Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Cell Physiol Biochem ; 36(3): 852-65, 2015.
Article in English | MEDLINE | ID: mdl-26044490

ABSTRACT

BACKGROUND/AIMS: Elevated levels of non-esterified fatty acids (NEFAs) are under suspicion to mediate ß-cell dysfunction and ß-cell loss in type 2 diabetes, a phenomenon known as lipotoxicity. Whereas saturated fatty acids show a strong cytotoxic effect upon insulin-producing cells, unsaturated fatty acids are not toxic and can even prevent toxicity. Experimental evidence suggests that oxidative stress mediates lipotoxicity and there is evidence that the subcellular site of ROS formation is the peroxisome. However, the interaction between unsaturated and saturated NEFAs in this process is unclear. METHODS: Toxicity of rat insulin-producing cells after NEFA incubation was measured by MTT and caspase assays. NEFA induced H2O2 formation was quantified by organelle specific expression of the H2O2 specific fluorescence sensor protein HyPer. RESULTS: The saturated NEFA palmitic acid had a significant toxic effect on the viability of rat insulin-producing cells. Unsaturated NEFAs with carbon chain lengths >14 showed, irrespective of the number of double bonds, a pronounced protection against palmitic acid induced toxicity. Palmitic acid induced H2O2 formation in the peroxisomes of insulin-producing cells. Oleic acid incubation led to lipid droplet formation, but in contrast to palmitic acid induced neither an ER stress response nor peroxisomal H2O2 generation. Furthermore, oleic acid prevented palmitic acid induced H2O2 production in the peroxisomes. CONCLUSION: Thus unsaturated NEFAs prevent deleterious hydrogen peroxide generation during peroxisomal ß-oxidation of long-chain saturated NEFAs in rat insulin-producing cells.


Subject(s)
Hydrogen Peroxide/metabolism , Insulin-Secreting Cells/drug effects , Oleic Acid/pharmacology , Palmitic Acid/toxicity , Peroxisomes/drug effects , Animals , Biological Assay , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Hydrogen Peroxide/antagonists & inhibitors , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Male , Palmitic Acid/antagonists & inhibitors , Peroxisomes/metabolism , Primary Cell Culture , Rats , Rats, Inbred Lew
2.
Metabolites ; 12(8)2022 Aug 08.
Article in English | MEDLINE | ID: mdl-36005604

ABSTRACT

Mammalian INDY (mINDY, NaCT, gene symbol SLC13A5) is a potential target for the treatment of metabolically associated fatty liver disease (MAFLD). This study evaluated the effects of a selective, cross-species active, non-competitive, non-substrate-like inhibitor of NaCT. First, the small molecule inhibitor ETG-5773 was evaluated for citrate and succinate uptake and fatty acid synthesis in cell lines expressing both human NaCT and mouse Nact. Once its suitability was established, the inhibitor was evaluated in a diet-induced obesity (DIO) mouse model. DIO mice treated with 15 mg/kg compound ETG-5773 twice daily for 28 days had reduced body weight, fasting blood glucose, and insulin, and improved glucose tolerance. Liver triglycerides were significantly reduced, and body composition was improved by reducing fat mass, supported by a significant reduction in the expression of genes for lipogenesis such as SREBF1 and SCD1. Most of these effects were also evident after a seven-day treatment with the same dose. Further mechanistic investigation in the seven-day study showed increased plasma ß-hydroxybutyrate and activated hepatic adenosine monophosphate-activated protein kinase (AMPK), reflecting findings from Indy (-/-) knockout mice. These results suggest that the inhibitor ETG-5773 blocked citrate uptake mediated by mouse and human NaCT to reduce liver steatosis and body fat and improve glucose regulation, proving the concept of NaCT inhibition as a future liver treatment for MAFLD.

3.
Free Radic Res ; 45(5): 501-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21332429

ABSTRACT

Hydrogen peroxide is an important mediator in cell signalling and cell death. Apart from the mitochondrion the peroxisome is the most important cellular site for the generation and scavenging of hydrogen peroxide. Peroxisomes contain various oxidases, e.g. for the metabolism of long-chain fatty acids, polyamines, and for the oxidation of urate, which form hydrogen peroxide. Widely-used chemical probes for the detection of hydrogen peroxide like dichlorofluorescein diacetate (DCFDA) often lack in specificity and the possibility of compartment-specific measurement. To overcome these disadvantages, Belousov et al. developed the novel hydrogen peroxide sensitive fluorescent protein HyPer. In the present study the HyPer protein was fused with the PTS1 tag for a specific hydrogen peroxide detection in peroxisomes. The localization of the HyPer protein in the peroxisomes was confirmed by immunofluorescence and the functionality by fluorescence microscopy and flow cytometry analyses. The presented HyPer-Peroxi fluorescent protein is a valuable tool for studying hydrogen peroxide generation within the peroxisomes.


Subject(s)
Hydrogen Peroxide/analysis , Molecular Probes/analysis , Peroxisomes/chemistry , Recombinant Proteins/metabolism , Animals , Catalase/metabolism , Cell Line, Tumor , Fluorescence , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Molecular Probes/metabolism , Oxidation-Reduction , Oxidoreductases/metabolism , Peroxisomes/metabolism , Rats , Substrate Specificity , Uric Acid/metabolism
4.
Diabetes ; 60(1): 200-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20971967

ABSTRACT

OBJECTIVE: Type 2 diabetes is a complex disease that is accompanied by elevated levels of nonesterified fatty acids (NEFAs), which contribute to ß-cell dysfunction and ß-cell loss, referred to as lipotoxicity. Experimental evidence suggests that oxidative stress is involved in lipotoxicity. In this study, we analyzed the molecular mechanisms of reactive oxygen species-mediated lipotoxicity in insulin-producing RINm5F cells and INS-1E cells as well as in primary rat islet cells. RESEARCH DESIGN AND METHODS: The toxicity of saturated NEFAs with different chain lengths upon insulin-producing cells was determined by MTT and propidium iodide (PI) viability assays. Catalase or superoxide dismutase overexpressing cells were used to analyze the nature and the cellular compartment of reactive oxygen species formation. With the new H2O2-sensitive fluorescent protein HyPer H2O2 formation induced by exposure to palmitic acid was determined. RESULTS: Only long-chain (>C14) saturated NEFAs were toxic to insulin-producing cells. Overexpression of catalase in the peroxisomes and in the cytosol, but not in the mitochondria, significantly reduced H2O2 formation and protected the cells against palmitic acid-induced toxicity. With the HyPer protein, H2O2 generation was directly detectable in the peroxisomes of RINm5F and INS-1E insulin-producing cells as well as in primary rat islet cells. CONCLUSIONS: The results demonstrate that H2O2 formation in the peroxisomes rather than in the mitochondria are responsible for NEFA-induced toxicity. Therefore, we propose a new concept of fatty acid-induced ß-cell lipotoxicity mediated via reactive oxygen species formation through peroxisomal ß- oxidation.


Subject(s)
Hydrogen Peroxide/pharmacology , Insulin-Secreting Cells/metabolism , Animals , Cell Culture Techniques , Cell Survival/drug effects , DNA Primers , Extracellular Matrix/physiology , Fluoresceins/pharmacology , Hep G2 Cells/pathology , Humans , Hydrogen Peroxide/metabolism , Immunohistochemistry , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Lentivirus/physiology , Male , Oxidative Stress , Peroxisome-Targeting Signal 1 Receptor , Peroxisomes/physiology , Polymerase Chain Reaction , Rats , Rats, Inbred Lew , Receptors, Cytoplasmic and Nuclear/genetics
SELECTION OF CITATIONS
SEARCH DETAIL