Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Eur J Cancer ; 37(17): 2264-74, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11677117

ABSTRACT

Axl is a receptor tyrosine kinase (RTK) with oncogenic potential and transforming activity. Since Axl bears structural similarities to cell adhesion molecules such as neural cell adhesion molecule (NCAM) (FNIII domains), it is thought that Axl might play a role in adhesion. In this study, we have analysed the expression of the Axl protein and its ligand, Gas6, in human lung cancer cell lines of different histological origin. Axl expression occurred in approximately 60% of non-small cell lung cancer (NSCLC) cell lines, which grow adherently, and in normal bronchial epithelial cells (NHBE), but not in cell lines of small cell lung cancer origin (SCLC), which grow in suspension. A number of SCLC sublines, which could be selected spontaneously or after oncogene transfection for adherent growth, all expressed Axl protein. Overexpression of Axl per se, however, did not induce any change in the adhesion phenotype. All Axl-expressing cell lines demonstrated a membrane-bound 140 kD form, as well as a soluble 85 kD form, detectable in supernatant, of Axl-RTK. Expression of the Axl ligand Gas6 was detected in approximately 80% of all cell lines investigated. We conclude from these data that loss of Axl expression is a feature of SCLC tumour cells. Axl expression appears to be a consequence of cellular adhesion and possibly influences differentiation in human lung cancers.


Subject(s)
Intercellular Signaling Peptides and Proteins , Lung Neoplasms/enzymology , Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/enzymology , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Adhesion/physiology , Cell Division/physiology , Gene Expression , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Protein Processing, Post-Translational , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Axl Receptor Tyrosine Kinase
2.
Cancer ; 92(5): 1231-6, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11571737

ABSTRACT

BACKGROUND: Specific CD44 isoforms are cell surface adhesion molecules and have been shown to be associated with tumor progression and metastasis. In lung carcinoma, CD44 expression has been reported to be a feature of nonsmall cell lung carcinoma (NSCLC) but not small cell lung carcinoma. A specific variant, CD44v6, was shown to be expressed only in a subset of NSCLC, namely the squamous cell and bronchoalveolar carcinomas, suggesting that CD44 may play a role in lung carcinoma differentiation. METHODS: To determine whether differential CD44 expression is an early event in the pathogenesis of lung carcinoma, the authors investigated the pattern of expression of the standard (CD44s) and variant (CD44v6) isoforms by immunohistochemistry in normal lung, nonneoplastic specimens, and bronchial biopsies of preneoplastic lesions. RESULTS: In normal bronchial epithelium and all nonneoplastic cases, CD44s expression was limited to the basement membrane and adjacent lower strata of the epithelium, whereas CD44v6 was expressed within the basement membrane only. However, aberrant expression of both CD44s and CD44v6 was observed in all preneoplastic lesions examined. In cases of dysplasia, squamous metaplasia, goblet cell hyperplasia, and basal cell hyperplasia, all epithelial strata showed immunoreactivity for both isoforms, in contrast to normal epithelium, in which immunoreactivity was noted to be restricted to the basal layer cells. In contrast, CD44s and CD44v6 expression was completely absent in nearly all cases of adenomatosis. CONCLUSIONS: Altered CD44s and/or CD44v6 expression appears to be a feature of all preneoplastic lesions in the lung, the precise nature of which varies according to histologic tumor type. Therefore, the authors conclude that CD44s and CD44v6 may lend themselves to be markers of preneoplastic changes in the lung.


Subject(s)
Hyaluronan Receptors/metabolism , Lung Neoplasms/metabolism , Precancerous Conditions/metabolism , Respiratory Mucosa/metabolism , Glycoproteins/metabolism , Humans , Immunohistochemistry , Lung/metabolism , Lung/pathology , Lung Neoplasms/pathology , Precancerous Conditions/pathology , Respiratory Mucosa/pathology
3.
Cancer Res ; 61(15): 5905-10, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11479232

ABSTRACT

Functional inactivation of the Rb and p53 pathways appears to be a rite of passage for all cancerous cells. However, p53 and Rb alterations are rare events in neuroendocrine gastroenteropancreatic (GEP) tumors. The CDKN2 locus on chromosome 9p21 sits at the nexus of both pathways harboring tumor suppressor genes, which restrain cell growth by affecting the function of pRb and p53. Therefore, we analyzed the implication of their inactivation in 37 primary neuroendocrine GEP tumors and two cell culture models. RT-PCR analysis revealed loss of expression of at least one of the tumor suppressor genes CDKN2A/p16, CDKN2B/p15, and CDKN2D/p14 with distinct genetic profiles, most frequently in nonfunctional pancreatic tumors (57%) and small intestinal carcinoids (44%), and less commonly in insulinomas (30%) and gastrinomas (22%). DNA analysis and methylation-specific PCR attributed loss of expression to either homozygous deletion or 5'CpG island hypermethylation. 5-Aza-2-deoxycytidine treatment reversed CDKN2A/p16 and CDKN2B/p15 silencing with concurrent growth restraint. Thus, tumor suppressor genes localized in the 9p21 gene cluster are specific targets of inactivation in neuroendocrine GEP tumors, and demethylating agents might hold promise for selective therapy.


Subject(s)
Cell Cycle Proteins , Chromosomes, Human, Pair 9/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Gastrointestinal Neoplasms/genetics , Genes, Tumor Suppressor/genetics , Multigene Family/genetics , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Tumor Suppressor Proteins , Carrier Proteins/genetics , Cell Division/genetics , Chromosome Aberrations , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p19 , DNA Methylation , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, p16/genetics , Humans , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
4.
Surgery ; 129(4): 490-7, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283541

ABSTRACT

BACKGROUND: p16(INK4a) alterations are considered to be an early event in pancreatic tumorigenesis and have been described in duct lesions adjacent to pancreatic cancers. This study evaluates whether duct lesions in chronic pancreatitis tissues of patients without pancreatic cancer also harbor genetic alterations in the p16(INK4a) tumor-suppressor gene, and thus represent high-risk precursors for pancreatic cancer. METHODS: Tissues were obtained from 20 pancreatic specimens taken from patients operated on for histologically verified chronic pancreatitis. Pancreatic intraductal neoplasias (PanIN) were identified in hematoxylin-and-eosin-stained slides. p16 protein expression was investigated immunohistochemically in all specimens. DNA from PanIN and non-PanIN tissue was analyzed genetically for p16(INK4a) mutations by single-strand conformation variation analysis and direct sequencing of the encoding region. Additionally, p16(INK4a) promoter methylation was analyzed by a methylation specific polymerase test. RESULTS: PanIN-1a lesions were identified in 10 of the 20 chronic pancreatitis specimens. Four of these 10 PanIN specimens (40%), but none of the 20 non-PanIN tissues, revealed a loss of p16 expression in immunohistochemistry. The mutational analysis of the p16(INK4a) gene showed 1 known polymorphism (c.442G > A; A148T) but no mutations. Two of the 10 specimens with PanIN revealed an inactivating hypermethylation of the p16(INK4a) promoter. CONCLUSIONS: This study shows for the first time that p16(INK4a) alterations can be observed in a considerable number of PanIN1 in chronic pancreatitis tissues not associated with pancreatic cancer. Therefore, p16(INK4a) alterations, especially promoter methylation, might indicate high-risk precursors in chronic pancreatitis that might progress to cancer.


Subject(s)
Genes, p16 , Mutation , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/genetics , Pancreatitis/complications , Pancreatitis/genetics , Adult , Aged , Base Sequence , Chronic Disease , DNA Methylation , DNA Primers/genetics , Female , Humans , Male , Middle Aged , Pancreatitis/pathology , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic , Risk Factors
5.
J Cancer Res Clin Oncol ; 127(2): 139-41, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11216916

ABSTRACT

Small cell lung cancer (SCLC) is a very malignant tumor known to grow aggressively and to metastasize early. It is well established that metastasis generally involves both tumor cell adhesion and proteolytic degradation of the extracellular matrix. However, SCLC cells cultured in vitro, such as the classic SCLC cell line NCI-H69, grow in floating aggregates and express only negligible proteolytic activity. In this report, we show that NCI-H69 cells can be selected for adherent growth. In contrast to parental suspension cells, the adherent cells were found to express tissue factor as well as gelatinolytic activity, attributable to matrix metalloproteinases 2 and 9. Such a switch of tumor cell characteristics, if it could occur in SCLC patients, might add to the understanding of the steps involved in the spreading of this highly metastatic type of lung cancer.


Subject(s)
Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Matrix Metalloproteinases/metabolism , Thromboplastin/biosynthesis , Carcinoma, Small Cell/enzymology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Tumor Cells, Cultured
6.
Pancreas ; 21(4): 369-75, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11075991

ABSTRACT

Multiple primary tumors in pancreatic cancer patients might indicate a genetic predisposition to the development of malignancies. In this study we evaluated whether the mutation rate of the TP53 and p16INK4a genes of pancreatic cancers differs in pancreatic cancer patients with and without multiple primaries. Furthermore, we investigated whether pancreatic cancer patients with multiple primaries carry germline mutations in either p16INK4a, TP53, or BRCA2 tumor suppressor genes to detect a genetic alteration that predisposes to the development of different primaries. Fourteen (23%) of 60 pancreatic cancer patients developed histologically verified additional primaries during their lifetimes. Normal constitutional and tumor DNA of the 14 patients with a positive cancer history, but negative family history, were analyzed for p16INK4a, TP53, and BRCA2 mutations by single-strand conformational variant (SSCV) analysis and direct sequencing. Hypermethylation of the p16INK4a promoter region in pancreatic cancers was identified by methylation-specific polymerase chain reaction (PCR; MSP). Four of 14 pancreatic carcinomas carried somatic intragenic p16INK4a mutations, and another four tumors revealed hypermethylation of the p16INK4a promoter region. Somatic intragenic TP53 mutations were identified in six of 14 tumors. None of the pancreatic cancer patients carried TP53 or BRCA2 germline mutations. In contrast, one of 14 pancreatic cancer patients with multiple primaries carried the p16INK4a mutation A68V in his germline. This mutation was localized in the conserved second ankyrin repeat of p16INK4a and did not occur in 100 control patients. The frequency of somatic TP53 and p16INK4a mutations in pancreatic cancer is similar in patients with and without multiple primaries. TP53 and BRCA2 germline mutations seem not to be significantly associated with the occurrence of multiple primaries in pancreatic cancer patients. However, p16INK4a germline mutations might be causative for tumor development in some pancreatic cancer patients with multiple primaries. The genetic investigation of patients with accumulation of different cancers even without a positive family history may be a new approach for the understanding of the relation of different cancers.


Subject(s)
Genes, p16 , Germ-Line Mutation , Neoplasms, Multiple Primary/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , BRCA2 Protein , DNA Methylation , Genes, p53 , Humans , Middle Aged , Neoplasm Proteins/genetics , Transcription Factors/genetics
7.
Digestion ; 62(2-3): 171-7, 2000.
Article in English | MEDLINE | ID: mdl-11025365

ABSTRACT

BACKGROUND/AIMS: The molecular mechanisms contributing to the tumorigenesis of insulinomas are poorly understood. Disruption of the cell cycle due to inactivation of the p16(INK4a) tumor-suppressor gene was identified in a variety of human tumors, including gastrinomas and nonfunctioning endocrine pancreatic carcinomas. In this study the role of p16(INK4a) in the tumorigenesis of insulinomas was evaluated. METHODS: Seventeen insulinomas (14 benign, 3 malignant) were analyzed for genetic alterations in the p16(INK4a) tumor-suppressor gene by SSCP, PCR-based deletion and methylation-specific assays. p16 expression was determined by immunohistochemistry. RESULTS: One malignant insulinoma showed a homozygous deletion of p16(INK4a) and another two benign insulinomas revealed aberrant methylation of the p16(INK4a) promoter region. All three tumors lacked p16 expression according to immunohistochemistry. None of the insulinomas carried intragenic p16(INK4a) mutations. In total, 17% of insulinomas had p16(INK4a) alterations. CONCLUSIONS: The p16(INK4a) tumor-suppressor gene contributes to tumorigenesis in only a small subset of insulinomas.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , Genes, p16/genetics , Insulinoma/genetics , Pancreatic Neoplasms/genetics , Cell Transformation, Neoplastic , Chromosome Deletion , DNA, Neoplasm/genetics , Humans , Immunohistochemistry , Polymerase Chain Reaction , Promoter Regions, Genetic
8.
J Clin Oncol ; 18(18): 3221-9, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10986054

ABSTRACT

PURPOSE: The aim of this study was to investigate the frequency of three (epi)genetic alterations (p53 and K-ras mutations and p16(INK4a) promoter hypermethylation) in symptomatic chronic smokers compared with patients with lung cancer and to evaluate the use of exfoliative material for such analyses. PATIENTS AND METHODS: Fifty-one patients with histologically confirmed lung cancer and 25 chronic smokers (> 20 pack-years) were investigated for mutations in the K-ras (codon 12) and p53 (codons 248, 249, and 273) genes and for allelic hypermethylation of the p16(INK4a) gene. DNA was isolated from sputum and bilateral bronchial lavage, and brushings were taken at bronchoscopy. RESULTS: Forty-one genetic lesions were detected within exfoliative material from the group of 51 patients with lung cancer and 10 lesions in the chronic smoker group. K-ras mutations occurred exclusively in the lung cancer group, whereas p53 mutations and p16(INK4a) promoter hypermethylation were also found in chronic smokers. Three of eight chronic smokers who harbored an (epi)genetic alteration were subsequently diagnosed with lung cancer. Analysis of sputum yielded information equivalent to that of samples obtained during bronchoscopy. CONCLUSION: p16(INK4a) promoter hypermethylation and p53 mutations can occur in chronic smokers before any clinical evidence of neoplasia and may be indicative of an increased risk of developing lung cancer or of early disease. K-ras mutations occur exclusively in the presence of clinically detectable neoplastic transformation. Molecular analysis of sputum for such markers may provide an effective means of screening chronic smokers to enable earlier detection and therapeutic intervention of lung cancer.


Subject(s)
Carrier Proteins/genetics , DNA Methylation , Genes, p53/genetics , Genes, ras/genetics , Lung Neoplasms/genetics , Mutation , Smoking/genetics , Aged , Aged, 80 and over , Biopsy , Bronchoalveolar Lavage Fluid/chemistry , Bronchoscopy , CpG Islands/physiology , Cyclin-Dependent Kinase Inhibitor p16 , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Female , Genetic Markers/genetics , Humans , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Male , Middle Aged , Polymerase Chain Reaction/methods , Precancerous Conditions/etiology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Promoter Regions, Genetic/physiology , Smoking/adverse effects , Smoking/pathology , Sputum/chemistry
9.
J Pathol ; 190(1): 69-75, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10640994

ABSTRACT

An 'enriched polymerase chain reaction (PCR)' protocol has been established for the sensitive detection of oncogene mutations in body fluid samples from cancer patients. This two-step protocol combines an allele-specific PCR clamping step followed by a PCR-RFLP (restriction fragment length polymorphism) confirmatory step. The method thus resembles a nested PCR technique starting directly from genomic DNA material and, in no more than 54 PCR cycles, allows the sensitive detection of one mutant allele in 10(3) normal alleles. This protocol was tested on bronchial cytology samples and sputum taken from lung cancer patients and point mutations could be detected both in codon 12 of K-ras and in three codons (248, 249, and 273) of the p53 gene. Comparing this protocol with a different 'enriched PCR' method based on repetitive PCR-RFLP steps, a high concordance was noted between the two methods. Although the present protocol seems to be less sensitive by approximately one order of magnitude, it is much easier to perform and thus could be applied to the rapid but sensitive detection of allelic subfractions in a population of cells derived from exfoliative material.


Subject(s)
DNA Mutational Analysis/methods , Genes, p53/genetics , Genes, ras/genetics , Lung Neoplasms/genetics , Polymerase Chain Reaction/methods , Bronchoalveolar Lavage Fluid , Cell Line , Feasibility Studies , Humans , Mutation, Missense , Polymorphism, Restriction Fragment Length , Sputum
10.
Cancer ; 86(1): 43-9, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10391562

ABSTRACT

BACKGROUND: Protein S is a plasma protein that serves as an important cofactor for activated protein C in the blood anticoagulation system. Protein S also acts as a mitogen on distinct cell types and is a ligand for Tyro3, a member of the Axl family of oncogenic receptor tyrosine kinases. This lends support to the hypothesis that protein S might also be involved in tumor cell regulation. METHODS: The expression of protein S and receptor Tyro3 was examined in 22 lung carcinoma cell lines and normal bronchial epithelial cells by reverse transcriptase-polymerase chain reaction. Secreted protein S was identified by Western blot analysis of cell supernatants and tested in a protein S-dependent clotting test for anticoagulant activity. Immunohistochemistry with anti-protein S polyvalent antiserum was also performed on 31 primary lung carcinoma specimens. RESULTS: Protein S mRNA and secreted protein were found in 11 of 12 cell lines of nonsmall cell lung carcinoma (NSCLC) origin and in normal bronchial epithelial cells, but they were found in only 4 of 10 small cell lung carcinoma (SCLC) cell lines. The majority of lung carcinoma cell lines that expressed protein S (13 of 15) also revealed expression of the cognate receptor, Tyro3. Protein S that was present in cell supernatant had anticoagulant activity comparable to that of plasma protein S, suggesting that it is gamma-carboxylated. In lung tumor tissue, protein S antigen was found in 20 of 31 cases examined, predominantly in tumors of the squamous cell and bronchioalveolar cell types. Protein S was found not only in tumor cells but also in cells of the normal bronchial epithelium, in alveolar macrophages, and in endothelium. CONCLUSIONS: To the authors' knowledge, their report is the first of the synthesis of an active anticoagulant protein in epithelial cells of human cancer. It suggests that protein S, by binding to a receptor (Tyro3), may influence local anticoagulation events or other, as yet unidentified, aspects of lung tumor development.


Subject(s)
Antigens, Neoplasm/analysis , Carcinoma, Non-Small-Cell Lung/physiopathology , Carcinoma, Small Cell/physiopathology , Lung Neoplasms/physiopathology , Protein S/biosynthesis , Receptor Protein-Tyrosine Kinases/metabolism , Base Sequence , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Small Cell/genetics , Humans , Lung Neoplasms/genetics , Molecular Sequence Data , Protein S/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Int J Cancer ; 80(6): 930-4, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10074928

ABSTRACT

A puzzling finding in various human tumors, including glioblastoma multiforme (GBM), is the stabilization of wild-type (wt) p53 protein. The biological significance of this phenomenon and the mechanism by which it occurs are unexplained. Recent reports have revealed that mdm2 exerts its negative regulation on the p53 signal by directly binding p53 protein and thereby instigating its proteasomal degradation. mdm2 has been shown to exist in alternatively spliced forms in human ovarian and bladder carcinomas, and recently in GBM, with loss or disruption of its p53 binding domain. Here we report that alternatively spliced transcripts of mdm2 are present in 7 of 16 human GBM primary cell cultures and in the established GBM cell lines LN 229 and LN 18. Sequencing demonstrated loss of the amino terminal p53 binding domain in these alternatively spliced mdm2 transcripts, and an out-of-frame splicing in the majority of cases. A significant correlation between the presence of mdm2 splice variants and increased expression of wt p53 protein was observed. Furthermore, in the presence of an mdm2 splice variant, wt p53 stabilization occurred despite coincident MDM2 amplification. Our findings suggest that wt p53 protein stabilization may arise as a consequence of alternative splicing of mdm2. Such a mechanism might account for wt p53 protein accumulation in GBM cells, even in the presence of MDM2 gene amplification.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplasm Proteins/metabolism , Nuclear Proteins , Protein Isoforms/metabolism , Proto-Oncogene Proteins/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Tumor Suppressor Protein p53/metabolism , Binding Sites , Brain Neoplasms/metabolism , Cysteine Endopeptidases/metabolism , Glioblastoma/metabolism , Half-Life , Humans , Multienzyme Complexes/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Proteasome Endopeptidase Complex , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Substrate Specificity , Tumor Cells, Cultured
12.
Pathobiology ; 67(4): 202-6, 1999.
Article in English | MEDLINE | ID: mdl-10738182

ABSTRACT

The present study investigates nitrosourea-induced rat (Rattus norvegicus) glioma cell lines for the functional status of the p16/Cdkn2a/Ink4a gene, which encodes the p16 cdk4 inhibitor and the alternative reading frame protein, p19ARF. We detected homozygous deletions of the p16/Cdkn2a/Ink4a gene locus in 4 of 5 glioma cell lines (C6, F98, RG2, and RGL.3), but not in the 9L gliosarcoma cell line or in a rat primary fibroblast cell line. RT-PCR demonstrated expression of the p16 and p19ARF mRNAs only in 9L cells and in rat fibroblasts. Comparative genomic in situ hybridization showed that the copy number of rat chromosome RNO5 was not altered in any of the glioma cell lines investigated, indicating that the deletions result from a discrete loss in the region of the p16/Cdkn2a/Ink4a locus. This is the first report of p16/Cdkn2a/Ink4a deletions present in nitrosourea-induced rat glioma cell lines. Since this genetic alteration is also commonly observed in human malignant glial tumors, our results validate the use of chemically induced rat glioma cell lines as an experimental model in the development of gene therapy strategies.


Subject(s)
Gene Deletion , Genes, p16/genetics , Glioma/genetics , Neoplasms, Experimental/genetics , Nervous System Neoplasms/genetics , Nitrosourea Compounds/toxicity , RNA, Neoplasm/analysis , Animals , Cell Division , Chromosomes/genetics , DNA Primers/chemistry , Fibroblasts/pathology , Glioma/chemically induced , Glioma/pathology , Homozygote , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Nervous System Neoplasms/chemically induced , Nervous System Neoplasms/pathology , Rats , Rats, Inbred F344/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
13.
Oncogene ; 16(23): 3057-68, 1998 Jun 11.
Article in English | MEDLINE | ID: mdl-9662339

ABSTRACT

Transitions from small cell (SCLC) to non-small cell lung cancer (NSCLC) cells have been documented both in vitro and in vivo and are thought to be an important step during tumor progression of human small cell lung cancer towards a treatment-resistant tumor state. We have screened NSCLC and SCLC cell lines for differences in the composition of nuclear transcription factors using consensus oligonucleotide sequences (SRE, Ets, TRE, CRE, B-motif, GAS, E-box). We found NSCLC cells to exhibit significantly higher AP-1 binding activity than SCLC cells consistent with the increased expression of CD44, an AP-1 target gene. To gain more insight into the molecular mechanisms underlying these differences, we analysed SCLC cell lines (NCI-N592 and NCI-H69) which were phenotypically transformed into NSCLC-type cells by transfection with activated H-ras and c-myc oncogenes. In these cells, ras-induced transition is accompanied by a strong induction of AP-1-binding activity along with increased expression of CD44 mRNA and protein. When analysing the composition of the AP-1 complex in more detail and comparing ras-induced versus phorbol ester-induced changes, we found Fra-1 to be the major component induced in ras-transfected but not in phorbol-ester treated or non-treated parental SCLC cells. This finding is paralleled by the observation that among the various members of the Fos and Jun family analysed (c-Fos, FosB, Fra-1, Fra-2, c-Jun, JunD, JunB) fra-1 is the only gene to be exclusively expressed in NSCLC cells but not in cells of SCLC origin. Our data, thus, point to a histiotype-related mechanism of recruitment among AP-1 proteins which may have bearings on the fate of lung cancer development.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Small Cell/genetics , Consensus Sequence , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-fos/genetics , Transcription Factor AP-1/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/pathology , DNA, Neoplasm/metabolism , Genes, ras , Humans , Lung Neoplasms/pathology , Phenotype , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Transcription, Genetic , Transformation, Genetic , Tumor Cells, Cultured
14.
Thromb Haemost ; 79(4): 773-7, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9569191

ABSTRACT

Resistance to activated protein C (APC resistance) is the most common cause of thrombophilia and linked to a single point mutation in the factor V gene (G-->A transition at nucleotide 1691). In the past, several PCR based methods have been proposed to determine the allelostatus of individual patients from small amounts of blood DNA including PCR followed by restriction fragment length polymorphism detection (PCR-RFLP), PCR using sequence-specific primers (PCR-SSP) and oligonucleotide ligation assay (OLA). Here, we present a novel approach based on the method of peptide nucleic acid(PNA)-mediated PCR clamping which is extremely sensitive to base pair mismatches. If PNAs specific for the two allelic variants are applied separately in each case a clear discrimination between a heterozygous or homozygous normal or homozygous Factor V Leiden status is possible and no further confirmation step is required. In a prospective study, 60 patients with suspected venous thrombosis events were tested and compared to the conventional PCR-RFLP technique. The concordance between both methods was 100%. PNA-based factor V genotyping, therefore, should be considered for large scale screening of those patients considered to be at risk for deep venous thrombosis.


Subject(s)
DNA Mutational Analysis/methods , Factor V/genetics , Oligopeptides/pharmacology , Polymerase Chain Reaction/methods , Thrombophilia/genetics , Alleles , DNA Primers , DNA Replication/drug effects , Factor V/analysis , Genotype , Humans , Magnesium/pharmacology , Nucleic Acid Denaturation , Nucleic Acid Synthesis Inhibitors/pharmacology , Polymorphism, Restriction Fragment Length , Protein C/metabolism , Substrate Specificity , Temperature , Thrombophilia/etiology
15.
Clin Cancer Res ; 4(2): 361-71, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9516924

ABSTRACT

RFLP-mediated PCR has been successfully applied as a reliable tool in the detection of ras mutations in many cancers and provides a basis for "mutant-enriched PCR" protocols. We have, therefore, modified this technique to the sensitive detection of K-ras codon 12 and also p53 "hot spot" mutations, which, frequently in lung cancer, affect codons at the positions 157, 175, 245, 248, 249, and 273. With a high sensitivity of 1 mutant allele in 10(4) normal alleles, our enrichment assay allows the detection of oncogene alleles when only a few tumor cells are present within a normal cell population. Brush cytology material obtained from the tumor site of 20 patients with endoscopically apparent bronchial carcinoma was compared to macroscopically normal mucosa taken from the contralateral bronchus ("control" cytology). We found K-ras codon 12 mutations in 5 cases (25%) and p53 mutations in 13 cases (65%) in the tumor-derived cell material but, with the exception of two cases, not in cell material taken from the control cytology. Seventy-five % of the samples analyzed showed that at least one of the two oncogenes was affected. In several cases, two p53 lesions were detected concomitantly. The majority of the mutations could be reconfirmed by an alternative approach exploiting changes in the genomic RFLP pattern induced by these mutations and were also demonstrated in separate diagnostic biopsies taken. Thus, we conclude that the established enriched PCR protocol ensures a high sensitivity and preserved specificity for the diagnosis of oncogene lesions associated with lung cancer. Because conventional techniques normally yield a lower incidence of corresponding ras and p53 mutations, we think that both the high rate and the heterogeneity of p53 mutations found in some cases are, indeed, related to the increased sensitivity of this new enriched PCR technique.


Subject(s)
Genes, p53 , Genes, ras , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Polymerase Chain Reaction/methods , Aged , Aged, 80 and over , Alleles , Biopsy , Codon , Cytological Techniques , DNA Primers , Exons , Female , Humans , Male , Middle Aged , Polymorphism, Restriction Fragment Length , Sensitivity and Specificity , Tumor Cells, Cultured
16.
Nucleic Acids Res ; 26(5): 1356-8, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9469850

ABSTRACT

For the rapid and sensitive detection of p53 'hot spot' mutations, we combined polymerase chain reaction based single-strand conformational polymorphism (PCR-SSCP) analysis with sequence specific-clamping by peptide nucleic acids (PNAs) in a one-step reaction tube protocol. For this purpose, we designed two PNA molecules comprising aa 246-250 of exon 7 and aa 270-275 of exon 8, respectively, to suppress the amplification of wild-type p53 allelic variants during PCR amplification. Using this method in a survey of 20 brush cytology samples from lung cancer patients, we were able to detect five p53 point mutations occurring in codons 248, 249 and 273 which could not be retrieved by conventional PCR-SSCP. Thus, allelic suppression by PNA molecules opens a way to largely improve the sensitivity of existing PCR-SSCP protocols (approximately 10-50-fold) and could be useful in the detection of 'hot spot' oncogene lesions in histological samples containing only a small number of cancer cells.


Subject(s)
Genes, p53 , Mutation , Polymerase Chain Reaction/methods , Polymorphism, Single-Stranded Conformational , Alleles , Base Sequence , DNA Primers/genetics , DNA, Neoplasm/genetics , Exons , Humans , Lung Neoplasms/genetics , Nucleic Acids , Peptides , Point Mutation , Polymerase Chain Reaction/statistics & numerical data , Sensitivity and Specificity
17.
Int J Cancer ; 71(2): 275-83, 1997 Apr 10.
Article in English | MEDLINE | ID: mdl-9139854

ABSTRACT

Activation of protein kinase C- (PKC) and Fos/Jun-dependent signal transduction pathways are thought to be major effects of oncogene action in different tumor systems including human non-small-cell lung carcinoma (NSCLC). We have previously shown that the phorbol ester analogue phorbol-myristate-acetate (PMA), which is a potent activator of PKC, can induce squamous-type cellular differentiation and the expression of proteinases, such as plasminogen activators and pro-cathepsin L, in several NSCLC cell lines. To investigate the PMA-dependent effect on proteinase secretion in more detail, we have now analysed the role of a downstream transmitter of PKC activity in this process, namely Fos, which is part of the AP-1 transcription factor in the nucleus. We transfected a cell line derived from an undifferentiated squamous-cell lung carcinoma with different chimeric fos-estrogen receptor constructs (fos-ER) which makes selective activation of this transcription factor possible. The resulting clones were treated either with PMA as activator of PKC, or with diethylstilbestrol (DES), an estrogen analogue binding to and thereby activating preformed Fos-ER molecules. We show that cells treated with either substance undergo similar phenotypic changes (change from cuboidal to spindle-cell type) and decrease their doubling rates and cloning efficiencies. This is paralleled by the induction of several proteinase genes such as t-PA, urokinase, and pro-cathepsins B and L. Contrary to activated PKC, Fos in this system seems to be unable to initiate terminal squamous-cell differentiation, as assessed by the production of cornified envelopes. It is, however, efficient in the stimulation of neutral or lysosomal proteinase secretion as determined by Western-blot analysis and zymography. This Fos-ER expressing system thus seems to be a valuable tool in the molecular dissection of pathways that lead to the activation and secretion of proteinases in NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Endopeptidases/metabolism , Gene Expression Regulation, Enzymologic , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Size/drug effects , DNA Primers/chemistry , Diethylstilbestrol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Immunoblotting , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Protein Kinase C/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Signal Transduction , Transfection , Tumor Cells, Cultured
18.
Lung Cancer ; 16(2-3): 151-72, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9152947

ABSTRACT

The CD44 transmembrane glycoprotein is expressed in most adult tissues and in the majority of neoplasias. Due to alternative splicing, this cell adhesion molecule exists in multiple isoforms some of which have been associated with specific types of tumours as well as with increased tumour metastasis. In this study, we have looked at the level and type of CD44 expression in lung cancer which represents a histologically heterogenous form of cancer composed of small cell lung carcinoma (SCLC) and non-small cell lung carcinoma (NSCLC), the latter subgroup comprising adenocarcinoma (ADC), bronchio-alveolar carcinoma (BAC), large cell carcinoma (LCC), and squamous cell carcinoma (SCC). We analysed 20 lung cancer cell lines and 64 primary tumours by RT-PCR and immunohistochemical detection of the CD44 standard and variant protein isoforms. Our results suggest that (i) CD44 is expressed in all histologically distinct subsets of lung cancer with a tendency SCC > BAC > ADC > LCC > SCLC, (ii) expression of the CD44 isoforms v5, v7, v8, and, most notably that of CD44 exon v6, strongly correlates with tumours of squamous cell and bronchio-alveolar carcinoma origin, tumours which commonly exhibit a comparatively low metastasizing potential, and (iii) the expression of CD44 isoforms is independent from the tumour size and lymph node status at surgery, the proliferative status of the tumour cell population (Ki67 antigen expression) and the histopathological grading (G1 to G3). Only non-differentiated tumours (G4), which were restricted to SCLC and LCC samples revealed markedly reduced CD44 standard and isoform antigen. In conclusion, our data point to a clear histiotype-related pattern of CD44 variant expression preferentially that of CD44v6 in SCC and BAC.


Subject(s)
Antigens, CD/biosynthesis , Hyaluronan Receptors/biosynthesis , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma, Bronchiolo-Alveolar/immunology , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Adult , Alternative Splicing , Antigens, CD/analysis , Carcinoma, Large Cell/immunology , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/immunology , Carcinoma, Small Cell/pathology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Division , Cell Line , DNA Primers , Exons , Humans , Hyaluronan Receptors/analysis , Immunohistochemistry , Ki-67 Antigen/analysis , Lung Neoplasms/classification , Neoplasm Staging , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Transcription, Genetic , Tumor Cells, Cultured
19.
Ann Hematol ; 74(1): 49-50, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9031617

ABSTRACT

Patients with polycythemia vera (PV) have a high risk of thrombosis. However, thrombosis is not sufficiently predictable with standard diagnostic procedures. We report on a patient with PV and recurrent thrombosis who nevertheless had a low platelet count while under therapy with hydroxyurea. As a result of duodenal ulcer and gastrointestinal bleeding, treatment with phenprocoumon was stopped years ago. Recently, heterozygosity for the factor V gene defect was diagnosed and anticoagulation therapy was reconsidered. In conclusion, the presence of resistance to activated protein C was an additional thrombotic risk factor that was important for our decision to change the treatment strategy in our patient.


Subject(s)
Polycythemia Vera/complications , Protein C/metabolism , Protein C/pharmacology , Thrombosis/complications , Thrombosis/epidemiology , Drug Resistance , Enzyme Activation , Factor V/genetics , Hematopoiesis/drug effects , Heterozygote , Humans , Hydroxyurea/therapeutic use , Male , Middle Aged , Mutation , Platelet Count/drug effects , Risk Factors
20.
J Biol Chem ; 269(9): 6866-73, 1994 Mar 04.
Article in English | MEDLINE | ID: mdl-8120048

ABSTRACT

Stimulation of quiescent cells with growth factors triggers changes in gene expression through multiple signal transduction pathways. One of these changes in Swiss 3T3 cells is the strong accumulation of T1 mRNA which encodes a secreted glycoprotein of the immunoglobulin superfamily. Proliferating cells continued to express T1 mRNA at a lower level, whereas growth arrest induced either by serum deprivation or by contact inhibition was paralleled by the disappearance of the T1 mRNA. T1 mRNA synthesis in response to serum and platelet-derived growth factor stimulation is mediated through protein kinase C-dependent and protein kinase C-independent pathways. Activation of protein kinase A also led to T1 gene expression. Ongoing protein synthesis is a prerequisite for T1 gene induction by growth factors which defines T1 as a delayed early serum-responsive gene. The ability of the immediate early transcription factors c-Fos and FosB to directly induce the T1 gene was demonstrated in a conditional expression system in the absence of protein synthesis. Furthermore, all known inducers of the T1 gene also lead to c-fos gene activation. Thus we show that the T1 gene is regulated by signals which are transduced through multiple pathways and provide evidence that the Fos proteins play an important role in the integration of these pathways.


Subject(s)
Gene Expression Regulation/drug effects , Glycoproteins/biosynthesis , Growth Substances/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/physiology , 3T3 Cells , Animals , Becaplermin , Blotting, Northern , Blotting, Western , Cell Division/drug effects , Cell Division/physiology , Cell Line , Cycloheximide/pharmacology , Epidermal Growth Factor/pharmacology , Estradiol/pharmacology , Genes, fos , Glycoproteins/isolation & purification , Insulin/pharmacology , Mice , Mitosis/drug effects , Mitosis/physiology , Platelet-Derived Growth Factor/pharmacology , Promoter Regions, Genetic , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-sis , RNA, Messenger/biosynthesis , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation , Transferrin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...