Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Acta Ophthalmol ; 89(1): 47-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19558573

ABSTRACT

PURPOSE: We investigated the immunohistochemical characteristics of corneal specimens in congenital aniridia and pemphigoid using various corneal markers to determine the status of the corneal epithelium. METHODS: Conjunctivalization was clinically suspected in all corneas. Ten aniridia and seven pemphigoid paraffin-embedded corneal specimens were stained with periodic Schiff reagent (PAS) and antibodies against CK3/12, CK12, CK19, breast cancer resistance protein 1 (BCRP) and p63. RESULTS: Aniridia: six cases contained goblet cells, four were negative. Both groups had cases with (three of six; one of four) and without CK19 positivity and cases with (two of six; three of four) and without p63 positivity. All aniridia cases except two in the goblet cell group were CK3/12- and CK12-positive and BCRP-negative. Pemphigoid: only one of the seven cases contained goblet cells. This case stained positively for CK19, 3/12, 12 and p63 and negatively for BCRP. The other six cases were positive for CK3/12, five of which were positive for CK12; only one case was CK19-positive. Three cases were p63-positive and two BCRP-positive. The CK12 staining was heterogenous in most cases and was often found in the superficial layer. CONCLUSION: Three different stages of epithelial characteristics were found in congenital aniridia and pemphigoid: (i) CK19-negative and inhomogenous CK12-positive cases indicating epithelium mainly from (partly) CK12-deficient limbal stem cells; (ii) CK19- and/or goblet cell-positive and CK12-positive cases with their epithelia originating from CK12-deficient limbal stem cells and from incursing conjunctival cells; and (iii) CK19-positive and CK12-negative cases consisting of conjunctival cells alone.


Subject(s)
Aniridia/metabolism , Biomarkers/metabolism , Epithelium, Corneal/metabolism , Pemphigoid, Benign Mucous Membrane/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Epithelial Cells/metabolism , Female , Goblet Cells/metabolism , Humans , Immunoenzyme Techniques , Keratin-12/metabolism , Keratin-19/metabolism , Keratin-3/metabolism , Male , Membrane Proteins/metabolism , Middle Aged , Young Adult
2.
Front Biosci (Elite Ed) ; 2(3): 1042-53, 2010 06 01.
Article in English | MEDLINE | ID: mdl-20515774

ABSTRACT

Differential expression of p53 isoforms might participate in the marked resistance towards conventional chemotherapy of renal cell carcinomas (RCCs). Therefore, we analysed their differential expression and regulation in RCCs. RCCs expressed a more p53 activating isoform pattern during tumor initiation and progression, in vivo. In vitro, two cell lines exhibiting a similar sensitivity towards Topotecan-induced cell death revealed a similar induction of p53 target genes but strongly differed in their extent of apoptosis. Furthermore, they strongly differed in their basal expression patterns and differential regulation of the isoforms. In conclusion, our study examined for the first time the differential expression and regulation of all p53 isoforms in a tumor in vivo. Furthermore, novel results in our in vitro studies show that p53 isoforms are strongly differentially regulated by chemotherapy in RCCs and that expression and regulation of so-called "p53-target genes" are obviously at least in part regulated by other transcription factors. In addition, our original findings show that p53 isoform expression in RCC cell lines is of minor importance for sensitivity towards chemotherapy.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Blotting, Western , Carcinoma, Renal Cell/pathology , DNA Primers , Humans , Kidney Neoplasms/pathology , Polymerase Chain Reaction , Protein Isoforms/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics
3.
Cell Oncol ; 32(1-2): 29-42, 2010.
Article in English | MEDLINE | ID: mdl-20208132

ABSTRACT

BACKGROUND: The importance of caspase-2 activation for mediating apoptosis in cancer is not clear and seems to differ between different tumour types. Furthermore, only few data have been obtained concerning the expression of caspase-2, which can be alternatively spliced into caspase-2L and caspase-2S, and the other PIDDosome members PIDD and RAIDD in human tumours in vivo. We, therefore, investigated their expression in renal cell carcinomas (RCCs) of the clear cell type in vivo and analysed the role of caspase-2 in chemotherapy-induced apoptosis in RCCs in vitro. METHODS: The analyses were performed by semiquantitative real-time PCR, Western Blot and Caspase-2 Assay. RESULTS: Our in vivo results showed an overall decrease in proapoptotic caspase-2L expression during tumour progression due to an increase in the relative share of caspase-2S mRNA in total caspase-2 mRNA expression. Furthermore, an increase in the expression of PIDD and RAIDD could be observed. In contrast, antiapoptotic BCL-2 expression increased only during early tumour stages, whereas expression decreased in pT3 RCCs. In vitro, caspase-2 activation in RCC cell lines coincidenced with sensitivity of tumour cells towards Topotecan-induced apoptosis. However, inhibition of caspase-2 could not prevent Topotecan-induced apoptosis. Interestingly, Topotecan-resistance could be overcome by the apoptosis-sensitizing drug HA14-1. CONCLUSION: Our study confirms the concept of a shift towards a more antiapoptotic transcriptional context during tumour progression in RCCs. Furthermore, it shows that caspase-2 participates in chemotherapy-induced apoptosis in RCCs although it is not mandatory for it. Additionally, inhibition of antiapoptotic BCL-2 family members might provide a possible way to overcome chemotherapy resistance of RCCs.


Subject(s)
Apoptosis/drug effects , CRADD Signaling Adaptor Protein/genetics , Carcinoma, Renal Cell/genetics , Carrier Proteins/genetics , Caspase 2/metabolism , Kidney Neoplasms/genetics , Up-Regulation , CRADD Signaling Adaptor Protein/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/physiopathology , Carrier Proteins/metabolism , Caspase 2/genetics , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Kidney Neoplasms/physiopathology , Male , Topotecan/pharmacology , Topotecan/therapeutic use , Tumor Cells, Cultured
4.
Cell Oncol ; 30(5): 419-33, 2008.
Article in English | MEDLINE | ID: mdl-18791273

ABSTRACT

Renal cell carcinomas (RCCs) exhibit a marked resistance towards apoptosis. Although most apoptotic stimuli converge at the level of the mitochondria, little is known about the mitochondrial apoptosis pathway in renal cell carcinomas. The aim of the present study, therefore, was to investigate the functionality of the mitochondrial apoptosis pathway in renal cell carcinoma cell lines by exposure to TRAIL, etoposide, HA14-1 and betulinic acid activating the mitochondria by different mechanisms. Sensitivity to TRAIL-induced apoptosis correlated with cleavage of the initiator caspase-8, but the mitochondrial apoptosis pathway was not induced. Similarly, etoposide and betulinic acid could not induce mitochondrial damage. In contrast, HA14-1 was able to activate mitochondrial apoptosis, thereby demonstrating functionally inducible signalling pathways downstream of the mitochondria. The intactness of the pathways upstream of the mitochondria was shown by pretreatment of TRAIL-sensitive cell lines with HA14-1, which could reconstitute TRAIL-induced mitochondrial damage and resulted in a synergistic apoptosis induction. Our results demonstrate that the apoptotic pathways upstream and downstream of the mitochondria are intact and inducible in renal cell carcinoma cell lines. However, resistance towards mitochondrial apoptosis is located on the level of the mitochondria themselves.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Benzopyrans/pharmacology , Drug Resistance, Neoplasm/physiology , Mitochondria/drug effects , Nitriles/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspase 8/drug effects , Caspase 8/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Cell Line, Tumor , Etoposide/pharmacology , Humans , Pentacyclic Triterpenes , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Triterpenes/pharmacology , Betulinic Acid
5.
Apoptosis ; 13(7): 938-49, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18516683

ABSTRACT

Activation of the initiator-caspase, caspase-8 is under tight control of multiple antiapoptotic regulators including ARC, cFlip(S), cFlip(L) and PED/PEA-15. Since there is little data regarding the expression of caspase-8 and its antiapoptotic regulators in human tumours in vivo, we analysed their expression in renal cell carcinomas (RCCs) to identify which of these genes might be crucial for the well known impaired apoptosis and--as a result--resistance towards chemotherapy and ionizing radiation of RCCs. Caspase-8, cFlip(S), cFlip(L) and PED/PEA-15 mRNA expression was significantly increased only in early stages of RCCs compared to non-neoplastic renal tissue. In contrast, ARC mRNA expression was significantly increased in RCCs of all stages without differences between the tumour stages and grades. Importantly, the relative mRNA expression ratio between ARC and caspase-8 was significantly increased during carcinogenesis and tumour progression. In contrast, the relative mRNA expression ratio between cFlip(S), cFlip(L) or PED/PEA-15 and caspase-8 remained constant during all tumour stages. In conclusion, our analysis revealed that ARC is the only caspase-8 inhibiting regulator being constantly overexpressed in RCCs. Furthermore, the balance between antiapoptotic ARC and proapoptotic caspase-8 is the only one to be disturbed during carcinogenesis and tumour progression of RCCs. This inhibition of Caspase-8 might therefore be one example for the multiple antiapoptotic functions of ARC in RCCs possibly contributing to the marked resistance of RCCs towards radio- and chemotherapy and reflects a shift of gene expression towards a more antiapoptotic context in RCCs.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carcinoma, Renal Cell/genetics , Caspase 8/genetics , Caspase Inhibitors , Kidney Neoplasms/genetics , Muscle Proteins/genetics , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Base Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Caspase 8/metabolism , Cell Line, Tumor , DNA Primers/genetics , Enzyme Activation , Female , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Middle Aged , Phosphoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
6.
Apoptosis ; 12(9): 1645-57, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17610067

ABSTRACT

Renal cell carcinomas (RCC) exhibit marked differences in susceptibility towards anticancer drug- and TRAIL-induced apoptosis. However, the underlying mechanisms determining apoptosis-sensitivity or -resistance are not well understood. The purpose of this study was to compare gene expression patterns induced by DNA-damage- and death receptor-induced apoptosis and to detect differentially expressed genes responsible for differences in apoptosis-susceptibility. Therefore, we performed a comparative cDNA-array analysis in an apoptosis-resistant and an apoptosis-sensitive RCC cell line. In the sensitive cell line an upregulation of multiple E2F1- and p53-inducible proapaptotic and cell-cycle regulating target genes by Topotecan as well as TRAIL was observed. Interestingly, several antiapoptotic NFkappaB-dependent target genes were also induced. In the resistant cell line, however, only a small number of E2F1-, p53- and NFkappaB-dependent target genes were differentially regulated. Conclusively, anticancer drug- as well as TRAIL-sensitivity go along with an upregulation of multiple proapoptotic genes. In contrast, the mechanisms of apoptosis-resistance are-at least in part-located upstream of gene induction and seem not to depend upon upregulation of de-novo-synthesized antiapoptotic genes. Conclusively, the proapoptotic stimuli are confronted with a cellular context which allows apoptosis to be conducted-in the sensitive cell line-or not-in the resistant cell line.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Topotecan/pharmacology , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Transcriptional Activation
7.
Eur J Nucl Med Mol Imaging ; 33(12): 1432-41, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16896672

ABSTRACT

PURPOSE: As primary osseous metastasis is the main adverse prognostic factor in patients with Ewing tumours, a NOD/scid mouse model for human Ewing tumour metastases has been established to examine the mechanisms of metastasis. The aim of this study was to evaluate the feasibility of diagnostic molecular imaging by small animal PET in this mouse model. METHODS: Human Ewing tumour cells were transplanted into immune-deficient NOD/scid mice via s.c injection (n=17) or i.v. injection (n=17). The animals (mean weight 23.2 g) were studied 2-7 weeks after transplantation using a submillimetre resolution animal PET scanner. To assess glucose utilisation and bone metabolism, mice were scanned after intravenous injection of 9.6 MBq (mean) 2-[(18)F]fluoro-2-deoxy-D: -glucose (FDG) or 9.4 MBq (mean) [(18)F]fluoride. Whole-body PET images were analysed visually and semi-quantitatively [%ID/g, tumour to non-tumour ratio (T/NT)]. Foci of pathological uptake were identified with respect to the physiological organ uptake in corresponding regions. RESULTS: Subcutaneously transplanted Ewing tumours demonstrated a moderately increased glucose uptake (median %ID/g 2.5; median T/NT 2.2). After i.v. transplantation, the pattern of metastasis was similar to that in patients with metastases in lung, bone and soft tissue. These metastases showed an increased FDG uptake (median %ID/g 3.6; median T/NT 2.7). Osseous metastases were additionally visible on [(18)F]fluoride PET by virtue of decreased [(18)F]fluoride uptake (osteolysis; median %ID/g 8.4; median T/NT 0.59). Metastases were confirmed immunohistologically. CONCLUSION: Diagnostic molecular imaging of Ewing tumours and their small metastases in an in vivo NOD/scid mouse model is feasible using a submillimetre resolution PET scanner.


Subject(s)
Disease Models, Animal , Positron-Emission Tomography/methods , Sarcoma, Ewing/diagnostic imaging , Sarcoma, Ewing/pathology , Animals , Cell Line, Tumor , Feasibility Studies , Fluorides/chemistry , Fluorine Radioisotopes/chemistry , Fluorodeoxyglucose F18 , Humans , Immunohistochemistry , Mice , Neoplasm Metastasis/diagnostic imaging , Sarcoma, Ewing/metabolism , Staining and Labeling , Transplantation, Heterologous
8.
Diagn Mol Pathol ; 15(2): 83-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16778588

ABSTRACT

The t(11;22) translocation is a diagnostic hallmark of various small round-cell tumors. This study correlates the performance of fluorescence in situ hybridization (FISH) and reverse transcription polymerase chain reaction (RT-PCR) in the detection of this translocation analyzing paraffin-embedded tissue specimens. As negative control samples, 10 cases of normal colon mucosa and 10 cases of colon carcinoma tissue were analyzed by FISH to determine a valid cutoff value for the diagnosis of a t(11;22) translocation. The mean number of false-positive nuclei differed significantly between disomic and polysomic control group cases (P=0.002). Therefore, the cutoff value was determined considering the pitfall polysomy. The analysis group consisted of 20 cases from the University of Düsseldorf and 10 cases from the University of Bonn. These cases were analyzed using PCR (Düsseldorf) and FISH (Bonn) using a single-blinded approach. Twenty-two cases (73.3%) were concordant in both methods. Five cases (16.7%) were discrepant, showing a positive result in FISH whereas PCR was negative. Three cases (10.0%) were analyzed by FISH, and PCR failed for nonoptimized tissue preparation. In conclusion, the detection of t(11;22) translocation is critically dependent on a thoroughly defined cutoff value for FISH and on appropriate tissue preparation for both methods. We recommend FISH as a sensitive screening tool in the detection of t(11;22) followed by subsequent PCR amplification of the specific chimeric transcript.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 22/genetics , In Situ Hybridization, Fluorescence/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Translocation, Genetic , Adult , Aged , Base Sequence , Carcinoma, Small Cell/diagnosis , Carcinoma, Small Cell/genetics , Case-Control Studies , Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , DNA Primers/genetics , Humans , In Situ Hybridization, Fluorescence/statistics & numerical data , Predictive Value of Tests , Reverse Transcriptase Polymerase Chain Reaction/statistics & numerical data , Sarcoma, Small Cell/diagnosis , Sarcoma, Small Cell/genetics , Single-Blind Method
9.
Cancer ; 108(2): 129-34, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16444703

ABSTRACT

BACKGROUND: Promoter hypermethylation is an important mechanism for silencing tumor-suppressor genes in cancer and a promising tool for development of molecular biomarkers. This study aimed to determine the prevalence of RAS association domain family protein 1A (RASSF1A) promoter hypermethylation in bronchial aspirates of patients with suspected lung cancer and to test whether this type of methylation assay could be used as a diagnostic adjunct to conventional cytology. METHODS: Two hundred three bronchial aspirates from patients with suspected lung cancer were analyzed for RASSF1A hypermethylation by using a sensitive quantitative methylation-specific polymerase chain reaction (QMSP). RESULTS: RASSF1A hypermethylation was found in 88% (35 of 40), 28% (31 of 111), and 100% (6 of 6) of bronchial aspirates collected from patients diagnosed with small cell lung cancer, nonsmall cell lung cancer, and combined small cell lung cancer, respectively. No hypermethylation was detected in patients diagnosed with nonneoplastic lung disease (0 of 46). Depending on histologic subtype, up to 82% of cases presenting with a negative histology showed a positive methylation assay. CONCLUSIONS: The QMSP analysis of RASSF1A hypermethylation enabled a highly specific distinction between patients diagnosed with lung cancer and those with nonneoplastic lung disease. These results suggested that a QMSP assay is a promising molecular tool for diagnosis of primary lung cancer.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Small Cell/diagnosis , DNA Methylation , Lung Neoplasms/diagnosis , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics , Adenocarcinoma/chemistry , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Biopsy, Needle , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/chemistry , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Case-Control Studies , Cell Line, Tumor , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Diagnosis, Differential , Female , Gene Silencing , Humans , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Molecular Diagnostic Techniques/methods , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Tumor Suppressor Proteins/analysis
10.
Clin Cancer Res ; 11(21): 7728-34, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16278393

ABSTRACT

PURPOSE: Recent studies have detected aberrant promoter methylation of adenomatous polyposis coli promoter 1 A (APC), cyclin-dependent kinase inhibitor-2A (p16(INK4a)), retinoic acid receptor beta2, and RAS association domain family protein 1 (RASSF1A) in bronchial aspirates and suggested their use as biomarkers for lung cancer diagnostics. The purpose of this study was to validate these candidate marker genes in a retrospective cohort study. EXPERIMENTAL DESIGN: Bronchial aspirates collected from a cohort comprising 247 patients with suspected lung cancer were investigated retrospectively regarding aberrant promoter methylation using a quantitative methylation-specific real-time PCR (QMSP). RESULTS: Eighty-nine patients were diagnosed with primary lung cancer, 102 had benign lung disease, and 56 showed miscellaneous other conditions. A panel consisting of APC, p16(INK4a), and RASSF1A emerged as useful combination. This panel detected aberrant methylation in bronchial aspirates of 22 of 35 (63%) and 21 of 44 (44%) centrally and peripherally located primary lung cancers, respectively. Bronchial aspirates also showed aberrant methylation in 5 of 7 (71%) patients with a recurrent lung cancer and in 8 of 30 (27%) cases without tumor recurrence. In contrast, only 1 of 102 patients with benign lung disease displayed a (false) positive test result. Rarely, aberrant methylation was found in patients with other malignancies (3 of 16). The QMSP assay correctly confirmed lung cancer in 8 of 12 (67%) cases with an ambiguous cytology. Moreover, it disclosed 9 of 26 (35%) of peripheral tumors lacking simultaneous cytologic or histologic diagnosis of malignancy. CONCLUSIONS: Our findings suggest that the QMSP assay could be applied as a reflex test in cases of suspected lung cancer that defy a definite diagnosis by conventional methods. Thus, the assay could be a useful diagnostic adjunct especially regarding peripheral tumors.


Subject(s)
Bronchi/metabolism , DNA Methylation , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Adenomatous Polyposis Coli Protein/metabolism , Adult , Aged , Aged, 80 and over , Biopsy , Bronchoscopy , Case-Control Studies , Cell Line, Tumor , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Primers/chemistry , False Positive Reactions , Female , Humans , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Polymerase Chain Reaction , Receptors, Retinoic Acid/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Sulfites/pharmacology , Temperature , Tumor Suppressor Proteins/genetics
11.
Anticancer Res ; 25(5): 3251-8, 2005.
Article in English | MEDLINE | ID: mdl-16101135

ABSTRACT

BACKGROUND: Testing microsatellite instability seems to be a useful tool for the initial screening of putative non-polyposis colorectal cancer (HNPCC), preceding analysis of germ-line mutations of DNA mismatch repair genes. However, diagnosis of microsatellite instability becomes complicated when highly-damaged DNA from formalin-fixed paraffin-embedded tissue specimens has to be investigated. MATERIALS AND METHODS: A new methodical approach was established based on special multiplex PCR regimes (e.g., on touch-up cycling conditions), allowing both sufficient, as well as specific, amplification of the Bethesda reference panel loci with low quality DNA as template. RESULTS: By applying our new method, microsatellite instability could be analyzed successfully in 75 out of 84 investigated tumors (89%), whereas, by using standard PCR protocols, microsatellite analysis failed in 36% of the investigated cases. CONCLUSION: Our new methodical approach should be recommended for the use of archival material since it allows an efficient and accurate amplification of the Bethesda marker fragments and is less dependent on the DNA quality.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA, Neoplasm/analysis , Microsatellite Repeats/genetics , Polymerase Chain Reaction/methods , Adaptor Proteins, Signal Transducing , Allelic Imbalance , Carrier Proteins , Colorectal Neoplasms, Hereditary Nonpolyposis/chemistry , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , DNA Damage , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Formaldehyde , Humans , Immunohistochemistry , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Paraffin Embedding , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Tissue Fixation
12.
Cancer Lett ; 222(2): 165-71, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15863265

ABSTRACT

In this study, we analyzed the role of the p53 status for paclitaxel/Taxol sensitivity in renal cell carcinomas (RCCs) of the clear cell type. Using immunohistochemistry, nuclear p53 accumulation could not be correlated to the paclitaxel/Taxol sensitivity. DNA sequencing detected a p53 gene mutation in two out of eight RCC cell lines, i.e. in exon 8 (cell line clearCa-6), and in exon 9 (cell line clearCa-5). No correlation, however, was found between the p53 status of our RCC cell lines and their paclitaxel/Taxol sensitivity as indicated by the IC50 values. However, paclitaxel-induced growth inhibition in paclitaxel-sensitive RCC cell lines was accompanied by an increase in apoptosis, irrespective of their p53 status. Although CD95 up-regulation was observed in renal cell carcinoma with wild-type p53 upon paclitaxel treatment, paclitaxel-induced apoptosis itself is triggered independently from the CD95 system. In conclusion, the p53 status cannot predict paclitaxel/Taxol sensitivity in RCC cell lines of the clear cell type.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Genes, p53 , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Paclitaxel/pharmacology , Apoptosis/drug effects , DNA Mutational Analysis , DNA, Neoplasm , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , Sequence Analysis, DNA , Tumor Cells, Cultured
13.
Int J Cancer ; 111(2): 224-8, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15197775

ABSTRACT

Aberrant promoter methylation is an important mechanism for gene silencing. In the present study, 50 Barrett's esophagus-associated esophageal adenocarcinomas (ADC), 50 cardiac ADC and 50 gastric ADC were investigated by means of methylation-specific real-time PCR for hypermethylation in the tumor suppressor genes APC, p16(INk4A) and p14(ARF). Additionally, expression of p16(INK4A) protein in the carcinomas was assessed using immunohistochemistry. Marked differences in hypermethylation were found between esophageal, cardiac and gastric ADC in the APC gene (78% vs. 32% vs. 84%) and in the p16(INK4A) gene (54% vs. 36% vs. 10%). Hypermethylation of p14(ARF) was absent from esophageal ADC and present infrequently in cardiac (2%) and gastric ADC (10%). Complete loss of p16(INK4A) protein expression was detectable in 45% of all tumors and was significantly associated with hypermethylation of the p16(INK4A) gene (p<0.0001, chi(2)-test). Our results suggest that hypermethylation of p16(INK4A) and APC are frequent findings in esophageal, cardiac and gastric ADC. Additionally, the data point to a tumor specific methylation pattern in upper gastrointestinal ADC.


Subject(s)
Adenocarcinoma/genetics , Cardia/pathology , DNA Methylation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/physiopathology , Gene Expression Profiling , Genes, APC , Genes, p16 , Stomach Neoplasms/genetics , Stomach Neoplasms/physiopathology , Tumor Suppressor Protein p14ARF/genetics , Adenocarcinoma/physiopathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/complications , Female , Humans , Immunohistochemistry , Male , Middle Aged
14.
Int J Cancer ; 111(2): 259-63, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15197780

ABSTRACT

In a retrospective analysis of 203 patients with small cell lung cancer (SCLC), we examined the prognostic value of c-kit expression on survival. Expression of c-kit was examined immunohistochemically in formalin-fixed, paraffin-embedded tissue sections. c-kit was observed in 87.7% of SCLC tumors. Using the Kaplan-Meier model, we found that lack of c-kit expression was associated with significantly shorter survival time compared to the presence of c-kit expression (mean survival 151 +/- 27 vs. 358 +/- 49 days, p = 0.0084). Moreover, the proportion of c-kit(+) cells within the tumor was also related to survival time. Patients with tumors in which >75% of cells stained positive for c-kit had a mean overall survival time of 424 (+/-72) compared to 295 (+/-67) days for patients with 25-75% c-kit(+) tumor cells. Patients with tumors containing <25% c-kit(+) cells had the worst survival, with 164 (+/-24) days (p = 0.0033). Further parameters associated with short survival times were low performance status, elevated levels of lactate dehydrogenase and higher stage according to the TNM classification. Multivariate analysis using the Cox regression model showed that the proportion of c-kit(+) cells within the tumor specimen was one of 3 independent prognostic parameters (p = 0.004) for overall survival next to TNM classification (p = 0.001) and performance status (p < 0.001).


Subject(s)
Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Gene Expression Profiling , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-kit/biosynthesis , Aged , Female , Humans , Male , Middle Aged , Multivariate Analysis , Prognosis , Survival Analysis
15.
Hum Pathol ; 35(3): 371-6, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15017595

ABSTRACT

Intestinal metaplasia in Barrett's esophagus (BIM) is a precancerous condition, whereas the carcinogenic potential of intestinal metaplasia of the cardia (CIM) is uncertain. Although clinically important, histological distinction between both conditions by endoscopic biopsies is considered problematic. In the present study, 4-mm samples of BIM (n=31) and CIM (n=9) were selected from esophagectomy specimens that had been resected for esophageal cancer. Slides were coded and stained with hematoxylin and eosin (H&E), Alcian blue-periodic acid-Schiff (PAS), cytokeratins (CK) 7 and 20, and CD10, which labels the intestinal brush border. The predictive value of these stains for the recognition of BIM and CIM was evaluated independently by two senior pathologists. With the use of H&E-stained slides exclusively, BIM samples were categorized correctly in 93.5% and 83.9% of cases (pathologists 1 and 2, respectively), and CIM samples, in 100% and 88.9% of cases. Alcian blue-PAS-positive goblet cells were identified by both investigators in all BIM and CIM samples. BIM-typical CK 7 and 20 immunostaining pattern was identified in 90.3%/83.9% of BIM but only in 11.1%/11.1% of CIM. CD10-positive brush border was present in 32.3%/25.8% of BIM and in 88.9%/88.9% of CIM. When HE-stained slides and immunohistologically stained slides were used together for tissue recognition, BIM were categorized correctly in 90.3%/80.6% of cases, and CIM, in 88.9%/88.9% of cases. In conclusion, BIM and CIM can be usually distinguished on the basis of HE sections. CK 7 and CK 20 expression pattern analysis discriminates correctly between BIM and CIM in the majority of cases. CD10-positive intestinal brush border is present in the majority of CIM but only in a minority of BIM. However, immunohistochemical investigations could not improve the diagnostic accuracy of HE histology alone.


Subject(s)
Barrett Esophagus/pathology , Cardia/pathology , Precancerous Conditions/pathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/metabolism , Biomarkers/analysis , Cardia/metabolism , Diagnosis, Differential , Esophagectomy , Female , Humans , Immunoenzyme Techniques , Intermediate Filament Proteins/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Keratin-20 , Keratin-7 , Keratins/metabolism , Male , Metaplasia/metabolism , Metaplasia/pathology , Middle Aged , Neprilysin/metabolism , Precancerous Conditions/metabolism , Reproducibility of Results
16.
Cardiology ; 100(3): 120-8, 2003.
Article in English | MEDLINE | ID: mdl-14631132

ABSTRACT

OBJECTIVE: We tested the hypothesis that patients with biopsy-proven inflammatory infiltrates have an impaired vasodilator capacity of the coronary microvasculation. METHODS: In 80 patients with clinically suspected inflammatory heart disease, coronary regulation was assessed with the argon method (1) at rest and maximal coronary flow (V(cor)/V(max)) and (2) at rest and minimal coronary resistance (R(cor)/R(min)) both before and after dipyridamole (0.5 mg/kg body weight) treatment. RESULTS: Compared to patients without evidence of myocardial inflammation in endomyocardial biopsy (n = 51) but similar demographic characteristics, patients with biopsy-proven inflammatory infiltrates (n = 29) showed significantly reduced maximal coronary flow (286 +/- 122 vs. 189 +/- 78 ml/min x 100 g; p = 0.001) and minimal coronary resistance was increased (0.40 +/- 0.17 vs. 0.60 +/- 0.27 mm Hg x min x 100 g/ml(-1), p = 0.001). The coronary reserve in patients with inflammatory infiltrates was markedly reduced (3.5 +/- 1.1 to 2.4 +/- 0.81, p = 0.001). CONCLUSION: Patients with biopsy-proven inflammatory infiltrates have a diminished coronary reserve due to reduced coronary vasodilator capacity. This may be due to the involvement of the intramural coronary vasculature in inflammatory heart disease.


Subject(s)
Coronary Circulation/physiology , Coronary Disease/pathology , Myocarditis/pathology , Adult , Biopsy, Needle , Blood Flow Velocity , Cardiac Output , Cohort Studies , Coronary Angiography , Coronary Disease/complications , Coronary Disease/diagnostic imaging , Echocardiography , Female , Humans , Hypertrophy, Left Ventricular/complications , Hypertrophy, Left Ventricular/diagnostic imaging , Immunohistochemistry , Male , Middle Aged , Myocarditis/complications , Probability , Prospective Studies , Risk Assessment , Sensitivity and Specificity , Severity of Illness Index , Vascular Resistance
17.
Hum Pathol ; 34(9): 850-6, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14562279

ABSTRACT

p63, a member of the p53 gene family, is known to encode functionally antagonistic protein isoforms. Although transactivating protein isoforms display p53-like functions, deltaNp63 isoforms act toward p53 in a dominant negative way. Using immunohistochemistry, we examined the expression of pan-p63 and deltaNp63 in 50 esophageal squamous cell carcinomas (SCCs) as well as in squamous low-grade intraepithelial neoplasias (S-LGINs; n = 4) and high-grade intraepithelial neoplasias (S-HGINs; n = 18). Additionally, 50 esophageal adenocarcinomas (ADCs) that arose in Barrett's esophagus (BE) as well as adjacent specialized metaplastic epithelium (SE; n = 41), low-grade intraepithelial neoplasias (B-LGINs; n = 27), and high-grade intraepithelial neoplasias (B-HGINs; n = 21) in BE were investigated. Furthermore, p63 gene amplification was determined by fluorescent differential polymerase chain reaction in a subset of 10 SCCs and 10 ADCs. Whereas in normal esophageal epithelium, expression of pan-p63 is invariably restricted to the basal cell layer, in 100% of S-LGINs, 94.4% of S-HGINs, and 88.0% of SCCs, expression of p63 was found in >75% of the cells. Concerning BE, only in a small subset of SEs (7.3%), B-LGINs (14.8%), B-HGINs (14.3%) and ADCs (16.0%) was a weak p63 protein expression (<10% positive cells) detectable, whereas the rest of the samples were completely negative. Expression of deltaNp63 was identical to expression of pan-p63 in the vast majority of samples. p63 gene amplification was found in 2 of 10 (20.0%) investigated SCCs and in 1 of 10 (10.0%) ADCs. In summary, strong expression of p63, especially its deltaNp63 isoforms, is a frequent finding in esophageal precancerous and cancerous squamous lesions, whereas this is not the case in carcinogenesis of BE. p63 gene amplification is an infrequent finding in esophageal SCCs and ADCs and does not correlate with protein overexpression.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma in Situ/metabolism , Carcinoma, Squamous Cell/metabolism , DNA-Binding Proteins/metabolism , Esophageal Neoplasms/metabolism , Gene Amplification , Membrane Proteins , Phosphoproteins/metabolism , Trans-Activators/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Count , DNA, Neoplasm/analysis , DNA-Binding Proteins/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Genes, Tumor Suppressor , Humans , Immunoenzyme Techniques , Male , Middle Aged , Phosphoproteins/genetics , Polymerase Chain Reaction , Protein Isoforms/genetics , Protein Isoforms/metabolism , Trans-Activators/genetics , Transcription Factors , Tumor Suppressor Proteins
18.
Anticancer Res ; 23(2B): 1489-93, 2003.
Article in English | MEDLINE | ID: mdl-12820414

ABSTRACT

BACKGROUND: The proto-oncogene c-myc is known to be involved in the regulation of proliferation, apoptosis and cell differentiation. MATERIALS AND METHODS: Amplification of c-myc was determined by means of differential PCR in 77 surgically treated stage I or II oesophageal squamous cell carcinomas (SCC) as well as in 43 locally advanced SCC (cT3-4 cN0-1 cM0) treated by radiochemotherapy and facultatively by surgery. The findings were correlated to overall survival and to response to radiochemotherapy. RESULTS: C-myc gene amplification was present in 8 out of 77 surgically treated SCC (10.4%) and in 13 out of 43 multimodally treated SCC (30.2%). Among the surgically treated tumours, the presence of c-myc amplification was correlated with high proliferative activity (p = 0.0399) but not with overall survival. Among the multimodally treated SCC, c-myc amplification tended to be correlated with response to chemotherapy and response to radiochemotherapy (not significant) whereas no impact on overall survival was found. CONCLUSION: Amplification of c-myc is found more frequently in advanced stages of oesophageal SCC than in early stages. C-myc amplification, however, does not influence the overall survival of oesophageal SCC patients treated either by surgery alone or by multimodal therapy.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Gene Amplification , Genes, myc , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/surgery , Cell Division , Cisplatin/administration & dosage , Clinical Trials as Topic , Combined Modality Therapy , Disease Progression , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/surgery , Esophagectomy , Etoposide/administration & dosage , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Leucovorin/administration & dosage , Male , Middle Aged , Multicenter Studies as Topic , Polymerase Chain Reaction , Prognosis , Prospective Studies , Proto-Oncogene Mas , Remission Induction , Survival Analysis , Treatment Outcome
19.
J Pathol ; 200(1): 16-22, 2003 May.
Article in English | MEDLINE | ID: mdl-12692836

ABSTRACT

The mitotic spindle assembly checkpoint modulates the timing of anaphase initiation in response to improper alignment of chromosomes at the metaphase plate. The BUB gene family encodes proteins which are part of a large multi-protein kinetochore complex and which are believed to be key components of the checkpoint regulatory pathway. Failure of this surveillance system can lead to genomic instability and could be responsible for the increased incidence of aneuploidy in gastric cancer. Since mutations of BUB genes have not been identified in gastric cancer to date, altered BUB expression levels may significantly impair mitotic checkpoint function. To explore this possibility, the expression levels of BUB1, BUBR1, and BUB3 were determined in 43 gastric carcinomas and corresponding normal gastric mucosa by reverse transcription-polymerase chain reaction (RT-PCR). Gene expression levels were compared with histopathological parameters and DNA ploidy, as well as with proliferative activity, measured by Ki-67 mRNA expression. To the authors' knowledge, this is the first study to investigate the expression levels of mitotic checkpoint genes together with DNA ploidy in gastric cancer. BUB1 was overexpressed in 84%, BUBR1 in 68%, and BUB3 in 79% of gastric cancers. This study also revealed that all three genes were simultaneously overexpressed in 61% of the tumours and that there was a statistically significant positive correlation between overexpression of BUB1, BUBR1 or BUB3 and Ki-67 expression (p < 0.001). Eighty-one per cent of the tumours were classified as aneuploid. However, no correlation was found between ploidy and BUB transcript expression levels. These results suggest that inactivation of the mitotic checkpoint genes BUB1, BUBR1, and BUB3 by epigenetic silencing does not seem to play a role in gastric carcinogenesis. The strong correlation of BUB expression level and tumour cell proliferation suggests that BUB overexpression is a proliferation-dependent phenomenon in gastric cancer. However, overexpression due to lack of normal BUB protein function or due to a yet unknown additional BUB function has to be considered.


Subject(s)
Adenocarcinoma/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Protein Kinases/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , DNA, Neoplasm/genetics , Female , Gastric Mucosa , Genes, Neoplasm/genetics , Humans , Ki-67 Antigen/genetics , Male , Middle Aged , Ploidies , Poly-ADP-Ribose Binding Proteins , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
20.
Hum Pathol ; 34(2): 174-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12612886

ABSTRACT

The CD95 (Apo-1/Fas) receptor-ligand system is a key regulator of apoptosis. Down-regulation of CD95 receptor and up-regulation of CD95 ligand has been reported in a variety of human tumors and is thought to confer a selective survival advantage. To explore the relevance of the CD95 system for tumor progression and prognosis in clear cell renal cell carcinomas (RCCs), we analyzed CD95 receptor and ligand expression in formalin-fixed tissue from 149 clear cell RCCs by immunohistochemistry. CD95 ligand expression could not be detected in nonneoplastic tubule epithelia and in clear cell RCCs. In contrast, CD95 receptor expression was found in the great majority of clear cell RCCs, and no down-regulation of CD95 receptor protein was evident when compared with nonneoplastic tubule epithelia. Although a significant increase (P = 0.004) of CD95 receptor expression was evident from well-differentiated (G1) to poorly differentiated (G3) RCCs, CD95 receptor expression was not correlated with tumor stage or survival of RCC patients. In conclusion, clear cell RCCs differ from other types of human cancer by their failure to down-regulate CD95 receptor expression or up-regulate CD95 ligand expression during tumor progression. These ex vivo observations suggest that down-regulation of CD95 receptor expression may not provide an additional selective growth advantage to RCC cells and thus further confirm our previous in vitro observations on a functional impairment of CD95-mediated apoptosis in RCC.


Subject(s)
Adenocarcinoma, Clear Cell/chemistry , Kidney Neoplasms/chemistry , fas Receptor/analysis , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Aged, 80 and over , Apoptosis , Female , Humans , Immunohistochemistry , Kidney/chemistry , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...