Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 97(8): 1106-15, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17876328

ABSTRACT

Recent evidence suggests that Runt-related transcription factors play a role in different human tumours. In the present study, the localisation of the Runt-related transcription factor-2 (Runx2), its transcriptional activity, as well as its regulation of expression was analysed in human pancreatic ductal adenocarcinoma (PDAC). Quantitative real-time PCR and immunohistochemistry were used for Runx2 expression and localisation analysis. Runt-related transcription factor-2 expression was silenced using specific siRNA oligonucleotides in pancreatic cancer cells (Panc-1) and immortalised pancreatic stellate cells (IPSCs). Overexpression of Runx2 was achieved using a full-length expression vector. TGF-beta1, BMP2, and other cytokines were assessed for their potential to regulate Runx2 expression. There was a 6.1-fold increase in median Runx2 mRNA levels in PDAC tissues compared to normal pancreatic tissues (P<0.0001). Runt-related transcription factor-2 was localised in pancreatic cancer cells, tubular complexes, and PanIN lesions of PDAC tissues as well as in tumour-associated fibroblasts/stellate cells. Coculture of IPSCs and Panc-1 cells, as well as treatment with TGF-beta1 and BMP2, led to increased Runx2 expression in Panc-1 cells. Runt-related transcription factor-2 overexpression was associated with decreased MMP1 release as well as decreased growth and invasion of Panc-1 cells. These effects were reversed by Runx2 silencing. In conclusion, Runx2 is overexpressed in PDAC, where it is regulated by certain cytokines such as TGF-beta1 and BMP2 in an auto- and paracrine manner. In addition, Runx2 has the potential to regulate the transcription of extracellular matrix modulators such as SPARC and MMP1, thereby influencing the tumour microenvironment.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Pancreatic Neoplasms/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression , Humans , Immunohistochemistry , Matrix Metalloproteinase 1/biosynthesis , Pancreatic Neoplasms/genetics , RNA, Messenger/analysis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection
2.
Neoplasia ; 9(2): 136-46, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17356710

ABSTRACT

Pancreatic carcinoma has an extremely bad prognosis due to lack of early diagnostic markers and lack of effective therapeutic strategies. Recently, we have established an in vitro model recapitulating the first steps in the carcinogenesis of the pancreas. SV40 large T antigen-immortalized bovine pancreatic duct cells formed intrapancreatic adenocarcinoma tumors on k-ras(mut) transfection after orthotopic injection in the nude mouse pancreas. Here we identified genes and proteins differentially expressed in the course of malignant transformation using reciprocal suppression subtractive hybridization and 2D gel electrophoresis and mass spectrometry, respectively. We identified 34 differentially expressed genes, expressed sequence tags, and 15 unique proteins. Differential expression was verified for some of the genes or proteins in samples from pancreatic carcinoma. Among these genes and proteins, the majority had already been described either to be influenced by a mutated ras or to be differentially expressed in pancreatic adenocarcinoma, thus proving the feasibility of our model. Other genes and proteins (e.g., BBC1, GLTSCR2, and rhoGDIalpha), up to now, have not been implicated in pancreatic tumor development. Thus, we were able to establish an in vitro model of pancreatic carcinogenesis, which enabled us to identify genes and proteins differentially expressed during the early steps of malignant transformation.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Animals , Antigens, Polyomavirus Transforming/physiology , Carcinoma, Pancreatic Ductal/pathology , Cattle , Cell Line, Transformed/metabolism , Cell Transformation, Viral/genetics , Chronic Disease , Disease Progression , Electrophoresis, Gel, Two-Dimensional , Genes, ras , Humans , Neoplasm Proteins/blood , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Pancreatic Ducts/cytology , Pancreatic Neoplasms/pathology , Pancreatitis/genetics , Pancreatitis/metabolism , Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Subtraction Technique
3.
Cancer Gene Ther ; 13(1): 65-73, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16096652

ABSTRACT

The prognosis of peritoneal spread from gastrointestinal cancer and subsequent malignant ascites is poor, and current medical treatments available are mostly ineffective. Targeted chemotherapy with intraperitoneal prodrug activation may be a beneficial new approach. L293 cells were genetically modified to express the cytochrome P450 enzyme 2B1 under the control of a cytomegalovirus immediate early promoter. This CYP2B1 enzyme converts ifosfamide to its active cytotoxic compounds. The cells are encapsulated in a cellulose sulfate formulation (Capcell). Adult Balb/c mice were inoculated intraperitoneally with 1 x 10(6) colon 26 cancer cells, previously transfected with GFP to emit a stable green fluorescence, by injection into the left lower abdominal quadrant. Two or five day's later animals were randomly subjected to either i.p. treatment with ifosfamide alone or ifosfamide combined with microencapsulated CYP2B1-expressing cells. Peritoneal tumor volume and tumor viability were assessed 10 days after tumor inoculation by means of fluorescence microscopy, spectroscopy and histology. Early i.p. treatment with ifosfamide and CYP2B1 cells resulted in a complete response. Treatment starting on day 5 and single-drug treatment with ifosfamide resulted in a partial response. These results suggest that targeted i.p. chemotherapy using a combination of a prodrug and its converting enzyme may be a successful treatment strategy for peritoneal spread from colorectal cancer.


Subject(s)
Cytochrome P-450 CYP2B1/pharmacology , Drug Compounding , Genetic Therapy/methods , Ifosfamide/therapeutic use , Peritoneal Neoplasms/therapy , Animals , Cytochrome P-450 CYP2B1/metabolism , Ifosfamide/metabolism , Ifosfamide/pharmacology , Male , Mice , Mice, Inbred BALB C , Peritoneal Neoplasms/metabolism , Prodrugs/metabolism , Prodrugs/pharmacology , Transfection , Tumor Cells, Cultured
4.
Pancreatology ; 2(4): 421-4, 2002.
Article in English | MEDLINE | ID: mdl-12138232

ABSTRACT

BACKGROUND/AIMS: Analysis of gene expression is dependent on normalization using housekeeping genes. However, many of these housekeeping genes (e.g. GAPDH, beta-actin) are upregulated in chronic pancreatitis and pancreatic cancer, and cannot be used for normalization. For this reason we tried to identify a housekeeping gene useful for expression analysis in pancreatic diseases. METHODS: RNA isolated from various tissues and states of disease was subjected to reverse transcription and subsequently amplified by PCR using primers for GAPDH and for the ribosomal highly basic 23-kDa (rb 23-kDa, RPL13A) protein. RESULTS: As anticipated, expression of GAPDH varied markedly in the different tissues, whereas the expression of rb 23-kDa was constant in all samples investigated. CONCLUSION: We recommend the use of the ribosomal highly basic 23-kDa protein as a standard for normalization at least for the pancreas and the prostate.


Subject(s)
Gene Expression , Pancreatic Diseases/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Molecular Weight , RNA, Messenger/metabolism , Reference Standards , Reverse Transcriptase Polymerase Chain Reaction
5.
Virchows Arch ; 438(6): 581-90, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11469690

ABSTRACT

Mutation of the K-ras gene is thought to be an early and important event in pancreatic carcinogenesis. In order to study the role of this molecular alteration in the transition from the normal to the neoplastic pancreatic cell, bovine pancreatic duct cells were first immortalized by SV40 large T antigen (Ag) complementary (c)DNA transfection and then transfected with a mutated K-ras gene. As did primary duct cells, the immortalized duct cells (more than 100 passages) expressed cytokeratins, carbonic anhydrase type-II, cystic fibrosis transmembrane conductance regulator (CFTR), and multidrug resistance (mdr). They grew as a single layer after transplantation under plastic domes and formed three-dimensional structures resembling ducts when grown on Matrigel. Cell growth was stimulated by insulin, epidermal growth factor (EGF), transforming growth factor (TGF)-alpha, but cells did not respond to gastrin and CCK-8. They did not form colonies in soft agar nor did they form tumors in nude mice. Immortalized cells transfected with mutated K-ras acquired the ability to form tumors after orthotopic injection into the nude mouse pancreas. It is concluded that SV 40 immortalized bovine pancreatic


Subject(s)
Cell Transformation, Neoplastic/pathology , Genes, ras/genetics , Mutation , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Transfection/methods , Animals , Antigens, Polyomavirus Transforming/genetics , Biomarkers/analysis , Cattle , Cell Division/drug effects , Cell Line, Transformed , Cell Transformation, Neoplastic/chemistry , Cell Transformation, Neoplastic/genetics , Clone Cells , DNA, Complementary/genetics , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fluorescent Antibody Technique, Indirect , Insulin/pharmacology , Mice , Mice, Nude , Pancreatic Ducts/drug effects , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/genetics , Polymerase Chain Reaction , RNA, Viral/biosynthesis , Transforming Growth Factor alpha/pharmacology
6.
Cancer Res ; 61(2): 550-5, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212248

ABSTRACT

Proliferation of fibrotic tissue (desmoplasia) is one of the hallmarks of several epithelial tumors including pancreatic adenocarcinoma. This tissue reaction may be deleterious or advantageous to the host or tumor. In a systematic analysis, we identified two growth factors expressed by human pancreatic carcinoma cells that are positively correlated with the ability to induce fibroblast proliferation both in vitro and in vivo, i.e., transforming growth factor (TGF)-beta1 and fibroblast growth factor-2. Here we demonstrate that the overexpression of TGF-beta1 induced up-regulation of matrix proteins and growth factors in the TGFbeta1-transfected pancreatic tumor cells. Furthermore, transfection of PANC-1 cells induces the same change in fibroblasts in either cocultivation experiments or when they are grown in conditioned medium from TGF-beta1-transfected PANC-1 cells. TGF-beta1-transfected pancreatic tumor cells induced a rich stroma after orthotopical transplantation in the nude mouse pancreas. The transfer of a single growth factor, TGF-beta1, conveys the ability to induce a fibroblast response similar to that seen in desmoplasia in human pancreatic adenocarcinoma. This effect cannot only be attributed to direct effects of TGF-beta1 but also results from the up-regulation of several other factors including collagen type I, connective tissue growth factor, and platelet-derived growth factor.


Subject(s)
Pancreatic Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Animals , Cell Division , Coculture Techniques , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Growth Substances/metabolism , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Transfection , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Transplantation, Heterologous , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , Up-Regulation
7.
Teratog Carcinog Mutagen ; 21(1): 97-106, 2001.
Article in English | MEDLINE | ID: mdl-11135324

ABSTRACT

CD44 is an integral cell-surface glycoprotein. Overexpression of the CD44 standard (CD44st) and its variants (CD44v) has been implicated in transformation and progression of many cancer types. Here, we investigated expression of CD44st, CD44v3-7, CD44v7/8, and v10 in five human pancreatic tumor cell lines and normal human pancreatic duct cells transfected with the SV40 large T antigen. CD44st and its variant proteins were quantified using immunocytochemistry and flow cytometry. CD44v7 was expressed at low levels, whereas CD44st, CD44v3, CD44 v4, CD44v, and CD44v6 were expressed at moderate levels in all pancreatic tumor cell lines. In contrast, CD44v7/8 and CD44v10 were expressed at very low levels in two out of the five pancreatic tumor cell lines. Overall, staining of CD44st and CD44 variants was significantly weaker compared to another surface molecule, ICAM-1, reported to be overexpressed in pancreatic cancer cells. Furthermore, the SV40 large T transfected duct cells showed only a weak staining for CD44st, CD44v5, and CD44v6. To determine a possible mechanism for the regulation of surface expression of CD44st, v5 and v6, we incubated Panc-1 cells with bFGF, TGF-beta1, EGF, TNFalpha, and IFNgamma. Only IFNgamma affected the CD44 expression by down-regulation of CD44v6. The constitutive expression of CD44 variants seems to be associated with the malignant state of invasive carcinoma.


Subject(s)
Adenocarcinoma/metabolism , Hyaluronan Receptors/metabolism , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Cell Line, Transformed/drug effects , Cell Line, Transformed/metabolism , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Growth Substances/pharmacology , Humans , Hyaluronan Receptors/classification , Pancreatic Ducts/cytology , Pancreatic Ducts/drug effects , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
8.
Ann N Y Acad Sci ; 880: 50-65, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415850

ABSTRACT

Although pancreatic adenocarcinoma has become one of the best characterized malignant diseases, severe diagnostic and therapeutic problems are still associated with this disease. The establishment of a molecular model of pancreatic carcinogenesis may provide tools that could result in earlier diagnosis of this disease and, in turn, improves prognosis. Since pancreatic adenocarcinoma seems to originate in epithelial cells in the pancreatic ducts, cultivation of native pancreatic duct epithelial cells (PDEC) is the initial step in the establishment of an in vitro model of pancreatic carcinogenesis. As these native cells survive only a short period in culture, the aim of this study was to establish a stable pancreatic duct cell line by immortalization with the SV40 large T antigen. Furthermore, initial steps in pancreatic carcinogenesis should possibly be imitated by additional transfections of mutated ki-ras and/or mutated p53 genes. By optimization of the isolation protocol and the culture medium, yield as well as proliferative activity of isolated PDEC was increased considerably. Transfection of SV40 large T antigen resulted in an increase in the proliferative lifetime of the isolated cells, but no real immortal phenotype was obtained. Moreover, one step in the transformation from the normal to the malignant phenotype was imitated successfully by additional transfection of mutated ki-ras.


Subject(s)
Cell Line, Transformed , Pancreatic Ducts/cytology , Animals , Antigens, Polyomavirus Transforming/genetics , Cattle , Cell Line , Cell Transformation, Neoplastic , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured
10.
Ann N Y Acad Sci ; 880: 337-51, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415878

ABSTRACT

The prognosis of pancreatic cancer is poor, and current medical treatment is mostly ineffective. The aim of this study was to design a new treatment modality in an animal model system. We describe here a novel treatment strategy employing a mouse model system for pancreatic carcinoma. Embryonal kidney epithelial cells were genetically modified to express the cytochrome P450 subenzyme 2B1 under the control of a cytomegalovirus (CMV) immediate early promoter. This CYP2B1 gene converts ifosfamide to its active cytotoxic compounds, phosphoramide mustard, which alkylates DNA, and acrolein, which alkylates proteins. The cells were then encapsulated in a cellulose sulphate formulation and implanted into preestablished tumors derived from a human pancreatic tumor cell line. Intraperitoneal administration of low-dose ifosfamide to tumor bearing mice that received the encapsulated cells results in partial or even complete tumor ablation. Such an in situ chemotherapy strategy utilizing genetically modified cells in an immunoprotected environment may prove useful for solid tumor therapy in man.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Cytochrome P-450 CYP2B1/genetics , Genetic Therapy/methods , Ifosfamide/therapeutic use , Pancreatic Neoplasms/therapy , Prodrugs/therapeutic use , Animals , Capsules , Cell Line , Cytochrome P-450 CYP2B1/biosynthesis , Disease Models, Animal , Gene Expression , Humans , Injections , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy
13.
Int J Pancreatol ; 25(1): 39-43, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10211420

ABSTRACT

CONCLUSION: Preparation of DNA from pancreatic juice for subsequent polymerase chain reaction (PCR) is difficult, but manageable. The protocol presented offers a simple and fast solution. This method might be applicable to other complicated samples, such as saliva, would secretions, or stool washings. BACKGROUND: Of all the biological samples used for PCR amplification, pancreatic juice is the most problematic because of the presence of potential inhibitory substances and the amount of nucleases. This demands a DNA preparation procedure that is suitable for routine diagnostic PCR, and is therefore efficient and safe. This is particularly true for pancreatic juice obtained during routine endoscopy. METHODS: We describe here a simple method utilizing modified phenol/chloroform extraction and precipitation directly from native pancreatic juice suitable for diagnostic PCR applications, such as oncogenes. RESULTS: DNA could be prepared in quantitative amounts from routine endoscopic specimens. DNA could also be prepared from samples kept several days at room temperature.


Subject(s)
DNA/isolation & purification , Molecular Biology/methods , Pancreatic Juice/chemistry , Polymerase Chain Reaction , Electrophoresis, Agar Gel , Genes, ras , Humans , Polymorphism, Restriction Fragment Length
14.
Pancreas ; 17(3): 262-5, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9788539

ABSTRACT

Preparation of pancreatic duct epithelial cells from adult organs is possible by limited digestion and outgrowth of cells. These primary cells are mitotically active for only a short period. Therefore transfection with SV40 large-T antigen is one method to obtain an immortalized cell clone. Because the transfection efficacy of primary cells with conventional vectors is comparatively low, our aim was to develop conditions with improved transfection rates. Best transfection rates (approximately 6% of the resting cells) were obtained by using the BES buffered saline (BBS) calcium phosphate (Ca-P) coprecipitation technique at low pH. By using these optimized transfection parameters, primary cultures of human pancreatic duct epithelial cells were successfully transfected with the plasmid pSV3neo, bearing the large- and small-T antigen of SV40. A G 418 resistant clone (E4) was maintained in culture for 14 months before reaching terminal crisis.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Epithelial Cells/metabolism , Pancreatic Ducts/metabolism , Transfection/methods , Animals , Cattle , Cell Separation , Cells, Cultured , Epithelial Cells/cytology , Galactose/metabolism , Humans , Immunohistochemistry , Keratins/metabolism , Pancreatic Ducts/cytology
15.
Gene Ther ; 5(8): 1070-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-10326030

ABSTRACT

The prognosis of pancreatic adenocarcinoma is poor and current treatment ineffective. A novel treatment strategy is described here using a mouse model system for pancreatic cancer. Cells that have been genetically modified to express the cytochrome P450 2B1 enzyme are encapsulated in cellulose sulphate and implanted into pre-established tumours derived from human pancreatic cells. Cytochrome P450 2B1 converts the chemotherapeutic agent ifosfamide to toxic metabolites. Administration of ifosfamide to tumour-bearing mice that were recipients of implanted encapsulated cells results in partial or even complete tumour ablation. These results suggest that in situ chemotherapy with genetically modified cells in an immunoprotected environment may prove useful for application in man.


Subject(s)
Adenocarcinoma/therapy , Cytochrome P-450 CYP2B1/genetics , Genetic Therapy/methods , Ifosfamide/therapeutic use , Pancreatic Neoplasms/therapy , Prodrugs/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Capsules , Cats , Combined Modality Therapy , Cytochrome P-450 CYP2B1/metabolism , Ifosfamide/metabolism , Injections, Intralesional , Kidney/cytology , Kidney/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Prodrugs/metabolism , Transfection
16.
Adv Exp Med Biol ; 451: 97-106, 1998.
Article in English | MEDLINE | ID: mdl-10026857

ABSTRACT

The prognosis of pancreatic adenocarcinoma is poor and current treatment is for the most part ineffective. We describe here a novel treatment strategy using a mouse model system for pancreatic cancer. Human embryonic epithelial cells have been genetically modified to express the cytochrome P450 2B1 enzyme under the control of a CMV immediate-early promoter. This CYP2B1 gene converts oxazaphosphorines (ifosfamide or cyclophosphamide) to their active cytotoxic compounds, phosphoramide mustard, which alkylates DNA, and acrolein, which alkylates proteins. A number of assays were performed to demonstrate the CYP2B1 gene function as well as toxic effects on neighbouring cells (bystander effect). The cells were then encapsulated in a cellulose sulphate formulation shown to be well tolerated in the pancreas of immunocompetent mice, and injected 1 cm away from pre-established tumours derived from a human pancreatic tumour cell line (PaCa-44). Intraperitoneal administration of low-dose ifosfamide to tumour bearing mice that received the encapsulated cells results in partial or even complete tumour ablation. Such an in situ chemotherapy strategy utilizing genetically modified cells in an immunoprotected environment may prove useful for solid tumour therapy in man.


Subject(s)
Adenocarcinoma/therapy , Cell Survival/drug effects , Cyclophosphamide/therapeutic use , Cytochrome P-450 CYP2B1/genetics , Cytochrome P-450 CYP2B1/metabolism , Ifosfamide/therapeutic use , Pancreatic Neoplasms/therapy , Adenocarcinoma/pathology , Animals , Biotransformation , Capsules , Cell Line , Cell Transplantation/methods , Cloning, Molecular , Cyclophosphamide/pharmacokinetics , Cyclophosphamide/toxicity , Genetic Therapy/methods , Humans , Ifosfamide/pharmacokinetics , Ifosfamide/toxicity , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Recombinant Proteins/metabolism , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...