Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ewa Gurgul-Convey is active.

Publication


Featured researches published by Ewa Gurgul-Convey.


Journal of Endocrinology | 2008

Interaction between pro-inflammatory and anti-inflammatory cytokines in insulin-producing cells

Kleber L.A. Souza; Ewa Gurgul-Convey; Matthias Elsner; Sigurd Lenzen

Pro-inflammatory cytokines cause beta-cell dysfunction and death. The aim of this study was to investigate the interactions between different pro- and anti-inflammatory cytokines and their effects on apoptotic beta-cell death pathways. Insulin-producing RINm5F cells were exposed to different combinations of cytokines. Gene expression analyses of manganese superoxide dismutase (MnSOD) and inducible nitric oxide synthase (iNOS) were performed by real-time RT-PCR. Cell viability was measured by the MTT assay, NFkappaB activation using a SEAP reporter gene assay, protein expression by western blotting and caspase-3 activity using the DEVD cleavage method. IL-1beta, tumour necrosis factor alpha (TNFalpha) and a combination of all three pro-inflammatory cytokines increased while IFNgamma alone did not affect NFkappaB activity and iNOS gene and protein expression. Interestingly, the anti-inflammatory cytokines IL-4, IL-13 and IL-10 decreased IL-1beta-stimulated NFkappaB activation and iNOS expression. IL-1beta, TNFalpha and the pro-inflammatory cytokine combination also increased MnSOD gene and protein expression. But IL-4, IL-13 and IL-10 did not affect MnSOD expression and did not modulate IL-1beta-stimulated MnSOD expression. Caspase-3 activity was increased by IL-1beta and the pro-inflammatory cytokine combination, and to a lesser extent by TNFalpha. In contrast, IFNgamma had no effect on caspase-3 activity. IL-4, IL-13 and IL-10 decreased caspase-3 activity and increased viability of insulin-producing cells treated with pro-inflammatory cytokines. The anti-inflammatory cytokines counteracted the cytotoxic effects of pro-inflammatory cytokines in insulin-producing cells. This was achieved through the reduction of nitrosative stress. Thus, a balance between the anti-inflammatory and the pro-inflammatory cytokines is of crucial importance for the prevention of pancreatic beta-cell destruction.


Journal of Molecular Medicine | 2011

Cytokine toxicity in insulin-producing cells is mediated by nitro-oxidative stress-induced hydroxyl radical formation in mitochondria

Ewa Gurgul-Convey; Ilir Mehmeti; Stephan Lortz; Sigurd Lenzen

Although nitric oxide (NO) and oxidative stress both contribute to proinflammatory cytokine toxicity in pancreatic β-cells during type 1 diabetes mellitus (T1DM) development, the interactions between NO and reactive oxygen species (ROS) in cytokine-mediated β-cell death have not been clarified. Exposure of insulin-producing RINm5F cells to IL-1β generated NO, while exposure to a combination of IL-1β, TNF-α, and IFN-γ, which simulates T1DM conditions, generated both NO and ROS. In theory, two reactions between NO and ROS are possible, one with the superoxide radical yielding peroxynitrite, and the other with hydrogen peroxide (H2O2) yielding hydroxyl radicals. Results of the present work exclude peroxynitrite involvement in cytokine toxicity to β-cells because its generation did not correlate with the toxic action of cytokines. On the other hand, we show that H2O2, produced upon exposure of insulin-producing cell clones and primary rat islet cells to cytokines almost exclusively in the mitochondria, reacted in the presence of trace metal (Fe++) with NO forming highly toxic hydroxyl radicals, thus explaining the severe toxicity that causes apoptotic β-cell death. Expression of the H2O2-inactivating enzyme catalase in mitochondria protected against cytokine toxicity by preventing hydroxyl radical formation. We therefore conclude that proinflammatory cytokine-mediated β-cell death is due to nitro-oxidative stress-mediated hydroxyl radical formation in the mitochondria.


Cytokine | 2011

Differential effects of proinflammatory cytokines on cell death and ER stress in insulin-secreting INS1E cells and the involvement of nitric oxide.

Stella Kacheva; Sigurd Lenzen; Ewa Gurgul-Convey

Proinflammatory cytokines produced by immune cells destroy pancreatic beta cells in type 1 diabetes. The aim of this study was to investigate the cytokine network and its effects in insulin-secreting cells. INS1E cells were exposed to different combinations of proinflammatory cytokines. Cytokine toxicity was estimated by MTT assay and caspase activation measurements. The NFκB-iNOS pathway was analyzed by a SEAP reporter gene assay, Western-blotting and nitrite measurements. Gene expression analyses of ER stress markers, Chop and Bip, were performed by real-time RT-PCR. Cytokines tested in this study, namely IL-1β, TNFα and IFNγ, had deleterious effects on beta cell viability. The most potent toxicity exhibited IL-1β and its combinations with other cytokines. The toxic effects of IL-1β towards cell viability, caspase activation and iNOS activity were dependent on nitric oxide and abolished by an iNOS blocker. IL-1β was the strongest inducer of the NFκB activation. An iNOS blocker inhibited IL-1β-mediated NFκB activation in the first, initial phase of cytokine action, but did not affect significantly NFκB activation after prolonged incubation. Interestingly iNOS protein expression was induced predominantly by IL-1β and decreased in the presence of an iNOS blocker in the case of a short time exposure. The changes in the expression of ER stress markers were also almost exclusively dependent on the IL-1β presence and counteracted by iNOS blockade. Thus cytokine-induced beta cell death is primarily IL-1β mediated with a NO-independent enhancement by TNFα and IFNγ. The deleterious effects on cell viability and function are crucially dependent on IL-1β-induced nitric oxide formation.


Diabetologia | 2013

Overexpression of the antioxidant enzyme catalase does not interfere with the glucose responsiveness of insulin-secreting INS-1E cells and rat islets

Stephan Lortz; Ewa Gurgul-Convey; O. Naujok; Sigurd Lenzen

Aims/hypothesisHydrogen peroxide (H2O2)-inactivating enzymes such as catalase are produced in extraordinarily low levels in beta cells. Whether this low expression might be related to a signalling function of H2O2 within the beta cell is unknown. A high level of H2O2-inactivating enzymes could potentially be incompatible with glucose-induced insulin secretion. Therefore the effect of catalase overexpression on mitochondrial function and physiological insulin secretion was studied in insulin-secreting INS-1E and primary islet cells.MethodsINS-1E and rat islet cells were lentivirally transduced to overexpress catalase in the cytosol (CytoCat) or in mitochondria (MitoCat). Cell viability and caspase-3 activation were assessed after cytokine incubation and hypoxia. Insulin secretion was quantified and expression of the gene encoding the mitochondrial uncoupling protein 2 (Ucp2) was measured in parallel to mitochondrial membrane potential and reactive oxygen species (ROS) formation.ResultsThe ability to secret insulin in a glucose-dependent manner was not suppressed by catalase overexpression, although the glucose-dependent increase in the mitochondrial membrane potential was attenuated in MitoCat cells along with an increased Ucp2 expression and reduced mitochondrial ROS formation. In addition, MitoCat overexpressing cells were significantly more resistant against pro-inflammatory cytokines and hypoxia than CytoCat and control cells.Conclusions/interpretationThe results demonstrate that an improved antioxidative defence status of insulin-secreting cells allowing efficient H2O2 inactivation is not incompatible with proper insulin secretory responsiveness to glucose stimulation and provide no support for a signalling role of H2O2 in insulin-secreting cells. Interestingly, the results also document for the first time that the decreased ROS formation with increasing glucose concentrations is of mitochondrial origin.


Biochimica et Biophysica Acta | 2011

Induction of the intrinsic apoptosis pathway in insulin-secreting cells is dependent on oxidative damage of mitochondria but independent of caspase-12 activation

Ilir Mehmeti; Ewa Gurgul-Convey; Sigurd Lenzen; Stephan Lortz

Pro-inflammatory cytokine-mediated beta cell apoptosis is activated through multiple signaling pathways involving mitochondria and endoplasmic reticulum. Activation of organelle-specific caspases has been implicated in the progression and execution of cell death. This study was therefore performed to elucidate the effects of pro-inflammatory cytokines on a possible cross-talk between the compartment-specific caspases 9 and 12 and their differential contribution to beta cell apoptosis. Moreover, the occurrence of ROS-mediated mitochondrial damage in response to beta cell toxic cytokines has been quantified. ER-specific caspase-12 was strongly activated in response to pro-inflammatory cytokines; however, its inhibition did not abolish cytokine-induced mitochondrial caspase-9 activation and loss of cell viability. In addition, there was a significant induction of oxidative mitochondrial DNA damage and elevated cardiolipin peroxidation in insulin-producing RINm5F cells and rat islet cells. Overexpression of the H(2)O(2) detoxifying enzyme catalase effectively reduced the observed cytokine-induced oxidative damage of mitochondrial structures. Taken together, the results strongly indicate that mitochondrial caspase-9 is not a downstream substrate of ER-specific caspase-12 and that pro-inflammatory cytokines cause apoptotic beta cell death through activation of caspase-9 primarily by hydroxyl radical-mediated mitochondrial damage.


Free Radical Biology and Medicine | 2009

Protection of insulin-producing cells against toxicity of dexamethasone by catalase overexpression

Leticia P Roma; José Roberto Bosqueiro; Daniel Andrade Da Cunha; Everardo M. Carneiro; Ewa Gurgul-Convey; Sigurd Lenzen; Antonio Carlos Boschero; Kleber L.A. Souza

Pancreatic beta cells are very sensitive to reactive oxygen species (ROS) and this might play an important role in beta cell death in diabetes. Dexamethasone is a synthetic diabetogenic glucocorticoid, which impairs pancreatic beta cell function. Therefore we investigated the toxicity of dexamethasone in RINm5F insulin-producing cells and its dependence on the expression level of the antioxidant enzyme catalase, which inactivates hydrogen peroxide. This was correlated with oxidative stress and cell death. An increased generation of ROS was observed in dexamethasone-treated cells together with an increase in caspase-3 activity and apoptosis rate. Interestingly, exposure to dexamethasone increased the cytosolic superoxide dismutase Cu/ZnSOD protein expression and activity, whereas the mitochondrial MnSOD isoform was not affected by the glucocorticoid. Catalase overexpression in insulin-producing cells prevented all the cytotoxic effects of dexamethasone. In conclusion, dexamethasone-induced cell death in insulin-producing cells is ROS mediated. Increased levels of expression and activity of the Cu/ZnSOD might favor the generation of hydrogen peroxide in dexamethasone-treated cells. Increased ROS scavenging capacity in insulin-producing cells, through overexpression of catalase, prevents a deleterious increase in hydrogen peroxide generation and thus prevents dexamethasone-induced apoptosis.


Journal of Biological Chemistry | 2010

Protection against Cytokine Toxicity through Endoplasmic Reticulum and Mitochondrial Stress Prevention by Prostacyclin Synthase Overexpression in Insulin-producing Cells

Ewa Gurgul-Convey; Sigurd Lenzen

Proinflammatory cytokines play a crucial role in the pathogenesis of type 1 diabetes mellitus. One of the cytokine-regulated pathways mediating inflammation in this autoimmune disease is the arachidonic acid metabolism pathway, comprising both the induction of cyclooxygenases and the production of different prostaglandins. Cytokine toxicity is mediated in many cell types, including pancreatic β cells through this pathway. Interestingly, some cell types have been shown to be insensitive to such toxicity, and this correlated with a high expression of prostacyclin synthase (PGIS). Using insulin-producing RINm5F cells as a model for pancreatic β cells, PGIS was overexpressed and exhibited a large protective effect against cytokine toxicity. This protective effect of PGIS against cytokine toxicity correlated with a decreased activation of the transcription factor NFκB and the inducible NO synthase promoter as well as a reduced inducible NO synthase protein expression and nitrite production. A reduction in the cytokine-stimulated endoplasmic reticulum and mitochondrial stress was also found in the PGIS-overexpressing cells. Moreover, cytokine-induced caspase-3 activation and reduction of glucose oxidation and cell proliferation were suppressed. Thus, PGIS overexpression apparently protects insulin-producing cells against cytokine toxicity via suppression of endoplasmic reticulum and mitochondrial stress-mediated cell death pathways.


Antioxidants & Redox Signaling | 2008

Relation between triketone structure, generation of reactive oxygen species, and selective toxicity of the diabetogenic agent alloxan.

Matthias Elsner; Ewa Gurgul-Convey; Sigurd Lenzen

The diabetogenic agent alloxan is a triketone that selectively destroys pancreatic beta cells. To investigate the importance of the triketone structure of alloxan for its cytotoxic potency, alloxan was compared with ninhydrin, also a triketone, and the amino derivative of alloxan uramil, which is not a triketone, because the 5-keto group of the alloxan is replaced by an amino group. Both compounds are cytotoxic but not diabetogenic. Ninhydrin was capable of generating cytotoxic reactive oxygen species (ROS) through redox cycling with dithiols, and uramil could also generate cytotoxic ROS. Both ninhydrin and uramil could not redox cycle with glutathione (GSH) and were not selectively toxic to beta cells; their structure does not allow selective cellular uptake via the GLUT2 glucose transporter. Thus, the results show that the 5-keto group in the pyrimidine ring structure of the triketone alloxan is crucially important for its ability to be selectively taken up into the beta cells via the specific glucose transporter GLUT2. The 5-keto group of the molecule enables redox cycling of alloxan through reaction with glutathione (GSH), thereby generating the cytotoxic ROS. Thus, the unique combination of these two properties confers on alloxan the beta cell-selective toxicity and diabetogenicity. Replacement of the 5-keto group by an amino group, as in uramil, abolishes selective beta cell toxicity because of the loss of the glucose analogue structure and the capability to generate ROS via redox cycling with GSH and cysteine.


Endocrinology | 2012

Mechanism of Prostacyclin-Induced Potentiation of Glucose-Induced Insulin Secretion

Ewa Gurgul-Convey; Katarzyna Hanzelka; Sigurd Lenzen

Arachidonic acid metabolites are crucial mediators of inflammation in diabetes. Although eicosanoids are established modulators of pancreatic β-cell function, the role of prostacyclin (prostaglandin I2) is unknown. Therefore, this study aimed to analyze the role of prostacyclin in β-cell function. Prostacyclin synthase (PGIS) was weakly expressed in rat islet cells but nevertheless significantly increased by incubation with 30 mM glucose, especially in non-β-cells. PGIS was overexpressed in INS1E cells, and the regulation of insulin secretion was analyzed. PGIS overexpression strongly potentiated glucose-induced insulin secretion along with increased insulin content and ATP production. Importantly, overexpression of PGIS potentiated only nutrient-induced insulin secretion. The effect of PGIS overexpression was mediated by prostacyclin released from insulin-secreting cells and dependent on prostacyclin receptor (IP receptor) activation, with concomitant cAMP production. The cAMP-mediated potentiation of glucose-induced insulin secretion by prostacyclin was independent of the protein kinase A pathway but strongly attenuated by the knockdown of the exchange protein directly activated by cAMP 2 (Epac2), pointing to a crucial role for Epac2 in this process. Thus, prostacyclin is a powerful potentiator of glucose-induced insulin secretion. It improves the secretory capacity by inducing insulin biosynthesis and probably by stimulating exocytosis. Our findings open a new therapeutical perspective for an improved treatment of type 2 diabetes.


Nitric Oxide | 2012

Is there a role for neuronal nitric oxide synthase (nNOS) in cytokine toxicity to pancreatic beta cells

Ewa Gurgul-Convey; Katarzyna Hanzelka; Sigurd Lenzen

Nitric oxide (NO), produced by the action of the inducible NO synthase, plays a crucial role in cytokine toxicity to pancreatic beta cells during type 1 diabetes development. It was the aim of this study to analyze the role of the neuronal NOS (nNOS) in proinflammatory cytokine-mediated beta cell toxicity. Expression of different isoforms of nitric oxide synthase in insulin-secreting INS1E cells and rat islets was analyzed by quantitative real-time PCR and Western blotting. The expression of nNOS in insulin-secreting INS1E cells was similar to that found in rat brain, while two other isoforms, namely the endothelial eNOS and inducible iNOS were not expressed in untreated cells. IL-1β alone or in combination with TNF-α and/or IFNγ induced iNOS but not eNOS expression. In contrast, nNOS expression was strongly decreased by the mixture of the three proinflammatory cytokines (IL-1β, TNF-α and IFNγ) both on the gene and protein level in INS1E cells and rat islet cells. The effects of cytokines on glucose-induced insulin-secretion followed the pattern of nNOS expression reduction and, on the other hand, of the iNOS induction. The data indicate that a low level of nitric oxide originating from the constitutive expression of nNOS in pancreatic beta cells is not deleterious. In particular since proinflammatory cytokines reduce this expression. This nNOS suppression can compensate for NO generation by low concentrations of IL-1β through iNOS induction. Thus, this basal nNOS expression level in pancreatic beta cells represents a protective element against cytokine toxicity.

Collaboration


Dive into the Ewa Gurgul-Convey's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ilir Mehmeti

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jolanta Jura

Jagiellonian University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amélie Hu

Université libre de Bruxelles

View shared research outputs
Researchain Logo
Decentralizing Knowledge