13.3. Each of 12 families studied was compatible with linkage to the marker D19S886. We have analysed 20 further families and found that the majority of these are consistent with a PJS gene on 19p13.3. Three families were, however, unlinked to 19p13.3 and none of the available PJS polyps from these families showed allele loss at D19S886. There were no obvious clinicopathological or ethnic differences between the 19p13.3 linked and unlinked families. There appears, therefore, to be a major PJS locus on chromosome 19p13.3 and the possibility exists of a minor locus (or loci) elsewhere.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Genetic Linkage , Peutz-Jeghers Syndrome/genetics , Female , Genetic Heterogeneity , Genetic Markers , Humans , Male , Pedigree
14.
Nature ; 391(6663): 184-7, 1998 Jan 08.
Article in English | MEDLINE | ID: mdl-9428765

ABSTRACT

Studies of hereditary cancer syndromes have contributed greatly to our understanding of molecular events involved in tumorigenesis. Here we investigate the molecular background of the Peutz-Jeghers syndrome (PJS), a rare hereditary disease in which there is predisposition to benign and malignant tumours of many organ systems. A locus for this condition was recently assigned to chromosome 19p. We have identified truncating germline mutations in a gene residing on chromosome 19p in multiple individuals affected by PJS. This previously identified but unmapped gene, LKB1, has strong homology to a cytoplasmic Xenopus serine/threonine protein kinase XEEK1, and weaker similarity to many other protein kinases. Peutz-Jeghers syndrome is therefore the first cancer-susceptibility syndrome to be identified that is due to inactivating mutations in a protein kinase.


Subject(s)
Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , Xenopus Proteins , AMP-Activated Protein Kinase Kinases , Amino Acid Sequence , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 19 , Female , Germ-Line Mutation , Humans , Male , Molecular Sequence Data , Pedigree , Peutz-Jeghers Syndrome/enzymology , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL