Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Cell Death Dis ; 14(1): 3, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596765

ABSTRACT

The emergence of resistance to systemic therapies in pancreatic ductal adenocarcinoma (PDAC) is still a major obstacle in clinical practice. Both, constitutive and inducible NF-κB activity are known as key players in this context. To identify differentially expressed and TRAIL resistance mediating NF-κB target genes, TRAIL sensitive and resistant PDAC cell lines were analyzed by transcriptome assays. In this context, A20 was identified as an NF-κB/RelA inducible target gene. Translational PDAC tissue analysis confirmed the correlation of elevated A20 protein expression with activated RelA expression in PDAC patients. In in vitro experiments, an elevated A20 expression is accompanied by a specific resistance toward TRAIL-mediated apoptosis but not to chemotherapeutic-induced cell death. This TRAIL resistance was attributed to A20´s E3-ligase activity-mediating Zink finger domain. Furthermore, the ubiquitin-binding scaffold protein p62 was identified as indispensable for the TRAIL-mediated apoptosis-inducing pathway affected by A20. The results of this study identify A20 as a possible therapeutic target to affect resistance to TRAIL-induced apoptosis in PDAC cells.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , NF-kappa B/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Apoptosis , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factor RelA/genetics , Pancreatic Neoplasms
2.
Cells ; 11(21)2022 11 04.
Article in English | MEDLINE | ID: mdl-36359900

ABSTRACT

Obesity and obesity-associated diseases represent one of the key health challenges of our time. In this context, aberrant hepatic lipid accumulation is a central pathological aspect of non-alcoholic fatty liver disease (NAFLD). By comparing methylation signatures of liver biopsies before and after bariatric surgery, we recently demonstrated the strong enrichment of differentially methylated heat shock factor 1 (HSF1) binding sites (>400-fold) in the process of liver remodeling, indicating a crucial role of HSF1 in modulating central aspects of NAFLD pathogenesis. Using cellular models of NAFLD, we were able to show that HSF1 is activated during fat accumulation in hepatocytes, mimicking conditions in patients before bariatric surgery. This induction was abolished by starving the cells, mimicking the situation after bariatric surgery. Regarding this connection, carnitine palmitoyltransferase 1 isoform A (CTP1a), a central regulator of lipid beta-oxidation, was identified as a HSF1 target gene by promoter analysis and HSF1 knockdown experiments. Finally, pharmacological activation of HSF1 through celastrol reduced fat accumulation in the cells in a HSF1-dependent manner. In conclusion, we were able to confirm the relevance of HSF1 activity and described a functional HSF1-CPT1a pathway in NAFLD pathogenesis.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/pathology , Lipid Metabolism/genetics , Obesity/metabolism , Lipids
3.
Int J Mol Sci ; 19(6)2018 Jun 04.
Article in English | MEDLINE | ID: mdl-29867042

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignant neoplasms and registers rising death rates in western countries. Due to its late detection in advanced stages, its extremely aggressive nature and the minimal effectiveness of currently available therapies, PDAC is a challenging problem in the clinical field. One characteristic of PDAC is a distinct desmoplasia consisting of fibroblasts, endothelial and immune cells as well as non-cellular components, contributing to therapy resistance. It is well established that the NF-κB signaling pathway controls inflammation, cancer progression and apoptosis resistance in PDAC. This study attempts to identify NF-κB target genes mediating therapy resistance of humane PDAC cell lines towards death ligand induced apoptosis. By using a genome wide unbiased approach the chemokine CX3CL1 was established as a central NF-κB target gene mediating therapy resistance. While no direct impact of CX3CL1 expression on cancer cell apoptosis was identified in co-culture assays it became apparent that CX3CL1 is acting in a paracrine fashion, leading to an increased recruitment of inflammatory cells. These inflammatory cells in turn mediate apoptosis resistance of PDAC cells. Therefore, our data dissect a bifunctional cross-signaling pathway in PDAC between tumor and immune cells giving rise to therapy resistance.


Subject(s)
Apoptosis , Carcinoma, Pancreatic Ductal/metabolism , Chemokine CX3CL1/metabolism , NF-kappa B/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/physiopathology , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Chemokine CX3CL1/immunology , Humans
4.
Biochim Biophys Acta Mol Cell Res ; 1864(5): 782-796, 2017 May.
Article in English | MEDLINE | ID: mdl-28188806

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest cancers. From a clinical view, the transcription factor NF-κB is of particular importance, since this pathway confers apoptosis resistance and limits drug efficacy. Whereas the role of the most abundant NF-κB subunit p65/RelA in therapeutic resistance is well documented, only little knowledge of the RelA downstream targets and their functional relevance in TRAIL mediated apoptosis in PDAC is available. In the present study TRAIL resistant and sensitive PDAC cell lines were analyzed for differentially expressed RelA target genes, to define RelA downstream targets mediating TRAIL resistance. The most upregulated target gene was then further functionally characterized. Unbiased genome-wide expression analysis demonstrated that the chemokine CCL20 represents the strongest TRAIL inducible direct RelA target gene in resistant PDAC cells. Unexpectedly, targeting CCL20 by siRNA, blocking antibodies or by downregulation of the sole CCL20 receptor CCR6 had no effect on PDAC cell death or cancer cell migration, arguing against an autocrine role of CCL20 in PDAC. However, by using an ex vivo indirect co-culture system we were able to show that CCL20 acts paracrine to recruit immune cells. Importantly, CCL20-recruited immune cells further increase TRAIL resistance of CCL20-producing PDAC cells. In conclusion, our data show a functional role of a RelA-CCL20 pathway in PDAC TRAIL resistance. We demonstrate how the therapy-induced cross-talk of cancer cells with immune cells affects treatment responses, knowledge needed to tailor novel bi-specific treatments, which target tumor cell as well as immune cells.


Subject(s)
Carcinoma, Pancreatic Ductal , Chemokine CCL20/physiology , Chemotaxis, Leukocyte/genetics , Drug Resistance, Neoplasm/genetics , Pancreatic Neoplasms , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Adult , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cells, Cultured , Chemokine CCL20/antagonists & inhibitors , Chemokine CCL20/genetics , Chemotaxis, Leukocyte/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Humans , Lymphocytes/drug effects , Lymphocytes/physiology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , RNA, Small Interfering/pharmacology
5.
Biochim Biophys Acta ; 1783(5): 941-52, 2008 May.
Article in English | MEDLINE | ID: mdl-18191642

ABSTRACT

The early response gene IEX-1 plays a complex role in the regulation of apoptosis. Depending on the cellular context and the apoptotic stimulus, IEX-1 is capable to either enhance or suppress apoptosis. To further dissect the molecular mechanisms involved in the modulation of apoptosis by IEX-1, we analysed the molecular crosstalk between IEX-1 and the NF-kappaB pathway. Using GST-pulldown assays, a direct interaction of IEX-1 with the C-terminal region of the subunit RelA/p65 harbouring the transactivation domain of the NF-kappaB transcription factor was shown. This interaction negatively regulates RelA/p65 dependent transactivation as shown by GAL4-and luciferase assay and was confirmed for the endogenous proteins by co-immunoprecipitation experiments. Using deletion constructs, we were able to map the C-terminal region of IEX-1 as the critical determinant of the interaction with RelA/p65. We could further show, that IEX-1 mediated NF-kappaB inhibition accounts for the reduced expression of the anti-apoptotic NF-kappaB target genes Bcl-2, Bcl-xL, cIAP1 and cIAP2, thereby sensitizing cells for apoptotic stimuli. Finally, ChIP-assays revealed that IEX-1 associates with the promoter of these genes. Altogether, our findings suggest a critical role of IEX-1 in the NF-kappaB dependent regulation of apoptotic responses.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/genetics , Membrane Proteins/metabolism , Transcription Factor RelA/antagonists & inhibitors , Transcriptional Activation , Apoptosis Regulatory Proteins/chemistry , Binding Sites , Cell Line , Cell Nucleus/metabolism , HeLa Cells , Humans , Membrane Proteins/chemistry , Promoter Regions, Genetic , Signal Transduction , Transcription Factor RelA/chemistry , Transcription Factor RelA/metabolism
6.
Biochem J ; 402(2): 367-75, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17107344

ABSTRACT

The stress response gene IEX-1 (immediate early gene-X-1) is involved in the regulation of cell growth and cellular viability. To some extent, these effects include an interference with the proteasomal turnover of certain regulatory proteins. Here, we show that IEX-1 directly attenuates the activity and formation of the 26 S proteasome in HEK-293 cells (human embryonic kidney cells). We further demonstrate that IEX-1 reduces the overall expression levels of certain protein components of the 19 S proteasomal subunit such as S5a/Rpn10 and S1/Rpn2, whereas the expression of other proteasomal proteins was less or not affected. In contrast with direct apoptotic stimuli, such as the anti-cancer drug etoposide, leading to caspase-dependent degradation of S1 and S5a, the effect of IEX-1 is independent of proteolytic cleavage of these proteins. Furthermore, the decreasing effect of IEX-1 on S5a and S1 expression is still seen in the presence of cycloheximide, but not in the presence of actinomycin D, and quantitative real-time PCR revealed lower mRNA levels of S5a and S1 in IEX-1-overexpressing cells, suggesting an interference of IEX-1 with the gene transcription of S5a and S1. Additionally, luciferase assays confirmed an interference of IEX-1 with the activity of the S5a promoter. These findings indicate a role of IEX-1 in the maintenance and assembly of the 26 S proteasome, obviously involving an altered gene expression of certain proteasomal proteins. Thereby, IEX-1 may essentially modulate signalling pathways related to 26 S proteasome activity and involved in cellular growth control and apoptosis.


Subject(s)
Immediate-Early Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cell Line , Gene Expression Regulation , Hexosyltransferases , Humans , Immediate-Early Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Promoter Regions, Genetic/genetics , Proteasome Endopeptidase Complex/genetics , Protein Biosynthesis , RNA-Binding Proteins , Transcription, Genetic/genetics
7.
J Biol Chem ; 280(26): 24849-56, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15855159

ABSTRACT

Immediate early gene X1 (IEX-1) represents a stress response gene involved in growth control and modulation of apoptosis. Here, we report a detailed analysis of IEX-1 with respect to its intracellular localization. By means of confocal laser scanning microscopy, a green fluorescent protein-IEX-1 fusion protein transfected into HeLa cells, as well as endogenous IEX-1, could be detected in distinct subnuclear structures. This particular subnuclear localization of IEX-1 was not observed with a green fluorescent protein-IEX-1 fusion protein lacking a putative nuclear localization sequence, along with a decreased effect on apoptosis. Double immunofluorescence staining revealed a partial co-localization of endogenous promyelocytic leukemia protein (PML) and IEX-1 in these subnuclear structures. Nuclear localization of IEX-1 is also enhanced upon treatment of cells with leptomycin B, an inhibitor of the nuclear exporter CRM1. These observations indicate that IEX-1 is specifically shuttled to and from the nucleus. Overexpression experiments using PML isoforms III and IV revealed distinct intranuclear interaction of IEX-1 and PML. Coprecipitation experiments showed physical interaction between IEX-1 and PML. The close structural relation of IEX-1-containing nuclear subdomains and PML nuclear bodies suggests a function of IEX-1 related to the multiple functions of these unique subnuclear regions, particularly during stress response and growth control.


Subject(s)
Cell Nucleus/metabolism , Genes, Immediate-Early , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Active Transport, Cell Nucleus , Apoptosis , Apoptosis Regulatory Proteins , Blotting, Western , Caspases/metabolism , Fatty Acids, Unsaturated/pharmacology , Glutathione Transferase/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoprecipitation , Membrane Proteins , Microscopy, Confocal , Microscopy, Fluorescence , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Protein Binding , Protein Isoforms , Recombinant Fusion Proteins/chemistry , Transcription Factors/metabolism , Transfection , Tumor Suppressor Proteins/metabolism
8.
Med Klin (Munich) ; 99(4): 185-90, 2004 Apr 15.
Article in German | MEDLINE | ID: mdl-15085288

ABSTRACT

BACKGROUND AND PURPOSE: The pancreatic ductal adenocarcinoma (PDAC) is characterized by a highly malignant phenotype and a profound chemoresistance. Thus, options for an effective treatment of this disease are still quite poor. In this study, it was investigated whether the autocrine secretion of interleukin-(IL-)1beta is related to a chemoresistant phenotype of PDAC cells in vivo. MATERIAL AND METHODS: Human PancTu1 PDAC cells were inoculated subcutaneously into female SCID mice. After 10 days of outgrowth, animals were randomized and left untreated or treated with an IL-1beta-RI antibody, etoposide, or a combination of both. After treatment for 14 days, tumor sizes were determined and each tumor was analyzed immunohistochemically for apoptosis (TUNEL), activated NF-kappaB (p65), and vascularization (CD31 staining). RESULTS: The combination of IL-1beta-RI antibody and etoposide led to a significantly reduced outgrowth of PancTu1 tumors in comparison to the monotherapies or no treatment. Accordingly, the number of apoptotic cells was significantly elevated in tumors of the combination group. After treatment with the IL-1beta-RI antibody, less activated NF-kappaB was present in tumors compared to the control group. Moreover, tumors of the combination group showed a clearly reduced vascularization. CONCLUSION: The autocrine secretion of IL-1beta contributes to a constitutively increased NF-kappaB activity in PDAC cells along with a chemoresistant phenotype.


Subject(s)
Autocrine Communication/physiology , Carcinoma, Pancreatic Ductal/pathology , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/physiology , Interleukin-1/antagonists & inhibitors , Interleukin-1/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/pathology , Sialoglycoproteins/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Stem Cell Assay , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Etoposide/pharmacology , Female , Humans , In Situ Nick-End Labeling , Interleukin 1 Receptor Antagonist Protein , Mice , Mice, SCID , Neoplasm Transplantation , Recombinant Proteins/pharmacology , Tumor Cells, Cultured/pathology
9.
Int J Cancer ; 104(4): 469-76, 2003 Apr 20.
Article in English | MEDLINE | ID: mdl-12584745

ABSTRACT

Sulfasalazine is commonly used as an anti inflammatory agent and is known as a potent inhibitor of NF-kappaB. Some pancreatic carcinomas are characterized by a constitutively elevated NF-kappaB activity accounting for chemoresistance. To elucidate whether blockade of NF-kappaB activity with sulfasalazine is suitable for overcoming this chemoresistance in vivo, we employed a mouse model with subcutaneously xenotransplanted human Capan-1 pancreatic carcinoma cells. Fourteen days upon tumor inoculation, animals were randomized in 6 groups, receiving no treatment, treatment with gemcitabine or with etoposide, either alone or in combination with sulfasalazine, or with sulfasalazine alone. Two therapy regimens were given with a 7-day interval in between. Upon treatment with etoposide or gemcitabine alone, tumor sizes were moderately reduced to 65-68% and 50-65%, respectively, as compared to untreated tumors. Sulfasalazine alone only decreased temporarily the tumor sizes. Sulfasalazine in combination with gemcitabine showed only partially higher reduction in tumor sizes than gemcitabine alone, whereas the combination with etoposide reduced significantly the tumor sizes in all experiments (down to 20%). TUNEL-staining showed higher numbers of apoptotic cells in tumors from the combination groups, in particular with etoposide, and proliferation as indicated by Ki67 staining was strongly reduced. Furthermore, combined treatment of sulfasalazine with the cytostatic drugs led to a decreased blood vessel density. Immunohistochemical staining of the activated p65 subunit showed that sulfasalazine treatment abolished the basal NF-kappaB activity in tumor xenografts. These data imply that the well established anti-inflammatory drug sulfasalazine sensitizes pancreatic carcinoma cells to anti cancer drugs, in particular to etoposide in vivo by inhibition of NF-kappaB. This combined chemotherapy offers great potential for improved anti-tumor responses in pancreatic carcinomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , NF-kappa B/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Sulfasalazine/therapeutic use , Animals , Apoptosis/drug effects , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Etoposide/pharmacology , Female , Humans , Mice , Mice, SCID , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...