Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 163: 196-209, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33359260

ABSTRACT

The idea to use megadoses of ascorbate (vitamin C) for cancer treatment has recently been revived. Despite clear efficacy in animal experimentation, our understanding of the cellular and molecular mechanisms of this treatment is still limited and suggests a combined oxidative and metabolic mechanism behind the selective cytotoxicity of ascorbate towards cancerous cells. To gain more insight into the cellular effects of high doses of ascorbate, we performed a detailed analysis of metabolic changes and cell survival of both luminal and basal-like breast cancer cells treated with ascorbate and revealed a distinctive metabolic shift virtually reversing the Warburg effect and triggering a severe disruption of redox homeostasis. High doses of ascorbate were cytotoxic against MCF7 and MDA-MB231 cells representing luminal and basal-like breast cancer phenotypes. Cell death was dependent on ascorbate-induced oxidative stress and accumulation of ROS, DNA damage, and depletion of essential intracellular co-factors including NAD+/NADH, associated with a multifaceted metabolic rewiring. This included a sharp disruption of glycolysis at the triose phosphate level, a rapid drop in ATP levels, and redirection of metabolites toward lipid droplet accumulation and increased metabolites and enzymatic activity in the pentose phosphate pathway (PPP). High doses of ascorbate also inhibited the TCA cycle and increased oxygen consumption. Together the severe disruptions of the intracellular metabolic homeostasis on multiple levels "redox crisis and energetic catastrophe" consequently trigger a rapid irreversible cell death.


Subject(s)
Breast Neoplasms , Animals , Ascorbic Acid/pharmacology , Breast Neoplasms/drug therapy , Cell Survival , Energy Metabolism , Female , Humans , Oxidation-Reduction , Oxidative Stress
2.
Commun Biol ; 3: 10, 2020.
Article in English | MEDLINE | ID: mdl-31909202

ABSTRACT

Gold compounds have a long history of use as immunosuppressants, but their precise mechanism of action is not completely understood. Using our recently developed liver-on-a-chip platform we now show that gold compounds containing planar N-heterocyclic carbene (NHC) ligands are potent ligands for the aryl hydrocarbon receptor (AHR). Further studies showed that the lead compound (MC3) activates TGFß1 signaling and suppresses CD4+ T-cell activation in vitro, in human and mouse T cells. Conversely, genetic knockdown or chemical inhibition of AHR activity or of TGFß1-SMAD-mediated signaling offsets the MC3-mediated immunosuppression. In scurfy mice, a mouse model of human immunodysregulation polyendocrinopathy enteropathy X-linked syndrome, MC3 treatment reduced autoimmune phenotypes and extended lifespan from 24 to 58 days. Our findings suggest that the immunosuppressive activity of gold compounds can be improved by introducing planar NHC ligands to activate the AHR-associated immunosuppressive pathway, thus expanding their potential clinical application for autoimmune diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Immunosuppression Therapy/methods , Organogold Compounds/immunology , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/drug effects , Transforming Growth Factor beta1/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival/drug effects , Hep G2 Cells , Humans , Male , Mice , Receptors, Aryl Hydrocarbon/metabolism , Transforming Growth Factor beta1/metabolism
3.
Sci Rep ; 9(1): 4616, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30874583

ABSTRACT

Organ-on-chip platforms provide models that allow the representation of human physiological processes in cell-based miniaturized systems. Potential pre-clinical applications include drug testing and toxicity studies. Here we describe the use of a multi-compartment micro-fluidic chip to recapitulate hepatic vitamin D metabolism (vitamin D to 25-hydroxyvitamin D) and renal bio-activation (25-hydroxyvitamin D to 1,25-dihydroxyvitamin D) in humans. In contrast to cultivation in conventional tissue culture settings, on-chip cultivation of HepG2 and RPTEC cells in interconnected chambers, used to mimic the liver and kidneys, respectively, resulted in the enhanced expression of vitamin D metabolizing enzymes (CYP2R1, CYP27B1 and CYP24A1). Pump-driven flow of vitamin D3-containing medium through the microfluidic chip produced eluate containing vitamin D3 metabolites. LC-MSMS showed a strong accumulation of 25-hydroxyvitamin D. The chip eluate induced the expression of differentiation markers in HL-60 (acute myeloid leukemia) cells, assessed by qPCR and FACS analysis, in a manner similar to treatment with reference standards indicating the presence of fully activated 1,25 dihydroxyvitamin D, although the latter was not detected in the eluate by LC-MSMS. Interestingly, 25-hydroxyvitamin D by itself led to weak activation of HL-60 cells suggesting that 25-hydroxyvitamin D is also an active metabolite. Our experiments demonstrate that complex metabolic interactions can be reconstructed outside the human body using dedicated organ-on-chip platforms. We therefore propose that such systems may be used to mimic the in vivo metabolism of various micronutrients and xenobiotics.


Subject(s)
Cholecalciferol/metabolism , Kidney/metabolism , Liver/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Activation, Metabolic/physiology , Animals , Cell Line , Cytochrome P-450 Enzyme System/metabolism , HL-60 Cells , Hep G2 Cells , Humans , Lab-On-A-Chip Devices , Microchip Analytical Procedures/methods , Microfluidic Analytical Techniques/methods , Microfluidics/methods , Vitamin D/analogs & derivatives , Vitamin D/metabolism , Vitamins/metabolism
4.
Commun Biol ; 1: 210, 2018.
Article in English | MEDLINE | ID: mdl-30511023

ABSTRACT

Cytotoxic activities of several Golgi-dispersing compounds including AMF-26/M-COPA, brefeldin A and golgicide A have previously been shown to induce autophagy or apoptosis. Here, we demonstrate that these Golgi disruptors also trigger ferroptosis, a non-apoptotic form of cell death characterized by iron-dependent oxidative degradation of lipids. Inhibitors of ferroptosis not only counteract cell death, but they also protect from Golgi dispersal and inhibition of protein secretion in response to several Golgi stress agents. Furthermore, the application of sublethal doses of ferroptosis-inducers such as erastin and sorafenib, low cystine growth conditions, or genetic knockdown of SLC7A11 and GPX4 all similarly protect cells from Golgi stress and lead to modulation of ACSL4, SLC7A5, SLC7A11 or GPX4 levels. Collectively, this study suggests a previously unrecognized function of the Golgi apparatus, which involves cellular redox control and prevents ferroptotic cell death.

5.
Int J Mol Sci ; 19(12)2018 Dec 09.
Article in English | MEDLINE | ID: mdl-30544880

ABSTRACT

The p38 MAPK pathway is known to influence the anti-tumor effects of several chemotherapeutics, including that of organometallic drugs. Previous studies have demonstrated the important role of p38 both as a regulator and a sensor of cellular reactive oxygen species (ROS) levels. Investigating the anti-cancer properties of novel 1,8-naphthalimide derivatives containing Rh(I) and Ru(II) N-heterocyclic carbene (NHC) ligands, we observed a profound induction of ROS by the complexes, which is most likely generated from mitochondria (mtROS). Further analyses revealed a rapid and consistent activation of p38 signaling by the naphthalimide-NHC conjugates, with the Ru(II) analogue-termed MC6-showing the strongest effect. In view of this, genetic as well as pharmacological inhibition of p38α, attenuated the anti-proliferative and pro-apoptotic effects of MC6 in HCT116 colon cancer cells, highlighting the involvement of this signaling molecule in the compound's toxicity. Furthermore, the influence of MC6 on p38 signaling appeared to be dependent on ROS levels as treatment with general- and mitochondria-targeted anti-oxidants abrogated p38 activation in response to MC6 as well as the molecule's cytotoxic- and apoptogenic response in HCT116 cells. Altogether, our results provide new insight into the molecular mechanisms of naphthalimide-metal NHC analogues via the ROS-induced activation of p38 MAPK, which may have therapeutic interest for the treatment of various cancer types.


Subject(s)
Apoptosis/drug effects , Colorectal Neoplasms/pathology , Heterocyclic Compounds/pharmacology , Methane/analogs & derivatives , Naphthalimides/pharmacology , Reactive Oxygen Species/metabolism , Ruthenium/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Heterocyclic Compounds/chemistry , Humans , Ligands , Membrane Potential, Mitochondrial/drug effects , Methane/chemistry , Methane/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Naphthalimides/chemistry , Ruthenium/chemistry , Signal Transduction/drug effects
6.
Cancer Metab ; 6: 11, 2018.
Article in English | MEDLINE | ID: mdl-30181873

ABSTRACT

BACKGROUND: We have previously identified 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], the bioactive form of vitamin D3, as a potent regulator of energy-utilization and nutrient-sensing pathways in prostate cancer cells. In the current study, we investigated the effects of 1,25(OH)2D3 on breast cancer (BCa) cell metabolism using cell lines representing distinct molecular subtypes, luminal (MCF-7 and T-47D), and triple-negative BCa (MDA-MB-231, MDA-MB-468, and HCC-1143). METHODS: 1,25(OH)2D3's effect on BCa cell metabolism was evaluated by employing a combination of real-time measurements of glycolysis/oxygen consumption rates using a biosensor chip system, GC/MS-based metabolomics, gene expression analysis, and assessment of overall energy levels. The influence of treatment on energy-related signaling molecules was investigated by immunoblotting. RESULTS: We show that 1,25(OH)2D3 significantly induces the expression and activity of the pentose phosphate pathway enzyme glucose-6-phosphate dehydrogenase (G6PD) in all BCa cell lines, however differentially influences glycolytic and respiratory rates in the same cells. Although 1,25(OH)2D3 treatment was found to induce seemingly anti-oxidant responses in MCF-7 cells, such as increased intracellular serine levels, and reduce the expression of its putative target gene thioredoxin-interacting protein (TXNIP), intracellular reactive oxygen species levels were found to be elevated. Serine accumulation in 1,25(OH)2D3-treated cells was not found to hamper the efficacy of chemotherapeutics, including 5-fluorouracil. Detailed analyses of the nature of TXNIP's regulation by 1,25(OH)2D3 included genetic and pharmacological inhibition of signaling molecules and metabolic enzymes including AMP-activated protein kinase and G6PD, as well as by studying the ITCH (E3 ubiquitin ligase)-TXNIP interaction. While these investigations demonstrated minimal involvement of such pathways in the observed non-canonical regulation of TXNIP, inhibition of estrogen receptor (ER) signaling by tamoxifen mirrored the reduction of TXNIP levels by 1,25(OH)2D3, demonstrating that the latter's negative regulation of ER expression is a potential mechanism of TXNIP modulation. CONCLUSIONS: Altogether, we propose that regulation of energy metabolism contributes to 1,25(OH)2D3's anti-cancer effects and that combining 1,25(OH)2D3 with drugs targeting metabolic networks in tumor cells may lead to synergistic effects.

7.
ACS Biomater Sci Eng ; 4(1): 78-89, 2018 Jan 08.
Article in English | MEDLINE | ID: mdl-33418680

ABSTRACT

Advances in organ-on-chip technologies for the application in in vitro drug development provide an attractive alternative approach to replace ethically controversial animal testing and to establish a basis for accelerated drug development. In recent years, various chip-based tissue culture systems have been developed, which are mostly optimized for cultivation of one single cell type or organoid structure and lack the representation of multi organ interactions. Here we present an optimized microfluidic chip design consisting of interconnected compartments, which provides the possibility to mimic the exchange between different organ specific cell types and enables to study interdependent cellular responses between organs and demonstrate that such tandem system can greatly improve the reproducibility and efficiency of toxicity studies. In a simplified liver-kidney-on-chip model, we showed that hepatic cells that grow in microfluidic conditions abundantly and stably expressed metabolism-related biomarkers. Moreover, we applied this system for investigating the biotransformation and toxicity of Aflatoxin B1 (AFB1) and Benzoalphapyrene (BαP), as well as the interaction with other chemicals. The results clearly demonstrate that the toxicity and metabolic response to drugs can be evaluated in a flow-dependent manner within our system, supporting the importance of advanced interconnected multiorgans in microfluidic devices for application in in vitro toxicity testing and as optimized tissue culture systems for in vitro drug screening.

8.
Arch Toxicol ; 92(3): 1133-1149, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29209748

ABSTRACT

The metabolic activity of hepatocytes is a central prerequisite for drug activity and a key element in drug-drug interaction. This central role in metabolism largely depends on the activity of the cytochrome P450 (CYP450) enzyme family, which is not only dependent on liver cell maturation but is also controlled in response to drug and chemical exposure. Here, we report the use of VividDye fluorogenic CYP450 substrates to directly measure and continuously monitor metabolic activity in living hepatocytes. We observed time- and dose-dependent correlation in response to established and putative CYP450 inducers acting through the aryl hydrocarbon receptor and drug combinations. Using repetitive addition of VividDye fluorogenic substrate on a daily basis, we demonstrated the new application of VividDye for monitoring the maturation and dedifferentiation of hepatic cells. Despite a lack of high specificity for individual CYP450 isoenzymes, our approach enables continuous monitoring of metabolic activity in living cells with no need to disrupt cultivation. Our assay can be integrated in in vitro liver-mimetic models for on-line monitoring and thus should enhance the reliability of these tissue model systems.


Subject(s)
Biological Assay/methods , Chromogenic Compounds/metabolism , Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/enzymology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biological Assay/instrumentation , Cell Differentiation/drug effects , Cells, Cultured , Cytochrome P-450 Enzyme System/analysis , Enzyme Induction/drug effects , Fluorescent Dyes/analysis , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/physiology , Humans , Indoles/pharmacology , Lab-On-A-Chip Devices , Male , Mice, Inbred C57BL , Oximes , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
9.
J Pediatr Gastroenterol Nutr ; 66(2): e28-e35, 2018 02.
Article in English | MEDLINE | ID: mdl-29095348

ABSTRACT

OBJECTIVES: Di(2-ethylhexyl) phthalate (DEHP) is a plasticizer used in many polyvinylchloride medical devices and is washed out easily. Thereby critically ill infants can become exposed to DEHP concentrations significantly exceeding the recommended threshold. We suspect DEHP to play an important role in the development of intestinal failure-associated liver disease. The aim of this study was therefore to determine the direct influence of DEHP on different liver cell types. METHODS: HepG2, human upcyte hepatocytes, primary murine hepatocytes, LX-2, human upcyte hepatic stellate cells, and liver organoids were cultured with DEHP (0.5-500 µmol/L) and parameters including cytotoxicity, cell-cell interactions, and expression of metabolizing enzymes were investigated. RESULTS: DEHP modulated the expression of xenobiotic metabolizing enzymes, reduced the formation of bile canaliculi and cell polarity, and inhibited Cyp-activity in hepatocytes. DEHP had a toxic effect on LX-2 and induced the fibrogenic activation of hepatic stellate cells. The mode of action of DEHP was different in monolayer cultures compared to 3D-liver organoids, which were more sensitive to DEHP. CONCLUSIONS: This study suggests that DEHP modulates expression and activity of drug-detoxifying liver enzymes in humans at a clinically relevant concentration. Furthermore, it may contribute to the development of cholestasis and fibrosis. These findings strongly support the opinion, that there is a significant potential for serious adverse effects of DEHP derived from medical devices on human health, especially in very young infants with immature livers.


Subject(s)
Cholestasis/chemically induced , Diethylhexyl Phthalate/pharmacology , Liver/drug effects , Animals , Cell Culture Techniques , Diethylhexyl Phthalate/adverse effects , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Liver/cytology , Liver/pathology , Mice , Real-Time Polymerase Chain Reaction
10.
PLoS One ; 12(4): e0174912, 2017.
Article in English | MEDLINE | ID: mdl-28426762

ABSTRACT

PURPOSE: Oct4 was reported to be one of the most important pluripotency transcription factors in the biology of stem cells including cancer stem cells, and progressed malignant cells. Here we report the investigation of gene expression control of Oct4 by selected human microRNAs and the physiological effect of Oct4 silencing in invasive cancer cells. METHODS AND RESULTS: High throughput luciferase activity assay revealed the microRNA-299-3p to be the most effective in reducing gene expression of Oct4, which was confirmed by Western blot analysis and Oct4 promoter activity in a target luciferase assay. Furthermore, it could be demonstrated that downregulation of Oct4 by microRNAs-299-3p in breast cancer and fibrosarcoma cells lead to a decreased invasiveness in a microfluidic chip assay. Additionally, microRNA-299-3p causes apoptosis in cancer cells. Comparison with Oct4 specific siRNA transfection confirmed that this effect is primary due to the blockade of Oct4 expression. CONCLUSION: The results suggest that microRNA-299-3p is an interesting target for potential clinical use. It may be able to decrease invasive behaviour of carcinoma cells; or even kill these cells by causing apoptosis.


Subject(s)
Apoptosis , Down-Regulation , MicroRNAs/physiology , Neoplasm Invasiveness/genetics , Octamer Transcription Factor-3/metabolism , 3' Untranslated Regions , Breast Neoplasms/pathology , Cell Line, Tumor , Cloning, Molecular , Female , Genetic Vectors , Homologous Recombination , Humans
11.
Biofabrication ; 9(1): 015014, 2017 Feb 03.
Article in English | MEDLINE | ID: mdl-28155845

ABSTRACT

The design and fabrication of a very large-scale liver-lobule (VLSLL)-on-a-chip device, providing a microphysiological niche for hepatocytes, is described. The device consists of an integrated network of liver-lobule-like hexagonal tissue-culture chambers constructed in a hybrid layout with a separate seed-feed network. As a key feature, each chamber contains a central outlet mimicking the central vein of a liver lobule. Separating chamber walls located between the culture area and feed network protects cells from the shear force of the convective flow. Arrays of designated passages convey nutrients to the cells by diffusion-dominated mass transport. We simulated the flow velocity, shear stress and diffusion of glucose molecules inside and outside the culture chambers under a continuous flow rate of 1 µl min-1. As proof of concept, human hepatocellular carcinoma cells (HepG2) were cultured for periods of 5 and 14 days and human-induced pluripotent stem cell (hiPSC)-derived hepatocytes for 21 days. Stabilized albumin secretion and urea synthesis were observed in the microfluidic devices and cells maintained morphology and functionality during the culture period. Furthermore, we observed 3D tissue-like structure and bile-canaliculi network formation in the chips. Future applications of the described platform include drug development and toxicity studies, as well as the modeling of patient-specific liver diseases, and integration in multi-organ human-on-a-chip systems.


Subject(s)
Cell Culture Techniques/methods , Lab-On-A-Chip Devices , Serum Albumin/analysis , Cell Culture Techniques/instrumentation , Cell Line , Diffusion , Dimethylpolysiloxanes/chemistry , Enzyme-Linked Immunosorbent Assay , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Serum Albumin/metabolism , Urea/metabolism
12.
Mol Oncol ; 10(6): 806-24, 2016 06.
Article in English | MEDLINE | ID: mdl-26887594

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) clinically has a very poor prognosis. No small molecule is available to reliably achieve cures. Meisoindigo is chemically related to the natural product indirubin and showed substantial efficiency in clinical chemotherapy for CML in China. However, its effect on PDAC is still unknown. Our results showed strong anti-proliferation effect of meisoindigo on gemcitabine-resistant PDACs. Using a recently established primary PDAC cell line, called Jopaca-1 with a larger CSCs population as model, we observed a reduction of CD133+ and ESA+/CD44+/CD24+ populations upon treatment and concomitantly a decreased expression of CSC-associated genes, and reduced cellular mobility and sphere formation. Investigating basic cellular metabolic responses, we detected lower oxygen consumption and glucose uptake, while intracellular ROS levels increased. This was effectively neutralized by the addition of antioxidants, indicating an essential role of the cellular redox balance. Further analysis on energy metabolism related signaling revealed that meisoindigo inhibited LKB1, but activated AMPK. Both of them were involved in cellular apoptosis. Additional in situ hybridization in tissue sections of PDAC patients reproducibly demonstrated co-expression and -localization of LKB1 and CD133 in malignant areas. Finally, we detected that CD133+/CD44+ were more vulnerable to meisoindigo, which could be mimicked by LKB1 siRNAs. Our results provide the first evidence, to our knowledge, that LKB1 sustains the CSC population in PDACs and demonstrate a clear benefit of meisoindigo in treatment of gemcitabine-resistant cells. This novel mechanism may provide a promising new treatment option for PDAC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Neoplastic Stem Cells/drug effects , Pancreas/drug effects , Pancreatic Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , AMP-Activated Protein Kinase Kinases , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Humans , Indoles/pharmacology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Gemcitabine , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...