Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dalton Trans ; 44(23): 10834-46, 2015 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-25806710

RESUMO

The crystal structure of the proton conducting perovskite BaCe(0.8)Y(0.2)O(3-δ) (BCY20) has been studied via high-resolution in situ neutron diffraction performed in controlled dry and humid (heavy water) oxygen flow. Two phase transitions, cubic Pm3[combining macron]m→R3[combining macron]c (775 °C)→Imma (250 °C) were observed on cooling from 1000 °C in dry O(2). A significant shift of the phase stability fields was observed on cooling in wet oxygen (pD(2)O ≈ 0.2 atm) with the R3[combining macron]c structure stabilised at 900 °C, and the R3[combining macron]c→Imma transition occurring at 675 °C. On cooling below 400 °C a monoclinic, I2/m, phase started to appear. The structural dependence on hydration level is primarily due to the de-stabilisation of the correlated, octahedra tilts as a consequence of structural relaxation around the oxygen vacancies present in the non-hydrated phase. The tendency of hydrated BaCe(0.8)Y(0.2)O(3-δ) to show octahedral tilting is also found to be enhanced, indicating that the deuteronic (protonic) defects influence the crystal structure, possibly via hydrogen bonding. Stabilisation of the monoclinic I2/m phase is attributed to the structural effect of deuterons that is inferred to increase on cooling as deuterons localise to a greater extent. Changing from wet oxidising (O(2) + D2O(g)) to wet reducing (5% H2 in Ar + D2O(g)) atmosphere did not influence the structure or the phase stability, indicating that Ce(4+) was not reduced under the present conditions. Based on the observed cell volume expansion protonic defects are present in the material at 900 °C at a D(2)O partial pressure of ∼0.2 atm. The origin of the chemical expansion is explained by the effective size of the oxygen vacancy being significantly smaller than the [OD] defect. Rietveld analysis has been used to locate possible sites for the deuterons in the high temperature, R3[combining macron]c and Imma, phases that are most relevant for proton transport.

2.
Biochem Pharmacol ; 76(12): 1748-56, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18831966

RESUMO

Potassium channel openers (KCOs) decrease insulin secretion from beta-cells. Some KCOs also protect against damage to beta-cell function and type 1 diabetes in animal models. Previously we have found that the KCO NNC 55-0118 counteracted islet cell dysfunction, and this was associated with a lowering of the mitochondrial membrane potential (Deltapsi). Presently we aimed to explore whether inhibition of insulin secretion per se or rather inhibition of mitochondrial function correlates to counteraction of beta-cell suppression. For this we used two novel KCOs (NNC 55-0321 and NNC 55-0462), which at certain concentrations have different actions regarding insulin secretion and the Deltapsi, with NNC 55-0321 being a potent inhibitor of Deltapsi and NNC 55-0462 being a potent inhibitor of insulin secretion. At 10 microM NNC 55-0321, but not with NNC 55-0462, the islet ATP content and ATP/ADP ratio was acutely decreased. This was accompanied by a complete protection against streptozotocin-induced suppression of islet insulin secretion using the former KCO. In cardiac research KCOs have been used to induce an ischemic preconditioning (IPC) response. In line with an IPC-like mechanism we found that NNC 55-0321 induced an initial free oxygen radical formation, PKC-epsilon isoform activation and a subsequent phosphorylation of the survival promoting factor Akt. Thus, KCOs may elicit mitochondrial events that resemble classical IPC seen in cardiomyocytes, and this could explain the enhanced islet cell function observed. KCOs with this property may be particularly interesting compounds to study as a rescue therapy during acute episodes of beta-cell suppression/destruction.


Assuntos
Guanidinas/farmacologia , Antagonistas da Insulina/farmacologia , Precondicionamento Isquêmico , Ilhotas Pancreáticas/fisiologia , Canais KATP/agonistas , Nitrilas/farmacologia , Substâncias Protetoras/farmacologia , Animais , Diazóxido/análogos & derivados , Diazóxido/farmacologia , Insulina/metabolismo , Secreção de Insulina , Masculino , Potenciais da Membrana , Membranas Mitocondriais/fisiologia , Proteína Oncogênica v-akt/metabolismo , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Estreptozocina
3.
Cell Transplant ; 15(8-9): 769-75, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17269447

RESUMO

Proinflammatory cytokines play a major role in rejection of pancreatic islet allografts and in type 1 diabetes (T1D). In rodent islets, exposure to IL-1beta alone or combined with IFN-gamma induces expression of inducible nitric oxide synthase (iNOS). Inhibition of iNOS or a deletion of the iNOS gene has been shown to be protective in animal models of T1D. In the present study we tested the hypothesis that transplantation of pancreatic islets deficient in iNOS (iNOS-/-) would permit increased graft survival. Pancreatic islets isolated from wild-type (wt) mice and iNOS-/- mice were allogeneically transplanted beneath the kidney capsule of spontaneously diabetic NOD mice. When blood glucose increased above 12.0 mM after preceding normalization of hyperglycemia, animals were sacrificed. Histological examinations of grafts were performed and graft gene expression was analyzed by real-time PCR. Transplantations of the two types of islets could reverse hyperglycemia and the grafts functioned for on average 1 week posttransplantation. Morphological examination of both types of islet grafts showed immune cell infiltration around and within the grafts. Remaining endocrine cells could be observed in wt and iNOS-/- islet grafts. In the removed grafts iNOS-/- islet tissue contained higher mRNA levels of insulin, proinsulin convertases (PC-1 and PC-2), and IL-1beta compared to transplanted wt islets. The assessments of insulin, PC-1 and PC-2 mRNAs of the grafts suggest that the iNOS-/- islets may be more resistant to destruction in the transplantation model used; however, this was not sufficient to prolong the period of normoglycemia posttransplantation.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Sobrevivência de Enxerto/genética , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico Sintase Tipo II/deficiência , Animais , Glicemia/análise , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Sobrevivência de Enxerto/fisiologia , Insulina/genética , Insulina/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ilhotas Pancreáticas/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 2/genética , Pró-Proteína Convertase 2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Mol Cell Endocrinol ; 240(1-2): 50-7, 2005 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-16023781

RESUMO

In rodent islets, exposure to interleukin-1beta (IL-1beta) and interferon-gamma (IFN-gamma) induces expression of inducible nitric oxide synthase (iNOS) and subsequent nitric oxide (NO) formation, which may inhibit islet function. However, cytokines may also induce NO-independent islet suppression. The present aim was to investigate the effect of cytokine exposure to iNOS deficient (iNOS-/-) mouse islets on various islet functions. Islets from iNOS-/- and wt mice exposed to IL-1beta or (IL-1beta + IFN-gamma) for 2-20 h showed different kinetics of glucose-stimulated insulin secretion. In iNOS-/- islets, IL-1beta at high glucose induced a delayed and prolonged stimulation of insulin secretion, and this was followed by an increase in phospholipase D mRNA expression. After 6 and 24 h, proinsulin convertase 1 and 2 (PC1 and PC2) mRNA expression was suppressed and proinsulin secretion increased from wt islets. In iNOS-/- islets, PC1 expression was recovered after 24 h, and there was no difference in proinsulin secretion. PDX-1 mRNA expression was suppressed independent of NO-formation. We conclude that cytokines induce both NO-dependent and NO-independent functional inhibition of murine beta-cells.


Assuntos
Citocinas/farmacologia , Proteínas de Homeodomínio/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/deficiência , Proinsulina/metabolismo , Transativadores/metabolismo , Animais , Glucose/farmacologia , Secreção de Insulina , Interferon gama/farmacologia , Interleucina-1/farmacologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Mutantes , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Fosfolipase D/genética , Fosfolipase D/metabolismo , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 2/genética , Pró-Proteína Convertase 2/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/metabolismo
5.
Mol Cell Endocrinol ; 220(1-2): 21-9, 2004 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-15196696

RESUMO

Cytokines may be involved in islet destruction during Type 1 diabetes. Exposure to interleukin-1beta (IL-1beta) or IL-1beta plus interferon-gamma (IFN-gamma) of rodent islets induces expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Subsequent formation of nitric oxide (NO) and prostaglandin E(2) (PGE(2)) may impair beta-cell function. Using iNOS deficient (iNOS -/-) islets, we have further investigated the relation between NO formation and PGE(2) induction. We found that iNOS -/- islets responded with a reduced PGE(2) formation following IL-1beta or (IL-1beta + IFN-gamma) treatment compared to wild-type (wt) islets, while COX-2 mRNA or protein content were unchanged. By the addition of an NO donor together with IL-1beta, PGE(2) formation could be stimulated from iNOS -/- islets. We conclude that the lowered capacity of PGE(2) formation observed from cytokine exposed iNOS -/- islets is due to a decreased stimulation of PGE(2) formation by the COX-2 enzyme in the absence of NO, rather then differences in expressed COX-2 protein.


Assuntos
Dinoprostona/biossíntese , Interleucina-1/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico Sintase/deficiência , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ciclo-Oxigenase 2 , Dinoprostona/farmacologia , Deleção de Genes , Insulina/metabolismo , Secreção de Insulina , Interferon gama/farmacologia , Ilhotas Pancreáticas/citologia , Isoenzimas/biossíntese , Isoenzimas/genética , Camundongos , Camundongos Knockout , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Prostaglandina-Endoperóxido Sintases/biossíntese , Prostaglandina-Endoperóxido Sintases/genética , RNA Mensageiro/metabolismo
6.
Immunol Lett ; 88(2): 141-5, 2003 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12880684

RESUMO

The cytokine IL-15 might contribute to inflammatory processes, but also act as an inhibitor of apoptosis in different cell lines. Furthermore, it has been reported that islet cells express IL-15 after exposure to proinflammatory cytokines, which could indicate a defence reaction. We aimed in this study to investigate if IL-15 could influence cell death and/or functional impairment of rat pancreatic islets induced by in vitro exposure to a combination of cytokines (25 U/ml IL-1beta+1000 U/ml IFN-gamma+1000 U/ml TNF-alpha). The effect of IL-15 itself on the function of rat pancreatic islets was also studied. Isolated rat islets were exposed for 24 h to IL-15 at different concentrations in the presence or absence of the cytokine mixture. The cytokines caused a strong inhibition of glucose-stimulated insulin release and the glucose oxidation rates. IL-15 (0.1-10 ng/ml) could not prevent the functional suppression caused by these effects. The cytokine combination caused a decline in whole islet DNA content and a marked increase in non-viable cells analysed by propidium iodide (PI) and annexin V staining. However, there was no significant decrease in whole islet DNA content when IL-15 (0.1 or 1.0 ng/ml) was present together with the cytokine mixture. On the other hand, IL-15 failed to influence the increase in cell death after PI and annexin V staining. If anything, IL-15 alone had a slight stimulatory effect (glucose oxidation rate) on islet cells. In conclusion, we can not exclude that IL-15 might antagonize some cytokine mediated cell death in islet cells, however, IL-15 fails to counteract functional suppression induced by cytokines.


Assuntos
Citocinas/farmacologia , Mediadores da Inflamação/farmacologia , Interleucina-15/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Animais , Morte Celular/efeitos dos fármacos , DNA/análise , DNA/genética , Citometria de Fluxo , Glucose/metabolismo , Insulina/metabolismo , Interferon gama/farmacologia , Interleucina-1/farmacologia , Interleucina-15/genética , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Oxirredução/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...