Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Pancreatology ; 21(5): 912-919, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33824054

ABSTRACT

BACKGROUND: Oncogenic Kras initiates and drives carcinogenesis in the pancreas by complex signaling networks, including activation of the NFκB pathway. Although recent evidence has shown that oncogenic gains in Nfκb2 collaborate with Kras in the carcinogenesis, no data at the level of genetics for the contribution of Nfκb2 is available so far. METHODS: We used Nfkb2 knock-out mice to decipher the role of the gene in Kras-driven carcinogenesis in vivo. RESULTS: We show that the Nfkb2 gene is needed for cancer initiation and progression in KrasG12D-driven models and this requirement of Nfkb2 is mechanistically connected to proliferative pathways. In contrast, Nfκb2 is dispensable in aggressive pancreatic ductal adenocarcinoma (PDAC) models relying on the simultaneous expression of the Kras oncogene and the mutated tumor suppressor p53. CONCLUSIONS: Our data add to the understanding of context-dependent requirements of oncogenic Kras signaling during pancreatic carcinogenesis.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Genes, ras , Mice , Pancreas , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics
2.
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
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.
J Biol Chem ; 289(4): 1917-29, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24311782

ABSTRACT

Although nuclear factor E2-related factor-2 (Nrf2) protects from carcinogen-induced tumorigenesis, underlying the rationale for using Nrf2 inducers in chemoprevention, this antioxidative transcription factor may also act as a proto-oncogene. Thus, an enhanced Nrf2 activity promotes formation and chemoresistance of colon cancer. One mechanism causing persistent Nrf2 activation is the adaptation of epithelial cells to oxidative stress during chronic inflammation, e.g. colonocytes in inflammatory bowel diseases, and the multifunctional stress response gene immediate early response-3 (IER3) has a crucial role under these conditions. We now demonstrate that colonic tissue from Ier3(-/-) mice subject of dextran sodium sulfate colitis exhibit greater Nrf2 activity than Ier3(+/+) mice, manifesting as increased nuclear Nrf2 protein level and Nrf2 target gene expression. Likewise, human NCM460 colonocytes subjected to shRNA-mediated IER3 knockdown exhibit greater Nrf2 activity compared with control cells, whereas IER3 overexpression attenuated Nrf2 activation. IER3-deficient NCM460 cells exhibited reduced reactive oxygen species levels, indicating increased antioxidative protection, as well as lower sensitivity to TRAIL or anticancer drug-induced apoptosis and greater clonogenicity. Knockdown of Nrf2 expression reversed these IER3-dependent effects. Further, the enhancing effect of IER3 deficiency on Nrf2 activity relates to the control of the inhibitory tyrosine kinase Fyn by the PI3K/Akt pathway. Thus, the PI3K inhibitor LY294002 or knockdown of Akt or Fyn expression abrogated the impact of IER3 deficiency on Nrf2 activity. In conclusion, the interference of IER3 with the PI3K/Akt-Fyn pathway represents a novel mechanism of Nrf2 regulation that may get lost in tumors and by which IER3 exerts its stress-adaptive and tumor-suppressive activity.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Colitis/metabolism , Colon/metabolism , Epithelial Cells/metabolism , Immediate-Early Proteins/metabolism , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line , Chromones/pharmacology , Colitis/chemically induced , Colitis/genetics , Colon/pathology , Dextran Sulfate/toxicity , Enzyme Inhibitors/pharmacology , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Immediate-Early Proteins/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Morpholines/pharmacology , NF-E2-Related Factor 2/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 304(7): G646-54, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23370678

ABSTRACT

Although a profound barrier dysfunction has been reported, little is known about the pathophysiological mechanism evoking gastrointestinal graft-vs.-host disease (GI-GvHD) and apparent therapeutic options. The aim of this study was to evaluate the influence of oral glutamine on the course of GI-GvHD in an acute semiallogenic graft-vs.-host disease (GvHD) in irradiated B6D2F1 mice. An acute semiallogenic GvHD was induced by intraperitoneal injection of lymphocytes from C57BL/6 mice to irradiated B6D2F1 mice. Half of the GvHD animals received oral glutamine supplementation for 6 days started at the time of lymphocyte transfer. Six days after induction of the semiallogenic GvHD, jejunum specimens were prepared. The expression of the proinflammatory cytokine TNF-α and the tight junction protein occludin was investigated by PCR. Histological changes along with the apoptotic response were evaluated and intestinal permeability was assessed. Animals with GvHD showed a strong increase in paracellular permeability as a sign of the disturbed barrier function. TNF-α expression was significantly increased and the expression of the tight junction protein occludin decreased. GvHD led to mucosal atrophy, crypt hyperplasia, crypt apoptosis, and a disintegration of the tight junctions. Glutamine-treated mice showed reduced expression of TNF-α, increased occludin expression, fewer histological changes in the jejunum, smaller number of apoptotic cells in the crypt, and reduced gastrointestinal permeability. In conclusion, oral glutamine seems to have beneficial effects on the severity of inflammatory changes in the course of GvHD and might be a therapeutic option.


Subject(s)
Glutamine/therapeutic use , Graft vs Host Disease/physiopathology , Animals , Disease Models, Animal , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Jejunum/drug effects , Jejunum/metabolism , Jejunum/pathology , Lymphocyte Transfusion/adverse effects , Mice , Occludin/biosynthesis , Permeability/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
6.
Antioxidants (Basel) ; 12(10)2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37891897

ABSTRACT

Metabolic compartmentalization of stroma-rich tumors, like pancreatic ductal adenocarcinoma (PDAC), greatly contributes to malignancy. This involves cancer cells importing lactate from the microenvironment (reverse Warburg cells) through monocarboxylate transporter-1 (MCT1) along with substantial phenotype alterations. Here, we report that the reverse Warburg phenotype of PDAC cells compensated for the shortage of glutamine as an essential metabolite for redox homeostasis. Thus, oxidative stress caused by glutamine depletion led to an Nrf2-dependent induction of MCT1 expression in pancreatic T3M4 and A818-6 cells. Moreover, greater MCT1 expression was detected in glutamine-scarce regions within tumor tissues from PDAC patients. MCT1-driven lactate uptake supported the neutralization of reactive oxygen species excessively produced under glutamine shortage and the resulting drop in glutathione levels that were restored by the imported lactate. Consequently, PDAC cells showed greater survival and growth under glutamine depletion when utilizing lactate through MCT1. Likewise, the glutamine uptake inhibitor V9302 and glutaminase-1 inhibitor CB839 induced oxidative stress in PDAC cells, along with cell death and cell cycle arrest that were again compensated by MCT1 upregulation and forced lactate uptake. Our findings show a novel mechanism by which PDAC cells adapt their metabolism to glutamine scarcity and by which they develop resistance against anticancer treatments based on glutamine uptake/metabolism inhibition.

7.
Eur J Intern Med ; 111: 54-62, 2023 05.
Article in English | MEDLINE | ID: mdl-36797118

ABSTRACT

INTRODUCTION: High-power short-duration ablation (HPSD) is an effective therapy for atrial fibrillation with thermal esophageal injury as a rare but relevant side effect. AIM AND METHODS: In this retrospective single-center analysis we evaluated the incidence and relevance of ablation-induced findings and the prevalence of ablation-independent incidental gastrointestinal findings. For 15 months all patients undergoing ablation were screened by postablation esophagogastroduodenoscopy. Pathological findings were followed up and treated if necessary. RESULTS: 286 consecutive patients (66±10 years; 54.9% male) were included. 19.6% of patients showed ablation-associated alterations (10.8% esophageal lesions, 10.8% gastroparesis, 1.7% both findings). Logistic multivariable regression analysis confirmed an influence of lower BMI on the occurrence of RFA-associated endoscopic findings (OR 0.936, 95% CI 0.878-0.997, p<0.05). 48.3% of patients demonstrated incidental gastrointestinal findings. In 1.0% neoplastic lesions were present, 9.4% showed precancerous lesions and in 4.2% neoplastic lesions of unknown dignity were found requiring further diagnostics or therapy. 18.1% of patients demonstrated findings associated with a potentially increased risk of bleeding under anticoagulation. Patients with clinically relevant incidental findings were significantly more often male, 68.8% vs. 49.5% (p<0.01). CONCLUSION: HPSD ablation is safe, no devasting complication occurred in any patient. It resulted in 19.6% ablation-induced thermal injury whereas incidental findings of the upper GI tract were found in 48.3% of patients. Due to the high prevalence of 14.7% of findings requiring further diagnostics, therapy, or surveillance in a cohort that is mimicking the general population, screening endoscopy of the upper GI tract seems to be reasonable in the general population.


Subject(s)
Atrial Fibrillation , Catheter Ablation , Humans , Male , Female , Atrial Fibrillation/diagnosis , Atrial Fibrillation/epidemiology , Atrial Fibrillation/surgery , Retrospective Studies , Prevalence , Esophagus/pathology , Endoscopy, Gastrointestinal , Catheter Ablation/adverse effects , Catheter Ablation/methods , Treatment Outcome
8.
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
9.
Carcinogenesis ; 33(1): 84-93, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22095073

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) and chronic pancreatitis, representing one risk factor for PDAC, are characterized by a marked desmoplasia enriched of pancreatic myofibroblasts (PMFs). Thus, PMFs are thought to essentially promote pancreatic tumorigenesis. We recently demonstrated that the adhesion molecule L1CAM is involved in epithelial-mesenchymal transition of PMF-cocultured H6c7 human ductal epithelial cells and that L1CAM is expressed already in ductal structures of chronic pancreatitis with even higher elevation in primary tumors and metastases of PDAC patients. This study aimed at investigating whether PMFs and L1CAM drive malignant transformation of pancreatic ductal epithelial cells by enhancing their tumorigenic potential. Cell culture experiments demonstrated that in the presence of PMFs, H6c7 cells exhibit a profound resistance against death ligand-induced apoptosis. This apoptosis protection was similarly observed in H6c7 cells stably overexpressing L1CAM. Intrapancreatic inoculation of H6c7 cells together with PMFs (H6c7co) resulted in tumor formation in 7/8 and liver metastases in 6/8 severe combined immunodeficiency (SCID) mice, whereas no tumors and metastases were detectable after inoculation of H6c7 cells alone. Likewise, tumor outgrowth and metastases resulted from inoculation of L1CAM-overexpressing H6c7 cells in 5/7 and 3/7 SCID mice, respectively, but not from inoculation of mock-transfected H6c7 cells. Treatment of H6c7co tumor-bearing mice with the L1CAM antibody L1-9.3/2a inhibited tumor formation and liver metastasis in 100 and 50%, respectively, of the treated animals. Overall, these data provide new insights into the mechanisms of how PMFs and L1CAM contribute to malignant transformation of pancreatic ductal epithelial cells in early stages of pancreatic tumorigenesis.


Subject(s)
Carcinoma, Pancreatic Ductal/etiology , Myofibroblasts/physiology , Neural Cell Adhesion Molecule L1/physiology , Pancreatic Neoplasms/etiology , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Liver Neoplasms/secondary , Mice , Mice, SCID , Pancreatic Neoplasms/pathology
10.
J Biol Chem ; 286(47): 40911-21, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21990354

ABSTRACT

Adaptation of epithelial cells to persistent oxidative stress plays an important role in inflammation-associated carcinogenesis. This adaptation process involves activation of Nrf2 (nuclear factor-E2-related factor-2), which has been recently shown to contribute to carcinogenesis through the induction of proteasomal gene expression and proteasome activity. To verify this possible link between inflammation, oxidative stress, and Nrf2-dependent proteasome activation, we explored the impact of inflammatory (M1) macrophages on the human colon epithelial cell line NCM460. Transwell cocultures with macrophages differentiated from granulocyte monocyte-colony-stimulating factor-treated monocytes led to an increased activity of Nrf2 in NCM460 cells along with an elevated proteasome activity. This higher proteasome activity resulted from Nrf2-dependent induction of proteasomal gene expression, as shown for the 19 and 20 S subunit proteins S5a and α5, respectively. These effects of macrophage coculture were preceded by an increase of reactive oxygen species in cocultured NCM460 cells and could be blocked by catalase or by the reactive oxygen species scavenger Tiron, whereas transient treatment of NCM460 cells with H(2)O(2) similarly led to Nrf2-dependent proteasome activation. Through the Nrf2-dependent increase of proteasomal gene expression and proteasome activity, the sensitivity of NCM460 cells to tumor necrosis factor-related apoptosis-inducing ligand- or irinotecan-induced apoptosis declined. These findings indicate that inflammatory conditions such as the presence of M1 macrophages and the resulting oxidative stress are involved in the Nrf2-dependent gain of proteasome activity in epithelial cells, e.g. colonocytes, giving rise of greater resistance to apoptosis. This mechanism might contribute to inflammation-associated carcinogenesis, e.g. of the colon.


Subject(s)
Apoptosis , Colon/cytology , Colon/metabolism , Macrophages/cytology , NF-E2-Related Factor 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Apoptosis/drug effects , Apoptosis/immunology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line , Coculture Techniques , Colon/drug effects , Colon/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intracellular Space/drug effects , Intracellular Space/metabolism , Irinotecan , Macrophages/metabolism , Proteasome Endopeptidase Complex/genetics , Reactive Oxygen Species/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology
11.
Cancers (Basel) ; 14(22)2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36428576

ABSTRACT

Tumor-related death is primarily caused by metastasis; consequently, understanding, preventing, and treating metastasis is essential to improving clinical outcomes. Metastasis is mainly governed by the dissemination of tumor cells in the systemic circulation: so-called circulating tumor cells (CTCs). CTCs typically arise from epithelial tumor cells that undergo epithelial-to-mesenchymal transition (EMT), resulting in the loss of cell-cell adhesions and polarity, and the reorganization of the cytoskeleton. Various oncogenic factors can induce EMT, among them the transforming growth factor (TGF)-ß, as well as Wnt and Notch signaling pathways. This entails the activation of numerous transcription factors, including ZEB, TWIST, and Snail proteins, acting as transcriptional repressors of epithelial markers, such as E-cadherin and inducers of mesenchymal markers such as vimentin. These genetic and phenotypic changes ultimately facilitate cancer cell migration. However, to successfully form distant metastases, CTCs must primarily withstand the hostile environment of circulation. This includes adaption to shear stress, avoiding being trapped by coagulation and surviving attacks of the immune system. Several applications of CTCs, from cancer diagnosis and screening to monitoring and even guided therapy, seek their way into clinical practice. This review describes the process leading to tumor metastasis, from the generation of CTCs in primary tumors to their dissemination into distant organs, as well as the importance of subtyping CTCs to improve personalized and targeted cancer therapy.

12.
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
13.
BMC Mol Biol ; 11: 64, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20799950

ABSTRACT

BACKGROUND: The L1 cell adhesion molecule (L1CAM) was originally identified as a neural adhesion molecule involved in axon guidance. In many human epithelial carcinomas L1CAM is overexpressed and thereby augments cell motility, invasion and metastasis formation. L1CAM positive carcinomas are associated with bad prognosis. Recent data point out that L1CAM is regulated in a fashion similar to epithelial-mesenchymal transition (EMT). Previous studies have implied the transcription factors Slug and/or ß-catenin in L1CAM transcriptional regulation. However, the regulation of human L1CAM expression at the transcriptional level is not well understood. RESULTS: To better understand the molecular basis of L1CAM transcriptional regulation, we carried out a detailed characterization of the human L1CAM promoter. We identified two transcription start sites, the first in front of a non-translated exon 0 (promoter 1) and the other next to the first protein-coding exon 1 (promoter 2). Both sites could be verified in endometrial carcinoma (EC) cell lines and appear to be used in a cell-type specific manner. The two identified promoter regions showed activity in luciferase reporter assays. Chromatin-IP analyses confirmed the in silico predicted E-boxes, binding sites for transcription factors Snail and Slug, as well as Lef-1 sites, which are related to ß-catenin-mediated transcriptional regulation, in both promoters. Overexpression of ß-catenin exclusively augmented activity of promoter 1 whereas Slug enhanced promoter 1 and 2 activity suggesting that both promoters can be active. Overexpression of ß-catenin or Slug could upregulate L1CAM expression in a cell-type specific manner. CONCLUSIONS: Our results, for the first time, provide evidence that the L1CAM gene has two functionally active promoter sites that are used in a cell-type specific manner. Slug and ß-catenin are involved L1CAM transcriptional regulation. Nevertheless, Slug rather than ß-catenin levels are correlated with L1CAM expression in EC cell lines. Our findings suggest that the L1CAM transcriptional regulation is more complex than anticipated and this study provides the basis for a better understanding of L1CAM regulation in non-neuronal/tumor cells.


Subject(s)
Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neural Cell Adhesion Molecule L1/metabolism , Promoter Regions, Genetic , Animals , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Humans , Neural Cell Adhesion Molecule L1/genetics , RNA, Messenger/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Initiation Site , beta Catenin/genetics , beta Catenin/metabolism
14.
Cells ; 9(11)2020 10 25.
Article in English | MEDLINE | ID: mdl-33113836

ABSTRACT

With a five-year survival rate under 9%, pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest tumors. Although the treatment options are slightly improving, PDAC is the second leading cause of cancer related death in 2020 in the US. In addition to a pronounced desmoplastic stroma reaction, pancreatic cancer is characterized by one of the lowest levels of oxygen availability within the tumor mass and these hypoxic conditions are known to contribute to tumor development and progression. In this context, the major hypoxia associated transcription factor family, HIF, regulates hundreds of genes involved in angiogenesis, metabolism, migration, invasion, immune escape and therapy resistance. Current research implications show, that hypoxia also modulates diverse areas of epigenetic mechanisms like non-coding RNAs, histone modifications or DNA methylation, which cooperate with the hypoxia-induced transcription factors as well as directly regulate the hypoxic response pathways. In this review, we will focus on hypoxia-mediated epigenetic alterations and their impact on pancreatic cancer.


Subject(s)
Epigenesis, Genetic , Hypoxia/genetics , Hypoxia/metabolism , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA Methylation , Disease Susceptibility , Energy Metabolism , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms/pathology
15.
Int J Oncol ; 34(1): 243-53, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19082495

ABSTRACT

We recently showed that the adhesion molecule L1CAM (CD171) is overexpressed in pancreatic adenocarcinoma (PDAC) essentially contributing to chemoresistance of PDAC cells. In search of the mechanisms of this effect we now identified alpha5-integrin as the L1CAM ligand being essential for L1CAM-mediated chemoresistance of these highly malignant tumor cells. Thus, blockade or knock-down of alpha5-integrin in the L1CAM expressing PDAC cell lines PT45-P1res, Colo357 and Panc1 increased anti-cancer drug sensitivity. In line with the previously reported NO-dependent caspase inhibition resulting from L1CAM induced iNOS expression, the loss of chemoresistance upon alpha5-integrin inhibition was preceded by decreased iNOS expression and enhanced caspase-3/-7 activation. Accordingly, the loss of anti-cancer drug protection by alpha5-integrin inhibition could be overcome by administration of the NO-donor SNAP. Moreover, the gain of chemoresistance of parental PT45-P1 cells when transfected with L1CAM was abrogated by alpha5-integrin inhibition, whereas transfection of PT45-P1 cells with an integrin binding-deficient L1CAM mutant (L1mutRGE) did neither induce chemoresistance or iNOS expression nor conferred sensitivity to alpha5-integrin inhibition as seen upon transfection with wild-type L1CAM. Thus, mutational loss of the integrin binding site in the L1CAM molecule or the blockade of alpha5-integrin abolished the induction of iNOS expression and chemoresistance by L1CAM, indicating that both a functional L1CAM and alpha5-integrin are indispensable of L1CAM-induced drug resistance in PDAC cells.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Drug Resistance, Neoplasm , Integrin alpha5/physiology , Neural Cell Adhesion Molecule L1/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Caspases/metabolism , Etoposide/therapeutic use , Flow Cytometry , Humans , Mutagenesis, Site-Directed , Neural Cell Adhesion Molecule L1/genetics , Nitric Oxide Synthase Type II/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
16.
Cancers (Basel) ; 11(12)2019 Dec 09.
Article in English | MEDLINE | ID: mdl-31835312

ABSTRACT

This series of 10 articles (four original articles, six reviews) is presented by international leaders in the field of NF-κB signaling in cancer and inflammation [...].

17.
Cancers (Basel) ; 11(10)2019 Sep 26.
Article in English | MEDLINE | ID: mdl-31561620

ABSTRACT

Pancreatic cancer is one of the carcinomas with the worst prognoses, as shown by its five-year survival rate of 9%. Although there have been new therapeutic innovations, the effectiveness of these therapies is still limited, resulting in pancreatic ductal adenocarcinoma (PDAC) becoming the second leading cause of cancer-related death in 2020 in the US. In addition to tumor cell intrinsic resistance mechanisms, this disease exhibits a complex stroma consisting of fibroblasts, immune cells, neuronal and vascular cells, along with extracellular matrix, all conferring therapeutic resistance by several mechanisms. The NF-κB pathway is involved in both the tumor cell-intrinsic and microenvironment-mediated therapeutic resistance by regulating the transcription of a plethora of target genes. These genes are involved in nearly all scenarios described as the hallmarks of cancer. In addition to classical regulators of apoptosis, NF-κB regulates the expression of chemokines and their receptors, both in the tumor cells and in cells of the microenvironment. These chemokines mediate autocrine and paracrine loops among tumor cells but also cross-signaling between tumor cells and the stroma. In this review, we will focus on NF-κB-mediated chemokine signaling, with an emphasis on therapy resistance in pancreatic cancer.

19.
Nat Commun ; 8(1): 2063, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234056

ABSTRACT

FOXO3 is consistently annotated as a human longevity gene. However, functional variants and underlying mechanisms for the association remain unknown. Here, we perform resequencing of the FOXO3 locus and single-nucleotide variant (SNV) genotyping in three European populations. We find two FOXO3 SNVs, rs12206094 and rs4946935, to be most significantly associated with longevity and further characterize them functionally. We experimentally validate the in silico predicted allele-dependent binding of transcription factors (CTCF, SRF) to the SNVs. Specifically, in luciferase reporter assays, the longevity alleles of both variants show considerable enhancer activities that are reversed by IGF-1 treatment. An eQTL database search reveals that the alleles are also associated with higher FOXO3 mRNA expression in various human tissues, which is in line with observations in long-lived model organisms. In summary, we present experimental evidence for a functional link between common intronic variants in FOXO3 and human longevity.


Subject(s)
Forkhead Box Protein O3/physiology , Longevity/genetics , Polymorphism, Single Nucleotide/genetics , White People/genetics , Age Factors , Aged , Aged, 80 and over , Alleles , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Computer Simulation , Female , Forkhead Box Protein O3/genetics , Haplotypes/genetics , Humans , Insulin-Like Growth Factor I/metabolism , Introns/genetics , Male , Middle Aged , RNA, Messenger/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL