Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
3.
Cell Death Dis ; 7(6): e2246, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27253410

ABSTRACT

The repeated treatment of cancer cells with chemo- or radiotherapy induces therapy resistance, but it was previously unknown whether the same effect occurs upon continuous exposure of cancer cells to diet-derived chemopreventive agents. We elucidated this interesting question in pancreatic ductal adenocarcinoma, which is a highly aggressive cancer entity with a marked resistance toward gemcitabine and other cytotoxic drugs. The isothiocyanate sulforaphane, present in cruciferous vegetables, and the polyphenol quercetin, present in many fruits and vegetables induced apoptosis and reduced viability in gemcitabine-sensitive BxPC-3 cells but not in non-malignant ductal pancreas cells and mesenchymal stromal cells. In turn, BxPC-3 cells were treated with increasing concentrations of gemcitabine, sulforaphane or quercetin for more than 1 year and the surviving subclones Bx-GEM, Bx-SF and Bx-Q were selected, respectively. While Bx-GEM cells acquired a total resistance, Bx-SF or Bx-Q cells largely kept their sensitivity as proved by MTT assay, annexin staining and FACS analysis. The evaluation of the self-renewal-, differentiation- and migration-potential by colony formation, differentiation or migration assays demonstrated that cancer stem cell features were enriched in gemcitabine-resistant cells, but decreased in sulforaphane- and quercetin-long time-treated cells. These results were confirmed by orthotopic xenotransplantation of cancer cells to the mouse pancreas, where Bx-GEM formed large, Bx-Q small and Bx-SF cells almost undetectable tumors. An mRNA expression profiling array and subsequent gene set enrichment analysis and qRT-PCR confirmed that tumor progression markers were enriched in Bx-GEM, but reduced in Bx-SF and Bx-Q cells. This study demonstrates that the continuous exposure of pancreatic cancer cells to sulforaphane or quercetin does not induce resistance in surviving cells but reduces tumorigenicity by inhibition of tumor progression markers. These results highlight that cancer cells may not adapt to the preventive and therapeutic effects of a regular fruit- and vegetable-based diet.


Subject(s)
Antineoplastic Agents/pharmacology , Diet , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms/pathology , Phytochemicals/pharmacology , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Survival/drug effects , Clone Cells , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isothiocyanates/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/genetics , Quercetin/pharmacology , Sulfoxides , Gemcitabine
5.
Cell Death Dis ; 5: e1471, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25321478

ABSTRACT

Giant cell tumor of bone (GCTB) is a very rare tumor entity, which is little examined owing to the lack of established cell lines and mouse models and the restriction of available primary cell lines. The stromal cells of GCTB have been made responsible for the aggressive growth and metastasis, emphasizing the presence of a cancer stem cell population. To identify and target such tumor-initiating cells, stromal cells were isolated from eight freshly resected GCTB tissues. Tumorigenic properties were examined by colony and spheroid formation, differentiation, migration, MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, immunohistochemistry, antibody protein array, Alu in situ hybridization, FACS analysis and xenotransplantation into fertilized chicken eggs and mice. A sub-population of the neoplastic stromal cells formed spheroids and colonies, differentiated to osteoblasts, migrated to wounded regions and expressed the metastasis marker CXC-chemokine receptor type 4, indicating self-renewal, invasion and differentiation potential. Compared with adherent-growing cells, markers for pluripotency, stemness and cancer progression, including the CSC surface marker c-Met, were enhanced in spheroidal cells. This c-Met-enriched sub-population formed xenograft tumors in fertilized chicken eggs and mice. Cabozantinib, an inhibitor of c-Met in phase II trials, eliminated CSC features with a higher therapeutic effect than standard chemotherapy. This study identifies a c-Met(+) tumorigenic sub-population within stromal GCTB cells and suggests the c-Met inhibitor cabozantinib as a new therapeutic option for targeted elimination of unresectable or recurrent GCTB.


Subject(s)
Anilides/therapeutic use , Carcinogenesis/pathology , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/pathology , Molecular Targeted Therapy , Proto-Oncogene Proteins c-met/metabolism , Pyridines/therapeutic use , Anilides/pharmacology , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Chickens , Female , Fertilization , Giant Cell Tumor of Bone/metabolism , Humans , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovum/metabolism , Pyridines/pharmacology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 4: e627, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23661005

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal malignancies. Cancer stem cells (CSCs), which are not targeted by current therapies, may be the reason for pronounced therapy resistance. A new treatment option in phase II trials is cabozantinib that inhibits the pancreatic CSC surface marker and tyrosine kinase receptor c-Met. The purpose of this study was to evaluate the effect of cabozantinib to stem-like features and therapy resistance. Established PDA cell lines, a gemcitabine-resistant subclone, non-malignant pancreatic ductal cells and primary spheroidal cultures from patient tumors were analyzed by MTT-assay, flow cytometry, colony and spheroid formation assays, western blotting, qRT-PCR, antibody protein array, immunohistochemistry and morphological features. Cabozantinib inhibited viability and spheroid formation and induced apoptosis in malignant cells with minor effects in non-malignant cells. After long-term cabozantinib treatment, PDA cells had altered anti- and pro-apoptotic signaling, but still responded to cabozantinib, as apoptosis only slightly decreased and viability only slightly increased suggesting a low resistance-inducing potential of cabozantinib. In parallel, c-Met expression and the pluripotency transcription factor SOX2 were downregulated, which might counteract development of full therapy resistance in long-term treated subclones. In single-treatment studies, cabozantinib increased efficacy of gemcitabine. Most importantly, cabozantinib strongly induced apoptosis and reduced viability in PDA cell lines, which are completely resistant toward gemcitabine. In primary, CSC-enriched spheroidal cultures cabozantinib downregulated CSC markers SOX2, c-Met and CD133 and induced apoptosis. These findings suggest that the clinical use of cabozantinib may be more effective than current chemotherapeutics.


Subject(s)
Anilides/pharmacology , Apoptosis/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Stem Cells/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/drug effects , Gemcitabine
7.
Br J Cancer ; 108(7): 1449-59, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23511563

ABSTRACT

BACKGROUND: The targeting of cancer stem cells by monoclonal antibodies offers new options for therapy. CD24 is a glycosylphosphatidylinositol-anchored membrane protein with a small protein core and a high level of glycosylation. It is overexpressed in many human carcinomas and is correlated with poor prognosis. CD24 is a marker for pancreatic and ovarian cancer stem cells, whereas breast cancer stem cells are negative for CD24. In cancer cell lines, changes of CD24 expression can alter cellular properties in vitro and tumour growth in vivo. We have shown before that monotherapy with monoclonal antibody (mAb) SWA11 to CD24 effectively retarded tumour growth in xenotransplanted mice. METHODS: Here, we have investigated in more detail the molecular mechanisms of mAb SWA11 therapeutic effects in A549 lung and SKOV3ip ovarian carcinoma models in scid/beige and CD1 mice, respectively. We focused on anti-proliferative, pro-apoptotic, anti-angiogenic and microenvironmental effects of SWA11 mAb treatment. RESULTS: We find that CD24 targeting is associated with changes in tumour cell proliferation and angiogenesis. The treatment lead to increased infiltration of tumour tissues with immune cells suggesting involvement of ADCC. We found that SWA11 mAb treatment strongly altered the intratumoural cytokine microenvironment. The addition of SWA11 mAb to gemcitabine treatment strongly potentiated its anti-cancer efficacy in A549 lung cancer model. CONCLUSION: Our data demonstrate that targeting of CD24 could be beneficial for the anti-cancer treatment combined with standard chemotherapy regimes.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD24 Antigen/immunology , Cytokines/immunology , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD24 Antigen/metabolism , Carcinoma, Ovarian Epithelial , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Proliferation , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Models, Animal , Female , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/immunology , Mice , Mice, SCID , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Transplantation, Heterologous , Xenograft Model Antitumor Assays , Gemcitabine
8.
Gut ; 58(7): 949-63, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18829980

ABSTRACT

BACKGROUND AND AIMS: Emerging evidence suggests that highly treatment-resistant tumour-initiating cells (TICs) play a central role in the pathogenesis of pancreatic cancer. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered to be a novel anticancer agent; however, recent studies have shown that many pancreatic cancer cells are resistant to apoptosis induction by TRAIL due to TRAIL-activated nuclear factor-kappaB (NF-kappaB) signalling. Several chemopreventive agents are able to inhibit NF-kappaB, and favourable results have been obtained--for example, for the broccoli compound sulforaphane-in preventing metastasis in clinical studies. The aim of the study was to identify TICs in pancreatic carcinoma for analysis of resistance mechanisms and for definition of sensitising agents. METHODS: TICs were defined by expression patterns of a CD44(+)/CD24(-), CD44(+)/CD24(+) or CD44(+)/CD133(+) phenotype and correlation to growth in immunodeficient mice, differentiation grade, clonogenic growth, sphere formation, aldehyde dehydrogenase (ALDH) activity and therapy resistance. RESULTS: Mechanistically, specific binding of transcriptionally active cRel-containing NF-kappaB complexes in TICs was observed. Sulforaphane prevented NF-kappaB binding, downregulated apoptosis inhibitors and induced apoptosis, together with prevention of clonogenicity. Gemcitabine, the chemopreventive agents resveratrol and wogonin, and the death ligand TRAIL were less effective. In a xenograft model, sulforaphane strongly blocked tumour growth and angiogenesis, while combination with TRAIL had an additive effect without obvious cytotoxicity in normal cells. Freshly isolated patient tumour cells expressing markers for TICs could be sensitised by sulforaphane for TRAIL-induced cytotoxicity. CONCLUSION: The data provide new insights into resistance mechanisms of TICs and suggest the combination of sulforaphane with TRAIL as a promising strategy for targeting of pancreatic TICs.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , NF-kappa B/antagonists & inhibitors , Pancreatic Neoplasms/prevention & control , Thiocyanates/therapeutic use , Animals , Cells, Cultured , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Isothiocyanates , Mice , Mice, Nude , NF-kappa B/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfoxides , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
9.
Br J Cancer ; 99(4): 622-31, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18665180

ABSTRACT

Little is known about the factors that enable the mobilisation of human mesenchymal stem cells (MSC) from the bone marrow into the blood stream and their recruitment to and retention in the tumour. We found specific migration of MSC towards growth factors present in pancreatic tumours, such as PDGF, EGF, VEGF and specific inhibitors Glivec, Erbitux and Avastin interfered with migration. Within a few hours, MSC migrated into spheroids consisting of pancreatic cancer cells, fibroblasts and endothelial cells as measured by time-lapse microscopy. Supernatant from subconfluent MSC increased sprouting of HUVEC due to VEGF production by MSC itself as demonstrated by RT-PCR and ELISA. Only few MSCs were differentiated into endothelial cells in vitro, whereas in vivo differentiation was not observed. Lentiviral GFP-marked MSCs, injected in nude mice xenografted with orthotopic pancreatic tumours, preferentially migrated into the tumours as observed by FACS analysis of green fluorescent cells. By immunofluorescence and intravital microscopic studies, we found the interaction of MSC with the endothelium of blood vessels. Mesenchymal stem cells supported tumour angiogenesis in vivo, that is CD31(+) vessel density was increased after the transfer of MSC compared with siVEGF-MSC. Our data demonstrate the migration of MSC toward tumour vessels and suggest a supportive role in angiogenesis.


Subject(s)
Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/blood supply , Vascular Endothelial Growth Factor A/metabolism , Actins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Benzamides , Bevacizumab , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation , Cells, Cultured , Cetuximab , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Imatinib Mesylate , Lentivirus/genetics , Male , Mesenchymal Stem Cell Transplantation , Mice , Mice, Nude , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Piperazines/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/metabolism , Pyrimidines/pharmacology , Spheroids, Cellular/pathology , Transplantation, Heterologous , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
10.
Cancer Gene Ther ; 15(4): 231-40, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18202717

ABSTRACT

Genetic modification of human bone marrow mesenchymal stem cells (MSC) is highly valuable for their exploitation in basic science and therapeutic applications, for example in cancer. We present here a new, fast and easy-to-use method to enrich a functional population of lentiviral (LV)-transduced MSC expressing enhanced green fluorescent protein (eGFP). We replaced the eGFP gene by a fusion gene of puromycin acetyltransferase and eGFP. Upon LV gene transfer and puromycin selection, we quickly obtained a pure transduced MSC population, in which growth, differentiation capacity and migration preferences were not compromised. Furthermore, we are the first to report the migration velocity of MSC among which 30% were moving and velocity of about 15 mum h(-1) was not altered by LV transduction. Manipulated MSC underwent senescence one passage earlier than non-transduced cells, suggesting the use for therapeutic intervention in early passage numbers. Upon tail vein application in nude mice, the majority of LV-transduced MSC could be detected in human orthotopic pancreatic tumor xenografts and to a minor extent in mouse liver, kidney and lung. Together, LV transduction of genes to MSC followed by puromycin selection is a powerful tool for basic research and improves the therapeutic prospects of MSC as vehicles in gene therapy.


Subject(s)
Lentivirus/genetics , Mesenchymal Stem Cells/cytology , Pancreatic Neoplasms/therapy , Transduction, Genetic , Animals , Cell Differentiation , Cell Line, Tumor , Cell Movement , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/pathology
11.
Cancer Gene Ther ; 14(3): 316-26, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17186015

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent, which selectively induces apoptosis in many transformed cells without apparent toxic side effects in normal tissue. We recently described the construction and characterization of a lentiviral vector for expression of TRAIL. In this report, we evaluate its suitability for therapeutic application. In vitro, we observed specific induction of apoptosis upon transduction in human lung cancer cells. Cell death was partially dependent on successful integration and TRAIL expression by the vectors, but was to some extent mediated by protein carryover, as we found TRAIL protein associated with virus particles. Transduction of subcutaneously growing lung tumors on nude mice with lentiviral TRAIL mediated a transient suppression of tumor growth. Analysis of tumor sections revealed that transduction efficiency of lentiviral control vector but not of lentiviral TRAIL vector was high. This was because of the direct cytotoxic activity of recombinant TRAIL present in viral particles, which prevented efficient tumor transduction. These data therefore suggest that enveloped viral vectors constitutively expressing TRAIL are well suited for ex vivo applications, such as the transduction of tumor-homing cells, but may have a lower effect when used directly for the transduction of tumor cells in vivo.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/therapy , Genetic Therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cells, Cultured , Female , Humans , Kidney/metabolism , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Transduction, Genetic , Tumor Necrosis Factor-alpha/pharmacology , Virion/metabolism
12.
Oncogene ; 26(23): 3352-63, 2007 May 17.
Article in English | MEDLINE | ID: mdl-17130830

ABSTRACT

The reciprocal chromosomal translocation t(4;11) is correlated with infant, childhood, adult and therapy-related high-risk acute leukemia. Here, we investigated the biological effects of MLL.AF4, AF4.MLL or the combination of both reciprocal fusion proteins in a conditional in vitro cell culture model system. Several parameters like cell growth, cell cycling capacity, apoptotic behavior and growth transformation were investigated under physiological and stress conditions. Co-transfected cells displayed the highest resistance against apoptotic triggers, cell cycling capacity and loss-of-contact inhibition. These analyses were complemented by gene expression profiling experiments and specific gene signatures were established for each of the three cell lines. Interestingly, co-transfected cells strongly upregulate the homeobox gene Nanog. In combination with Oct4, the Nanog homeoprotein is steering maintenance of pluripotency and self-renewal in embryonic stem cells. Transcription of Nanog and other stem cell factors, like Oct4 and Bmi1, was verified in biopsy material of t(4;11) patient cells which express both reciprocal t(4;11) fusion genes. In conclusion, the presence of both reciprocal MLL fusion proteins confers biological properties known from t(4;11) leukemia, suggesting that each of the two fusion proteins contribute specific properties and, in combination, also synergistic effects to the leukemic phenotype.


Subject(s)
Apoptosis , Cell Cycle , Chromosomes, Human, Pair 4/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Gene Expression Profiling , Humans , Mice , Mutation/genetics , Polymerase Chain Reaction , Transcription Factors/genetics
13.
Rehabilitation (Stuttg) ; 45(5): 289-98, 2006 Oct.
Article in German | MEDLINE | ID: mdl-17024613

ABSTRACT

BACKGROUND: Urinary incontinence following stroke is an extensive problem for the patients and their relatives that influences the well-being and care in the future. There are a lot of therapeutic interventions available, their effectiveness, however, is not known in detail. For rehabilitation practice the ongoing question is how Urinary Incontinence (UI) can best be treated in a way that the patients daily life is not compromised. METHOD: The search for clinical trials was carried out in PubMed, CINAHL, and Cochrane Library, restricted to German and English papers published between 1989 and April 2005. Medical, nursing and physiotherapeutic interventions for treating UI after stroke were described and analysed. RESULTS: The clinical trials were divided into process-oriented trials and those looking at individual interventions. The process trials could be divided into three different groups with an overall success of 82-95 %, 50-56 % and 23-36 % respectively. Behavioral methods (caregiver-induced, patient-active and other interventions) and medical interventions are available. The studies of the most successful group include staff education and application of interventions based on an assessment procedure and a guideline. No clinical trial on individual interventions reached a result like the process-oriented studies. CONCLUSION: For treating urinary incontinence a multimodal approach is necessary: special education of the nurses, applying and acting in a problem-solving process, for example in the Rehabilitation Cycle and delivering care based on an assessment procedure and guidelines. Development of a guideline for treating urinary incontinence after stroke can be recommended. Further research in the efficacy of individual interventions is needed.


Subject(s)
Practice Guidelines as Topic , Practice Patterns, Physicians'/statistics & numerical data , Quality of Life , Stroke Rehabilitation , Stroke/epidemiology , Urinary Incontinence/rehabilitation , Comorbidity , Humans , Treatment Outcome , Urinary Incontinence/epidemiology
14.
Cell Death Differ ; 13(10): 1740-51, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16470224

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in many transformed cells, suggesting TRAIL as an ideal candidate for cancer gene therapy. A main obstacle in cancer therapy is intrinsic or acquired therapy resistance of malignant cells. To study induction of resistance against TRAIL, we generated lentiviral vectors allowing efficient TRAIL expression and apoptosis induction in a variety of human cancer cell lines. Within days upon TRAIL overexpression, cells became resistant towards TRAIL, but not to CD95 ligation or DNA damage by cisplatin. Cell surface expression of TRAIL receptors 1 and 2 was completely abrogated in resistant cells due to intracellular retention of the receptors by TRAIL. SiRNA directed against TRAIL resensitized the resistant cells by restoring cell surface expression of TRAIL receptors. These findings represent a novel resistance mechanism towards TRAIL, specifically caused by TRAIL overexpression, and question the use of TRAIL expression in tumor-cell targeting gene therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Lentivirus/genetics , Membrane Glycoproteins/genetics , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/genetics , Apoptosis , Apoptosis Regulatory Proteins/antagonists & inhibitors , Base Sequence , Cell Line, Tumor , Cisplatin/pharmacology , Death Domain Receptor Signaling Adaptor Proteins , Drug Resistance, Neoplasm , Endoplasmic Reticulum/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Jurkat Cells , Membrane Glycoproteins/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Transduction, Genetic , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors
15.
Cell Death Differ ; 13(1): 31-40, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16003386

ABSTRACT

The CD95 (Apo-1/Fas)/CD95 ligand (CD95L) system is best characterized as a trigger of apoptosis. Nevertheless, despite broad expression of CD95L and CD95 in the developing brain, absence of functional CD95 (lpr mice) or CD95L (gld mice) does not alter neuronal numbers. Here, we report that in embryonic hippocampal and cortical neurons in vivo and in vitro CD95L does not induce apoptosis. Triggering of CD95 in cultured immature neurons substantially increases neurite branches by promoting their formation. The branching increase occurs in a caspase-independent and death domain-dependent manner and is paralleled by an increase in the nonphosphorylated form of Tau. Most importantly, lpr and gld mutants exhibit a reduced number of dendritic branches in vivo at the time when synapse formation takes place. These data reveal a novel function for the CD95 system and add to the picture of guidance molecules in the developing brain.


Subject(s)
Neurons/cytology , Neurons/physiology , fas Receptor/physiology , Animals , Apoptosis , Caspases/metabolism , Cell Differentiation , Cells, Cultured , Fas Ligand Protein , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/physiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Neurites/ultrastructure , Neuronal Plasticity , Signal Transduction , Transcription Factors/metabolism , Tumor Necrosis Factors/deficiency , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/physiology , fas Receptor/genetics
16.
Cancer Lett ; 242(1): 104-11, 2006 Oct 08.
Article in English | MEDLINE | ID: mdl-16338063

ABSTRACT

The glucocorticoid dexamethasone is frequently used as co-treatment in cytotoxic cancer therapy, e.g. to prevent nausea, to protect normal tissue or for other reasons. While the potent pro-apoptotic properties and the supportive effects of glucocorticoids to tumour therapy in lymphoid cells are well studied, the impact to cytotoxic treatment of colorectal and hepatocellular carcinoma is unknown. We tested apoptosis-induction, viability, tumour growth and protein expression using 8 established cell lines, 18 surgical specimen and a xenograft on nude mice. In the presence of dexamethasone we found strong inhibition of apoptosis in response to 5-FU, cisplatin, gemcitabine or gamma-irradiation, enhanced viability and tumour growth of colorectal and hepatocellular carcinomas. No correlation with age, gender, histology, TNM, the p53 status and induction of therapy resistance by dexamethasone co-treatment could be detected. These data show that glucocorticoid-induced resistance occurs not occasionally but is common in colorectal and hepatocellular carcinomas implicating that the use of glucocorticoids may be harmful for cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Colorectal Neoplasms/drug therapy , Dexamethasone/pharmacology , Liver Neoplasms/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Female , Glucocorticoids/metabolism , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation
17.
Cancer Biol Ther ; 5(1): 59-64, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16294015

ABSTRACT

PURPOSE: Glucocorticoids such as dexamethasone are widely used for medication of urological diseases, e.g., as cotreatment of advanced prostate cancer, to improve appetite, weight loss, fatigue, relieve bone pain, diminish ureteric obstruction, to reduce the production of adrenal androgens, as an antiemetic in patients undergoing chemo- and/or radiotherapy together with serving as "standard" therapy arm in randomized studies. While the potent pro-apoptotic properties and the supportive effects of glucocorticoids to tumor therapy in lymphoid cells are well studied, the impact to growth of prostate and other urological carcinomas is unknown. METHODS: We isolated cells from surgical resections of 21 prostate tumors and measured apoptosis and viability in these primary cells and 17 established cell lines from human prostate, bladder, renal cell and testicular carcinomas. RESULTS: We found that dexamethasone induces resistance regarding exposure to several cytotoxic agents such as taxol, gemcitabine, cisplatin, 5-FU and gamma-irradiation in 86% of the freshly isolated prostate tumors and in 100% of the established urological cell lines. No difference in dexamethasone-mediated protection was found in normal, benign and malignant prostate tumors. CONCLUSIONS: These data show for the first time that dexamethasone induced therapy resistance in urological carcinomas is not the exception but a more common phenomenon and implicate that glucocorticoids may have two faces in cancer therapy, a beneficial and a dangerous one.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Dexamethasone/adverse effects , Drug Resistance, Neoplasm/drug effects , Urologic Neoplasms/therapy , Apoptosis , Female , Humans , Male , Radiation Tolerance/drug effects , Urologic Neoplasms/drug therapy , Urologic Neoplasms/radiotherapy
18.
Br J Cancer ; 92(6): 1084-8, 2005 Mar 28.
Article in English | MEDLINE | ID: mdl-15756274

ABSTRACT

Chemotherapy for lung cancer not only has severe side effects but frequently also exhibits limited, if any clinical effectiveness. Dexamethasone (DEX) and similar glucocorticoids (GCs) such as prednisone are often used in the clinical setting, for example, as cotreatment to prevent nausea and other symptoms. Clinical trials evaluating the impact of GCs on tumour control and patient survival of lung carcinoma have never been performed. Therefore, we isolated cancer cells from resected lung tumour specimens and treated them with cisplatin in the presence or absence of DEX. Cell number of viable and dead cells was evaluated by trypan blue exclusion and viability was measured by the MTT-assay. We found that DEX induced resistance toward cisplatin in all of 10 examined tumour samples. Similar results were found using gemcitabine as cytotoxic drug. Survival of drug-treated lung carcinoma cells in the presence of DEX was longlasting as examined 2 and 3 weeks after cisplatin treatment of a lung carcinoma cell line. These data corroborate recent in vitro and in vivo xenograft findings and rise additional concerns about the widespread combined use of DEX with antineoplastic drugs in the clinical management of patients with lung cancer.


Subject(s)
Cisplatin/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dexamethasone/pharmacology , Lung Neoplasms/drug therapy , Aged , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Gemcitabine
19.
Gene Ther ; 11(6): 534-43, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14999225

ABSTRACT

Gene transfer vectors based on the adeno-associated virus (AAV) are used for various experimental and clinical therapeutic approaches. In the present study, we demonstrate the utility of rAAV as a tumoricidal agent in human colorectal cancer. We constructed an rAAV vector that expresses tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL/Apo2L) and used it to transduce human colorectal cancer cells. TRAIL belongs to the TNF superfamily of cytokines that are involved in various immune responses and apoptotic processes. It has been shown to induce cell death specifically in cancer cells. Transduction with AAV.TRAIL gave rise to rapid expression of TRAIL, followed by induction of apoptosis, which could be inhibited by the caspase inhibitor z-VAD.fmk, in several human colon cancer cell lines. The apoptotic mechanism included activation of caspase-3, as well as cytochrome c release from mitochondria. The outgrowth of human colorectal tumors grown in mice was completely blocked by transduction with AAV.TRAIL in vitro, while in vivo transduction significantly inhibited the growth of established tumors. AAV vectors could provide a safe method of gene delivery and offer a novel method of using TRAIL as a therapeutic protein.


Subject(s)
Colorectal Neoplasms/therapy , Dependovirus/genetics , Genetic Therapy/methods , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Apoptosis , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cells, Cultured , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Gene Expression , Hepatocytes/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , TNF-Related Apoptosis-Inducing Ligand , Transduction, Genetic/methods , Tumor Necrosis Factor-alpha/metabolism
20.
Br J Cancer ; 89(11): 2155-62, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14647152

ABSTRACT

In the present study, we demonstrate the utility of a non-tumour-forming T-cell line for the inducible gene transfer of tumour necrosis factor (TNF)-related apoptosis-inducing ligand (Apo2L/TRAIL), which has been shown to selectively induce apoptosis in malignant but not in normal cells. To generate T cells inducible for TRAIL expression, we stably transfected Jurkat cells with TRAIL in the context of the Tet-On system. The switched on cells strongly expressed TRAIL mRNA, whose protein product was expressed on the cell surface. Paracrine induction of apoptosis in human target tumour cells was solely found for membrane-bound TRAIL. The Jurkat-TRAIL cells itself survived due to clonal selection of TRAIL-resistant cells. Jurkat-TRAIL cells had an additive effect with cytotoxic drugs in vitro, since cell death was enhanced. To elucidate the antitumoral activity of these Jurkat-TRAIL cells in vivo, we injected them intratumorally in xenografts of human Burkitt lymphomas. Switching on expression of TRAIL by adding tetracycline to the drinking water of the mice strongly reduced tumour growth by apoptosis in a caspase-dependent manner. Thus, non-tumour-forming T-cell lines offer a novel method for gene transfer and inducible expression of TRAIL in tumour therapy.


Subject(s)
Burkitt Lymphoma/therapy , Gene Transfer Techniques , Lymphoma, B-Cell/therapy , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Apoptosis , Apoptosis Regulatory Proteins , Burkitt Lymphoma/pathology , Cell Line, Tumor , Humans , Jurkat Cells , Lymphoma, B-Cell/pathology , Mice , Mice, Nude , Neoplasm Transplantation , T-Lymphocytes/transplantation , TNF-Related Apoptosis-Inducing Ligand , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...