Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 5(6): 1621-34, 2014 Mar 30.
Article in English | MEDLINE | ID: mdl-24742583

ABSTRACT

The extreme aggressiveness of pancreatic ductal adenocarcinoma (PDA) has been associated with blocked gap junctional intercellular communication (GJIC) and the presence of cancer stem cells (CSCs). We examined whether disturbed GJIC is responsible for a CSC phenotype in established and primary cancer cells and patient tissue of PDA using interdisciplinary methods based in physiology, cell and molecular biology, histology and epigenetics. Flux of fluorescent dyes and gemcitabine through gap junctions (GJs) was intact in less aggressive cells but not in highly malignant cells with morphological dysfunctional GJs. Among several connexins, only Cx43 was expressed on the cell surface of less aggressive and GJIC-competent cells, whereas Cx43 surface expression was absent in highly malignant, E-cadherin-negative and GJIC-incompetent cells. The levels of total Cx43 protein and Cx43 phosphorylated at Ser368 and Ser279/282 were high in normal tissue but low to absent in malignant tissue. si-RNA-mediated inhibition of Cx43 expression in GJIC-competent cells prevented GJIC and induced colony formation and the expression of stem cell-related factors. The bioactive substance sulforaphane enhanced Cx43 and E-cadherin levels, inhibited the CSC markers c-Met and CD133, improved the functional morphology of GJs and enhanced GJIC. Sulforaphane altered the phosphorylation of several kinases and their substrates and inhibition of GSK3, JNK and PKC prevented sulforaphane-induced CX43 expression. The sulforaphane-mediated expression of Cx43 was not correlated with enhanced Cx43 RNA expression, acetylated histone binding and Cx43 promoter de-methylation, suggesting that posttranslational phosphorylation is the dominant regulatory mechanism. Together, the absence of Cx43 prevents GJIC and enhances aggressiveness, whereas sulforaphane counteracts this process, and our findings highlight dietary co-treatment as a viable treatment option for PDA.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Cell Communication/drug effects , Connexin 43/metabolism , Gap Junctions/drug effects , Isothiocyanates/pharmacology , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Cell Proliferation/drug effects , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Gap Junctions/metabolism , Gap Junctions/pathology , Humans , Immunoenzyme Techniques , Microscopy, Electron , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Sulfoxides , Tumor Cells, Cultured , Gemcitabine
2.
Int J Cancer ; 134(10): 2489-503, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24615157

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal malignancies characterized by an intense tumor stroma with hypoperfused regions, a significant inflammatory response and pronounced therapy resistance. New therapeutic agents are urgently needed. The plant-derived agent triptolide also known as "thunder god vine" has a long history in traditional Chinese medicine for treatment of rheumatoid arthritis and cancer and is now in a clinical phase II trial for establishing the efficacy against a placebo. The authors mimicked the situation in patient tumors by induction of hypoxia in experimental models of pancreatic cancer stem cells (CSCs) and evaluated the therapeutic effect of triptolide. Hypoxia led to induction of colony and spheroid formation, aldehyde dehydrogenase 1 (ALDH1) and NF-κB activity, migratory potential and a switch in morphology to a fibroblastoid phenotype, as well as stem cell- and epithelial-mesenchymal transition-associated protein expression. Triptolide efficiently inhibited hypoxia-induced transcriptional signaling and downregulated epithelial-mesenchymal transition (EMT) and CSC features in established highly malignant cell lines, whereas sensitive cancer cells or nonmalignant cells were less affected. In vivo triptolide inhibited tumor take and tumor growth. In primary CSCs isolated from patient tumors, triptolide downregulated markers of CSCs, proliferation and mesenchymal cells along with upregulation of markers for apoptosis and epithelial cells. This study is the first to show that triptolide reverses EMT and CSC characteristics and therefore may be superior to current chemotherapeutics for treatment of PDA.


Subject(s)
Diterpenes/pharmacology , Epithelial-Mesenchymal Transition/drug effects , NF-kappa B/metabolism , Pancreatic Neoplasms/prevention & control , Phenanthrenes/pharmacology , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents, Alkylating/pharmacology , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/prevention & control , Cell Hypoxia , Cell Line, Tumor , Cell Movement/drug effects , Chick Embryo , Down-Regulation/drug effects , Epoxy Compounds/pharmacology , Humans , Isoenzymes/metabolism , Mice , Mice, Inbred Strains , Mice, Nude , NF-kappa B/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/metabolism , RNA Interference , Retinal Dehydrogenase/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays
3.
Int J Oncol ; 44(5): 1470-80, 2014 May.
Article in English | MEDLINE | ID: mdl-24626333

ABSTRACT

Advanced androgen-independent prostate cancer (AIPC) is an aggressive malignancy with a poor prognosis. Apoptosis-resistant cancer stem cells (CSCs) have been identified in AIPC and are not eliminated by current therapeutics. Novel therapeutic options, which are currently being evaluated in patient studies, include TRAIL and the broccoli-derived isothiocyanate sulforaphane. Although neither agent targets normal cells, TRAIL induces apoptosis in most cancer cells, and sulforaphane eliminates CSCs. In this study, the established AIPC cell lines DU145 and PC3, with enriched CSC features, and primary patient-derived prostate CSCs were treated with sulforaphane and recombinant soluble TRAIL. We examined the effects of these drugs on NF-κB activity, self-renewal and differentiation potential, and stem cell signaling via spheroid- and colony-forming assays, FACS and western blot analyses, immunohistochemistry, and an antibody protein array in vitro and after xenotransplantation. We largely found a stronger effect of sulforaphane on CSC properties compared to TRAIL, though the agents acted synergistically when applied in combination. This was associated with the inhibition of TRAIL-induced NF-κB binding; CXCR4, Jagged1, Notch 1, SOX 2, and Nanog expression; ALDH1 activity inhibition; and the elimination of differentiation and self-renewal potential. In vivo, tumor engraftment and tumor growth were strongly inhibited, without the induction of liver necrosis or other obvious side effects. These findings suggest that sulforaphane shifts the balance from TRAIL-induced survival signals to apoptosis and thus explains the observed synergistic effect. A nutritional strategy for high sulforaphane intake may target the cancer-specific activity of TRAIL in CSCs.


Subject(s)
Antineoplastic Agents/pharmacology , Isothiocyanates/pharmacology , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Chick Embryo , Drug Synergism , Humans , Male , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Sulfoxides , Xenograft Model Antitumor Assays , NF-kappaB-Inducing Kinase
4.
J Pathol ; 227(3): 325-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22262369

ABSTRACT

Involvement of dysregulated autophagy in cancer growth and progression has been shown in different tumour entities, including pancreatic ductal adenocarcinoma (PDA). PDA is an extremely aggressive tumour characterized by a small population of highly therapy-resistant cancer stem cells (CSCs) capable of self-renewal and migration. We examined whether autophagy might be involved in the survival of CSCs despite nutrition and oxygen deprivation typical for the hypoxic tumour microenvironment of PDA. Immunohistochemistry revealed that markers for hypoxia, CSCs and autophagy are co-expressed in patient-derived tissue of PDA. Hypoxia starvation (H/S) enhanced clonogenic survival and migration of established pancreatic cancer cells with stem-like properties (CSC(high)), while pancreatic tumour cells with fewer stem cell markers (CSC(low)) did not survive these conditions. Electron microscopy revealed more advanced autophagic vesicles in CSC(high) cells, which exhibited higher expression of autophagy-related genes under normoxic conditions and relative to CSC(low) cells, as found by RT-PCR and western blot analysis. LC3 was already fully converted to the active LC3-II form in both cell lines, as evaluated by western blot and detection of accumulated GFP-LC3 protein by fluorescence microscopy. H/S increased formation of autophagic and acid vesicles, as well as expression of autophagy-related genes, to a higher extent in CSC(high) cells. Modulation of autophagy by inhibitors and activators resensitized CSC(high) to apoptosis and diminished clonogenicity, spheroid formation, expression of CSC-related genes, migratory activity and tumourigenicity in mice. Our data suggest that enhanced autophagy levels may enable survival of CSC(high) cells under H/S. Interference with autophagy-activating or -inhibiting drugs disturbs the fine-tuned physiological balance of enhanced autophagy in CSC and switches survival signalling to suicide.


Subject(s)
Autophagy , Carcinoma, Pancreatic Ductal/pathology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Electron , Microscopy, Fluorescence , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/ultrastructure , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/ultrastructure , Polymerase Chain Reaction , Time Factors , Tumor Burden
5.
Mol Ther ; 19(1): 188-95, 2011 01.
Article in English | MEDLINE | ID: mdl-20940707

ABSTRACT

Despite intense efforts to develop treatments against pancreatic cancer, agents that cure this highly resistant and metastasizing disease are not available. Considerable attention has focused on broccoli compound sulforaphane (SF), which is suggested as combination therapy for targeting of pancreatic cancer stem cells (CSCs). However, there are concerns that antioxidative properties of SF may interfere with cytotoxic drugs-as suggested, e.g., for vitamins. Therefore we investigated a combination therapy using established pancreatic CSCs. Although cisplatin (CIS), gemcitabine (GEM), doxorubicin, 5-flurouracil, or SF effectively induced apoptosis and prevented viability, combination of a drug with SF increased toxicity. Similarly, SF potentiated the drug effect in established prostate CSCs revealing that SF enhances drug cytotoxicity also in other tumor entities. Most importantly, combined treatment intensified inhibition of clonogenicity and spheroid formation and aldehyde dehydrogenase 1 (ALDH1) activity along with Notch-1 and c-Rel expression indicating that CSC characteristics are targeted. In vivo, combination treatment was most effective and totally abolished growth of CSC xenografts and tumor-initiating potential. No pronounced side effects were observed in normal cells or mice. Our data suggest that SF increases the effectiveness of various cytotoxic drugs against CSCs without inducing additional toxicity in mice.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Thiocyanates/pharmacology , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , Female , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Isothiocyanates , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/pathology , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-rel/antagonists & inhibitors , Proto-Oncogene Proteins c-rel/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Retinal Dehydrogenase , Spheroids, Cellular , Sulfoxides , Tumor Stem Cell Assay/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...